ESMO Supporter 2017

Displaying One Session

Hall 8 Poster Display session
Date
11.09.2017
Time
13:15 - 14:15
Location
Hall 8
Poster display session Poster Display session

BASIC SCIENCE (ID 5671)

Lecture Time
13:15 - 13:15
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15
Poster display session Poster Display session

7P - GSKJ4, an H3K27me3 demethylase inhibitor, effectively suppresses the breast cancer stem cells (ID 2053)

Presentation Number
7P
Lecture Time
13:15 - 13:15
Speakers
  • N. Yan
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Breast cancer stem cells (BCSCs) are responsible for breast cancer metastasis and treatment failure. Hence, eliminating BCSCs poses possibility to eradicate breast cancer. Recently, studies have been suggested that H3K27me3 is implicated in maintenance of cancer stem cells (CSCs), however, the roles of H3K27me3 in BCSCs remain poorly investigated. Hence, we aimed to explore the functionality of H3K27me3 in BCSCs.

Methods

Here, we firstly determined the global level of H3K27me3 in mammosphere-derived cells. Then, we detected the effect of GSKJ4 in cell viability through CCK8 assay. Next, we tested the impact of GSKJ4 on BCSCs expansions with CD44+CD24- phenotype and ALDH1-positive via flowcytometry. In addition, the impact on self-renewal capacity of BCSCs were also asked using mammosphere formation assay and colony formation assay. Further, western blot and Q-PCR were conducted to explore the effect of GSKJ4 in the expression level of stemness-related markers. Finally, we determined the influence of GSKJ4 on tumorigenicity using a xenograft model and investigated the underlying mechanisms.

Results

We identify H3K27me3 as a negative modulator of stemness of BCSCs and suggest GSKJ4 is a promising drug targeting BCSCs. we show that the H3K27me3 level is decreased in mammosphere-derived BCSCs. In breast cancer cells, we demonstrate that GSKJ4 could markedly inhibit the proliferation. Strikingly, we show that GSKJ4 could effectively suppress BCSCs including its expansion, self-renewal capacity, and the expression of stemness-related markers. Additionally, our xenograft model confirms that GSKJ4 is able to effectively inhibit the tumorigenicity of MDA-MB-231. Mechanistically, the inhibition effect of GSKJ4 in BCSCs is via inhibiting JMJD3 and UTX thus causing increment of H3K27me3 level, which results in suppressing stemness factors including NANOG, SOX2 and OCT4.

Conclusions

Our results provide strong supports that epigenetic modification is associated with maintenance of properties of BCSCs and reveal that GSKJ4 is capable to be a prospective agent targeting BCSCs.

Legal entity responsible for the study

N/A

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

8P - The impact of rotenone-modulated oxidative stress on the survival of human breast cancer stem cells (CD24-/CD44+) (ID 5102)

Presentation Number
8P
Lecture Time
13:15 - 13:15
Speakers
  • S. Wanandi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Cancer stem cells (CSCs) have been proven to be tumorigenic and may be responsible for the resistance to chemo-radiation therapy, disease recurrence and metastasis. Chemo-radiation therapy modulates oxidative stress in cancer cells, leading to cellular adaption response including modulation of cell survival and antioxidant defense mechanisms. However, the redox status alteration of breast CSCs is not yet clearly understood. The aim of this study was to elaborate the impact of rotenone-modulated oxidative stress on the survival of human breast CSCs (CD24-/CD44+) which might be beneficial to understand the underlying mechanism of chemo-radiation therapy resistance.

Methods

Human breast CSCs (CD24-/CD44+) and non-CSCs (CD24-/CD44-) were treated with rotenone and DMSO (vehicle) for 6 hours, respectively. The effects of rotenone on oxidative stress were assessed by analysing intracellular reactive oxygen species (ROS) level using dihydroethidium assay, as well as mRNA expression and specific activity of MnSOD antioxidant. Finally, cell survival was determined using MTS assay, as well as through analysis of survivin mRNA expression.

Results

Our results showed that rotenone could not modulate the superoxide level of human breast CSCs (CD24-/CD44+), in contrast to that of non-CSCs (CD24-/CD44-). Albeit MnSOD synthesis in human breast CSCs has been excessively enhanced following rotenone treatment, the enzyme activity was still lower than in non-CSCs. Importantly, the cell viability of CSCs was higher than that of non-CSCs, which related to the increase of survivin.

Conclusions

We conclude that human breast CSCs (CD24-/CD44+) could survive better than their counterpart non-CSCs (CD24-/CD44-) when treated with rotenone. This impact might be associated with the increase of antioxidant MnSOD expression and survivin mRNA expression.

Legal entity responsible for the study

Faculty of Medicine, Universitas Indonesia

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

9P - Kynurenine-3-monooxygenase (KMO) protein promotes triple negative breast cancer progression (ID 3601)

Presentation Number
9P
Lecture Time
13:15 - 13:15
Speakers
  • C. Liu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Triple-negative breast cancer (TNBC) remains a difficult-to-treat cancer and the biology beneath TNBC is a research interest. Tryptophan-kynurenine metabolism plays an important role in epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs) and immune escape. Previous studies have focused on the expression and function of the first step and the rate-limiting enzyme in tumor cells, whereas the second step catabolic enzyme kynurenine 3-monooxygenase (KMO) was rarely addressed in tumorigenesis. Hence, we sought to investigate of the mechanism and functions of KMO in TNBC carcinogenesis.

Methods

KMO gene alteration and mRNA transcripts were analyzed from the The Cancer Genome Atlas (TCGA) database. MDA-MB-231 and MDA-MB-468 TNBC cell lines were used for in vitro studies. Cell proliferation, colony formation, transwell migration/invasion assays and tumorsphere forming ability were used for functional study. Signal transduction pathways were assessed by Western blot, quantitative real-time PCR and reporter assays. The effect of KMO on tumor growth was tested in nude mice with breast cancer xenografts.

Results

TCGA analysis showed high-frequency of KMO amplification alterations, which was related to poor overall survival in breast cancers. KMO transcripts were up-regulated in the tumor tissues of breast cancers, especially in TNBC. The functional assays showed that ectopic KMO expression promoted tumorigenesis, including cell growth and abilities of colony formation, migration, invasion, and tumorsphere formation. Moreover, western blot analysis revealed expressions of epithelial marker E-cadherin were decreased and mesenchymal markers N-cadherin, and Twist were increased by KMO overexpression in MDA-MB-468 cells. Interestingly, the mRNA and protein levels of pluripotency genes including CD44, Nanog, Oct4, and SOX-2 were also suppressed by KMO knockdown. Data of reporter gene assay showed that the activities of Nanog, Oct4, and SOX-2 promoters were enhanced by KMO overexpression. Furthermore, knockdown KMO decreased the xenografted tumor growth of MDA-MB-468 cells, suggesting its oncogenic role in TNBC.

Conclusions

Our data highlight the novel and critical roles of KMO in TNBC progression and metastasis.

Clinical trial identification

Not applicable

Legal entity responsible for the study

Taipei Veterans General Hospital

Funding

Taipei Veterans General Hospital

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

10P - PIM1 kinase promotes cell migration via SHP2 in triple-negative breast cancer (ID 2297)

Presentation Number
10P
Lecture Time
13:15 - 13:15
Speakers
  • F. Braso Maristany
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Triple-negative breast cancers (TNBCs) are aggressive and associated with poor prognosis. We have recently demonstrated that PIM1 regulates cell death, tumour growth and chemotherapy response in TNBC. This study aims to further explore the molecular mechanisms by which PIM1 promotes malignant phenotypes in TNBC, in particular cell migration.

Methods

The HumanHT-12 v4 expression array was used to interrogate changes in gene expression upon PIM1 knockdown in TNBC cell lines. Transwell migration assays and time-lapse live-cell imaging were used to study the role of PIM1 in cell migration. To assess the morphology of TNBC cells we stained F-actin with 488-phalloidin. Phospho-kinase arrays were used to elucidate the pathway by which PIM1 may control cell migration.

Results

Gene expression analysis revealed PTPN11 as the most downregulated gene upon PIM1 knockdown in TNBC cell lines. These results were validated by qRTPCR in 3 TNBC cell lines. PTPN11 encodes for the phosphatase SHP2, known to be relevant for the migration of TNBC cells. We therefore studied whether PIM1 was also required for this phenotype in TNBC cell lines. PIM1 knockdown led to a defect on 2D-transwell migration in MDA-MB-231 and SUM159 cells, similar to that observed upon SHP2 knockdown. Interestingly, SHP2 knockdown did not affect short-term cell population growth of TNBC cells, suggesting that PIM1 exerts its role in cell population growth via different mechanisms, as demonstrated previously. Upon PIM1 knockdown, MDA-MB-231 showed lower motility persistence, increased circularity and a reduction of F-actin filaments. To understand the common downstream targets of PIM1 and SHP2 and elucidate the pathway by which PIM1 may control cell migration, we used phospho-kinase arrays. These revealed decreased phosphorylation of PLCg1, FAK and PYK2, proteins involved in cell migration, upon either PIM1 or SHP2 knockdown.

Conclusions

These data suggest that PIM1 regulates cell migration by controlling PTPN11/SHP2 expression and provide further evidence for PIM1 as a target for TNBC therapy, not only to induce apoptosis and prevent tumour growth, but also to prevent TNBC migration.

Clinical trial identification

N/A

Legal entity responsible for the study

King's College London/Breast Cancer Now

Funding

Breast Cancer Now

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

11P - SHP-1 agonist SC-43 enhanced the anti-tumor effect of docetaxel through suppressing p-STAT3 in triple negative breast cancer cells (ID 1581)

Presentation Number
11P
Lecture Time
13:15 - 13:15
Speakers
  • K. Chen
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Triple negative breast cancer (TNBC) is an aggressive cancer and its prognosis remains poor. Combinational therapies are a promising strategy for enhancing treatment efficacy. Blockade of STAT3 signaling have been shown to enhance the response of cancer cells to conventional chemotherapeutic agents. Here we used a SHP-1 agonist SC-43 to dephosphorylate STAT3, thereby suppressing oncogenic STAT3 signaling, and tested it in combination with docetaxel in TNBC cells.

Methods

TNBC cell lines (HCC-1937, MDA-MB-468, MDA-MB-231) were used for in vitro studies. Cell viability was examined by MTT assay. Combination index was determined using Calcusyn anaysis. Apoptosis was examined by flow cytometry and western blot. Signal transduction pathways in cells were assessed by western blot. In vivo efficacy of SC-43 in combination with docetaxel was tested in nude mice with breast cancer xenografts.

Results

To exam expression of SHP-1 in clinical samples, we analyzed mRNA expression of SHP-1 gene (ptpn6) in a public TNBC dataset (The Cancer Genome Atlas, TCGA). We found that higher SHP-1 mRNA expression is associated with better overall survival in TNBC patients. Sequential combination of docetaxel and SC-43 in vitroshowed enhanced anti-proliferation and apoptosis associated with decreased p-STAT3 and decreased STAT3-downstream effector cyclin D1 in the TNBC cell lines MDA-MB-231, MDA-MB-468 and HCC-1937. Ectopic expression of STAT3 reduced theincreased cytotoxicity induced by the combination therapy. In addition, this sequential combination showed enhanced SHP-1 activity compared to SC-43 alone. Furthermore, siRNA against SHP-1 reduced apoptosis induced by the combination treatment. Moreover, by ectopic expression of SHP-1 mutants that caused SC-43 to lose its SHP-1 agonist capability, the SC-43-induced p-STAT3 signaling inhibition was reduced in the cells subjected to the combination treatment, suggesting SHP-1 plays a crucial role in docetaxel-SC-43-mediated TNBC cell apoptosis, Importantly, combination of docetaxel and SC-43 showed enhanced anti-tumor growth compared to single-agent therapy in MDA-MB-231 xenografted tumor mice.

Conclusions

SHP-1 agonist SC-43 enhanced the anti-tumor effect of docetaxel by SHP-1 dependent STAT3 inhibition in human TNBC cells. We suggest a therapeutic potential of SHP-1 agonist in combination with docetaxel for TNBC.

Legal entity responsible for the study

National Taiwan University Hospital

Funding

NHRI-EX106-10608BI

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

12P - Delineating the mechanisms of resistance to panHER inhibitors in HER2+ breast cancer cells (ID 3981)

Presentation Number
12P
Lecture Time
13:15 - 13:15
Speakers
  • O. Kutuk
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Despite the increase in patient survival rates promoted by increased screening and prevention efforts, much faster tumor genome sequencing and developed smart targeted therapies, de novo or acquired chemoresistance remains to be a significant factor for treatment failure in breast cancer therapeutics. Conventional chemotherapy, radiotherapy as well as targeted therapies activate mitochondrial cell death machinery to eliminate cancer cells. BCL‐2 protein family members regulate mitochondrial cell death pathway by controlling mitochondrial outer membrane permeabilization. Neratinib and dacomitinib are potent and irreversible pan‐EGFR inhibitors, which block their autophosphorylation and downstream signaling. Moreover, neratinib and dacomitinib have been shown to activate cell death in HER2‐overexpressing cell lines. The aim of our study was to identify molecular pathways responsible for panHER2 inhibitor resistance in HER2+ breast cancer cells.

Methods

The expression of EGFR and BCL‐2 protein family members was determined by immunoblotting and qPCR. CellTiter‐Glo was used to measure cell viability and AnnexinV/PI staining and flow cytometer was used to evaluate apoptotic response. BH3 profiling was used to determine the apoptotic blocks and mitochondrial cell death priming in breast cancer cells.

Results

Here we showed that increased MCL‐1 and decreased BIM mediate resistance to neratinib in ZR‐75‐30 and SKBR3 cells while increased BCL‐XL and BCL‐2 and decreased BIM promoted neratinib resistance in BT474 cells. Cells were also cross-resistant to dacomitinib. BH3 profiles of HER2+ breast cancer cells efficiently predicted anti‐apoptotic protein dependence and development of resistance to panHER2 inhibitors. Adding specific ERK1/2 inhibitor SCH772984 to neratinib or dacomitinib led to increased apoptotic response in SKBR3 and ZR‐75‐30 cells, but we did not detect a similar response in BT‐474 cells. Reactivation of ERK1/2 was primarily responsible for acquired resistance in SKBR3 and ZR‐75‐30 cells. Intriguingly, both ERK1/2 and Akt/NFkappaB pathways were responsible for neratinib resistance in BT474 cells.

Conclusions

Our results showed that different mitochondrial apoptotic blocks mediated acquired panHER2 resistance in HER2+ breast cancer cell lines as well as highlighted the potential of BH3 profiling assay in prediction of panHER2 resistance in breast cancer cells.

Legal entity responsible for the study

Ozgur Kutuk

Funding

Baskent University

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

13P - AXL as a potential primary and secondary trastuzumab resistance mechanism in breast cancer cells with HER2 overexpression (ID 4867)

Presentation Number
13P
Lecture Time
13:15 - 13:15
Speakers
  • J. Cejalvo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Breast cancer (BC) is a heterogeneous disease, HER2+ represents between 15-30% of all subtypes. Trastuzumab (T), a monoclonal antibody able to inhibit HER2 activation, has been successfully employed in HER2 amplified tumors both in adjuvant and in metastatic settings, conferring an improvement in DFS, PFS and OS. Despite these results, many patients experience primary or secondary resistance to therapy. The mechanism of resistance is unclear, our aim is to assess AXL, a receptor tyrosine kinases (RTK) implicated in epithelial-to-mesenchymal transition, as potential mechanisms of resistance.

Methods

We used two cell lines to investigate possible mechanisms of primary and secondary resistance to T in HER2+ and hormone receptor negative BC. AU565 sensitive to T (AU565-S), and HCC1954 a primary T-resistant cell line. A third cell line with acquired resistance to T (AU565-R) was generated by treating AU565-S cells with constant dose of T (15mg/mL) for 4 months. Cell viability was estimated by MTT assay. We explored the expression of AXL by Western blot (WB) and quantitative reverse transcriptase PCR (qRT-PCR).

Results

The cell viability analysis at 7 days assay confirmed AU565-S as sensible to T, HCC1954 as primary resistant and the development of a secondary resistance to T (AU565-R) (50% of increased viability from AU565-S). HER2 overexpression in all three cells lines were confirmed by WB and FISH. qRT-PCR indicated an important up-regulation of AXL at mRNA levels in AU565-R and HCC1954 compared to AU565-S (p < 0.05). In the same line, WB analyses showed a significantly increase in AXL protein expression in AU565-R and HCC1954 (2.03 and 7.37 fold, respectively). Finally, a selective AXL inhibitor (TP-0903) has demonstrated reduction of viability in all cell lines and significant restoration of sensitivity to T in AU565-R (p < 0.01).

Conclusions

Our results suggest: 1) AXL could be a potential mechanism of both primary and secondary resistance to T; 2) combination therapy with AXL inhibitor plus T restored T sensitivity in in vitro model with AXL overexpressed. These results merit further study and to explore this RTK as possible therapeutic targets in case of anti-HER2 treatment failure.

Legal entity responsible for the study

Hospital Clinico Universitario of Valencia. Biomedical Research Institute INCLIVA

Funding

None

Disclosure

J.A. Perez Fidalgo: Received fees from AstraZeneca, Ipsen, Novartis, Pfizer and Roche for participation in speaker bureaus. Had travel/accomodation expenses paid/reimbursed by AstraZeneca, Roche and Sandoz. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

14P - Alterations to trastuzumab-induced antibody-dependent cell-mediated cytotoxicity (T-ADCC) in a lapatinib-resistant HER2+ breast cancer cell line model (ID 3555)

Presentation Number
14P
Lecture Time
13:15 - 13:15
Speakers
  • N. Gaynor
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Lapatinib is a dual targeting (EGFR/HER2) small molecule tyrosine kinase inhibitor (TKI) approved for the treatment of HER2+ breast cancer. Lapatinib treatment can often lead to acquired resistance and refractory disease. There is no data available on the sensitivity of lapatinib-resistant breast cancer cells to immune cell-mediated cytotoxicity. To investigate the consequences of a lapatinib resistant phenotype on the immune response, we examined T-ADCC and the expression levels of immune-related proteins in a cell line model of lapatinib-resistant HER2+ breast cancer.

Methods

Lapatinib-resistant SKBR3 cells (SKBR3-Lap) were generated by continuous exposure to 250nM lapatinib for 6 months alongside untreated parental cells (SKBR3-Par). FACS-based assays employing peripheral blood mononuclear cells (PBMCs) isolated from healthy volunteers were used to measure direct PBMC-mediated cytotoxicity and T-ADCC. Comparative DNA microarray studies (SK-Par vs. SK-LAP) identified differentially expressed immune-related genes which were subsequently examined at the protein level by Western blot.

Results

T-ADCC was 45-50% lower in SKBR3-Lap compared to the SKBR3-Par cell line at the three effector to target cell ratios examined - 1:1 (p = 0.01), 5:1 (p = 0.001) and 10:1 (p = 0.024). Direct immune cell-mediated cytotoxicity was low (<7% at 10:1) for both cell lines. Microarray data identified significant alterations in the growth factor receptor EGFR, the immunosuppressive adenosine receptor A2AR and immune response modulating MHC Class I, HLA-E, and NKTR in SK-LAP compared to SK-Par. Western blots established changes to these targets at the protein level. In addition, protein levels of HER2 were not significantly reduced and programmed death ligand 1 (PD-L1) levels were increased (p = 0.045) in SKBR3-Lap.

Conclusions

Resistance to lapatinib is associated with an attenuated T- ADCC response and an altered profile of immune-related proteins in this HER2+ breast cancer model. Further investigation is warranted to explore if targeting proteins such as A2AR or PD-L1 could play a role in ADCC response in this model.

Legal entity responsible for the study

Cancer Biotherapeutics, National Institute for Cellular Biotechnology, Dublin City University

Funding

Cancer Clinical Research Trust

Disclosure

N. O'Donovan: Research funding from GlaxoSmithKline. J. Crown: Honoraria from Novartis. Research funding from Roche and GlaxoSmithKline. D. Collins: Currently funded by a Roche Postdoctoral Fellowship and has received funding from Roche Products Ireland Ltd. as part of the IRCSET Enterprise Partnership Scheme. All other authors have declared no conflicts of interest.

Disclosure

N. O\'Donovan: Research funding from GlaxoSmithKline. J. Crown: Honoraria from Novartis. Research funding from Roche and GlaxoSmithKline. D. Collins: Currently funded by a Roche Postdoctoral Fellowship and has received funding from Roche Products Ireland Ltd. as part of the IRCSET Enterprise Partnership Scheme. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

15P - Estrogen-dependent breast cancer: The importance of androgen receptor in exemestane treatment (ID 3578)

Presentation Number
15P
Lecture Time
13:15 - 13:15
Speakers
  • C. Amaral
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Exemestane (Exe) is a third-generation steroidal aromatase inhibitor (AI) that is a standard therapeutic approach for post-menopausal women with estrogen-receptor positive (ER+) breast cancer. Besides its clinical benefit, acquired resistance may develop. Thus, to avoid this drawback it is urgent to find new targets that can improve breast cancer treatment. It is known that 85-95% of the ER+ breast cancers, overexpress androgen receptor (AR), that has a dual function in breast cancer depending on hormonal cell status. It has been described that in AIs-sensitive breast cancer cells this receptor promotes cell death. Several clinical trials are ongoing to study the efficacy of combining AR antagonists, as bicalutamide (CDX), with Exe, but the benefit of targeting AR is not well defined. In that way, this work will investigate the biological significance of AR in Exe-treated breast cancer cells and the effectiveness of targeting AR.

Methods

In ER+ breast cancer cells that overexpress aromatase (MCF-7aro), it was investigated the in vitro effects of the AR antagonist CDX in Exe-treated cells. The cell impact in viability and cell proliferation was studied using MTT assay and flow cytometry, respectively. The cell death was explored by evaluating caspase activities. The expression/activation of AR and the effects on PI3K and MAPK pathways were studied by Western-blot.

Results

Exe induces an overexpression and hyperactivation of AR in MCF-7aro cells. By blocking AR with CDX, it was observed an increase in the reduction of viability and proliferation of Exe-treated cells, when comparing to Exe alone. An increase in activation of caspases-9, -8 and -7 was also observed for the combination. In addition, CDX inhibits the Exe-induced activation of cell proliferation/survival MAPK pathway and caused no effect on PI3K pathway.

Conclusions

This study suggests that, contrary what is described for other AIs, the AR as a pro-survival role in sensitive breast cancer cells treated with Exe and that by targeting AR it is possible to improve the clinical efficacy of Exe, by inhibiting cell proliferation and inducing apoptosis. This work contributes to the understanding of the link between AR and Exe and will highlight new targets to improve breast cancer treatment.

Legal entity responsible for the study

UCIBIO, REQUIMTE, Faculty of Pharmacy, University of Porto

Funding

Fundação para a Ciência e a Tecnologia (FCT): Amaral C. (SFRH/BPD/98304/2013) and Augusto T. (BD/128333/2017) grants

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

16P - Phosphatidylinositol 3-kinase (PI3Kα)/AKT axis blockade with taselisib or ipatasertib enhances the efficacy of anti-microtubule drugs in human breast cancer cells (ID 929)

Presentation Number
16P
Lecture Time
13:15 - 13:15
Speakers
  • A. Diana
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The phosphatidylinositol 3-kinase (PI3Ks) pathway is commonly altereted in breast cancer patients, but its role is still unclear. Taselisib, a mutant PI3Kα selective inhibitor, and ipatasertib, an AKT inhibitor, are currently under investigation in clinical trials in combination with paclitaxel or hormonal therapies in breast cancer. The aim of this study was to evaluate if PI3K or AKT inhibition can prevent resistance to chemotherapy and potentiate its efficacy.

Methods

The efficacy of combined treatment of ipatasertib and taselisib plus vinorelbine or paclitaxel or eribulin was evaluated in vitro on human breast cancer cells (with different expression profile of hormonal receptors, HER2, and of PI3Kα mutation) on cell survival by using MTT (3,(4,5-dimethylthiazol-2)2,5 difeniltetrazolium bromide) and on cell apoptosis by flow-cytometry analysis. We also investigated the effect of combined treatment on downstream intracellular signaling, by western blot analysis, and on metastatic properties, by migration assays. Finally, we analyzed changes in cell cytoskeleton by immunofluorescence.

Results

A significant synergism of ipatasertib or taselib plus anti-microtubule chemotherapy in terms of anti-proliferative, pro-apoptotic and anti-metastatic effect was observed. The combined treatment completely inhibited the activation of proteins downstream of PI3K and MAPK pathways and affected the expression of survivin. Combined treatments completely disorganized the cytoskeleton in human breast cancer cells, thus suggesting a potential mechanism for this combination.

Conclusions

Targeting PI3K may enhance the efficacy of anti-microtubule drugs in human breast cancer.

Legal entity responsible for the study

Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy.

Funding

This work has been supported by Associazione Italiana Per La Ricerca Sul Cancro (AIRC)-Project MFAG 2013-N.14392 and drugs were kindly provided by Genentech (Research proposal nr. OR-214726 for taselisib and nr. OR-214797 for ipatasertib).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

18P - Potential miRNAs involved in molecular pathways mediating the anticancer effects of short term starvation in breast cancer cells treated with doxorubicin (ID 5115)

Presentation Number
18P
Lecture Time
13:15 - 13:15
Speakers
  • D. Fanale
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In recent years, increasing evidences showed that several types of dietary approaches restricting food intake, including Short Term Starvation (STS), may exert a protective role against aging and other age-related pathologies as well as cancer. Interestingly, the dietary restriction showed significant anticancer effects able to prevent cancer onset, slow its progression and improve therapy response. Since recent studies showed that miRNAs may modulate sensibility/resistance to antiblastic therapy, the aim of our study was to investigate the STS-induced molecular changes in breast cancer cells treated with doxorubicin, focusing our attention on miRNA expression profile.

Methods

Vitality assays were used to assess the effects of STS on cell proliferation. Using a TaqMan Low Density Array A human microRNA microarray analysis, the expression profile of 377 miRNAs was analyzed in healthy and malignant breast cells, MCF10A and MDA-MB-231 respectively, treated for 24h with 1µM doxorubicin under STS conditions for 48h. In addition, the expression of mRNAs and miRNAs specifically induced by STS was analyzed in MCF-7, MDA-MB-231 and SkBr3 cells using Real-time PCR analyses.

Results

In vitro cell vitality assays showed that STS, in association with doxorubicin treatment, significantly reduces breast cancer cell proliferation and viability, whereas it appears to protect healthy breast cells from chemotherapeutic treatment. Microarray analysis showed that a subset of miRNAs involved in molecular pathways related to drug sensitivity/resistance was found to be differentially expressed in breast cancer cells following the doxorubicin treatment and STS. Finally, expression analysis of hypothetical miRNA gene targets involved in therapy response have confirmed the coherence of our results.

Conclusions

This work establishes, for the first time, an interesting link between anticancer effects of STS and miRNA expression changes in doxorubicin-treated breast cancer cells, suggesting the potential involvement of some miRNAs in molecular pathways mediating the effects of STS in breast cancer.

Clinical trial identification

Not applicable

Legal entity responsible for the study

Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

19P - An endothelial premetastatic-like niche is promoted by tumor-secreted factors derived from highly metastatic breast cancer cells in vitro (ID 4365)

Presentation Number
19P
Lecture Time
13:15 - 13:15
Speakers
  • M. Gallardo Vera
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

It is well established that primary tumour can modify secondary organs long before the circulating tumor cells reach their metastatic targets. These modifications are mainly exerted through tumor-secreted factors and can act on several cell types to form what is called the pre-metastatic niche. Pre-metastatic endothelial niche play a key role in the extravasation process during metastasis. Here we describe the effect of the mixture of secreted factors by tumor cells over endothelial changes that facilitates tumor cell transendothelial migration.

Methods

Adhesion assay Human umbilical vein endothelial cells (HUVEC) cells were stimulated or not with TNF-α [10ng/ml] or Tumor Secreted Factors (TSFs) [10 µg/ml] from MDA-MB-231 or MCF-7 cell lines. HUVEC monolayers were co-cultured with a U937(3H) cells. After 3h firm attached U937(3H) were lysed and radioactivity was measured. Vascular permeability assay HUVEC monolayers were cultured in Boyden chambers. Cells were stimulated or not with TNF-α [10ng/ml] or TSFs [10 µg/ml] and incubated for 12h. Afterwards a dextran-FITC solution was added for 20 min and the bottom well fluorescence was quantified. Transendothelial migration assay HUVEC monolayers were cultured in Boyden chambers. Cells were stimulated or not with TNF-α [10ng/ml] or TSFs [10 µg/ml] and incubated for 10h. Fluorescent labeled MDA-MB-231 cell suspension 2x104 was added (24h) and the migrant cells were counted under an epifluorescence microscope.

Results

Adhesion assay revealed that HUVEC stimulated with MDA-231 TSFs attached U937 cells 6-fold comparing to the MCF-7 TSFs or control, in a similar way as TNF-α did. The MDA-TSFs were able to increase the HUVEC monolayer permeability exceeded about 30% of the TNF-α induced permeability. A 1.5-fold increase of transendothelial migration cells was observed in HUVEC stimulated with MDA-MB-231 TSFs.

Conclusions

Tumor secreted factors derived from highly metastatic cell line MDA-MB-231 are capable to induce a premetastatic-like endothelial state, increasing the tumor cell transendothelial migration, adhesion to the HUVEC monolayer as well as vascular permeability enhancement.

Clinical trial identification

11-62-2014

Legal entity responsible for the study

Instituto de Investigaciones Biomédicas, UNAM

Funding

Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), fellowship 509589 from CONACYT.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

20P - Selective accumulation of the rat adherent natural killer cells in mammary tumor tissues (ID 4424)

Presentation Number
20P
Lecture Time
13:15 - 13:15
Speakers
  • S. Gu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In the present study, we attempted to clarify what kind of adhesion molecules and tumor cytotoxic killer activity A-NK cells can express, when cultured for long period in vitro, and then tried experimentally to augment the selective accumulation the A-NK cells into rat mammary tumors, in combination with the prior injection of various kinds of adjuvants into the tumor region. The mechanisms by which the effector cells accumulate in tumor tissue will be discussed.

Methods

1. Animals: Specific pathogen-free (SPF) female rats. 2. Preparation of A-NK cells: A-NK cells were isolated from splenic lymphocytes. 3. Antibodies: Monoclonal antibodies, and Anti adhesion molecule antibodies. 4. Immunohistochemical staining and Flow cytometric analysis. 5. Preparation of mammary tumor bearing rats.

Results

Immunocytochemical and flow-cytometric analysis revealed that most of the A-NK cells strongly expressed lymphocyte-function-associated antigen 1 throughout the incubation. All A-NK cells from 8-150-day cultures, particularly those cultured for 8 days, showed significant cytolytic activity against all targets. Peritumoral injection of various kinds of adjuvant, particularly Freund's complete adjuvant plus bacillus Calmetee-Guerin, resulted in a marked accumulation of A-NK cells in mammary tumor tissues 24 h after injection, and simultaneously in the formation of vessels resembling high-endothelium venules, and expression of the ICAM-1 molecule on the tumor cells in the sites of tumor tissues. When A-NK cells were intravenously administered, significant retardation of tumor growth and prolongation of survival of tumor-bearing rats were observed in the groups that received the prior injection of adjuvants.

Conclusions

These results indicate that the prior injection of proper adjuvant into the peritumoral region is effective for the selective accumulation or infiltration of A-NK cells into the sites of tumor tissues, and results in the marked retardation of tumor growth.

Clinical trial identification

none

Legal entity responsible for the study

School of Rehabilitation Sciences

Funding

None

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster display session Poster Display session

21P - Inhibition of nitric oxide synthase (NOS) reduces the effect of stress hormone signalling in breast cancer (ID 3086)

Presentation Number
21P
Lecture Time
13:15 - 13:15
Speakers
  • R. Flaherty
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Expression of nitric oxide synthase (NOS) has been found to correlate with tumour progression in breast cancer, indicating that NO activity may drive malignant growth. Previously we have shown that the stress hormone cortisol acts through a nitric oxide synthase (NOS) mediated pathway to induce production of nitric oxide (NO), and can induce DNA damage in breast cancer.

Methods

Breast cancer cell lines MCF-7 and MDA-MB-231 as well as the mouse mammary tumour cell line 66CL4 were exposed to cortisol and levels of intracellular NO were measured using composite electrochemical sensors. DNA damage was quantified using immunofluorescence and expression of iNOS and metastatic markers VEGF and TWIST were examined using qPCR. An in vivo syngeneic breast cancer model was also used to examine the effect of L-NAME, a NOS inhibitor, on tumour aggressiveness and metastasis in conjunction with daily restraint stress (2hrs) (n = 4/group, repeated in duplicate).

Results

Cortisol significantly increased the expression of iNOS, the generation of NO and DNA damage in breast cancer cells and this was blocked by the NOS inhibitor L-NAME. A significant increase in VEGF and TWIST expression was also observed in response to cortisol. Furthermore, L-NAME also significantly reduced primary tumour growth in stressed mice and reduced the number of metastatic sites/mouse. Tumour microvasculature (as evidenced by CD31 expression) was significantly increased in stressed mice and this was reduced with L-NAME treatment.

Conclusions

We demonstrated that L-NAME, through inhibition of NO signalling, is effective in reducing primary tumour formation and metastatic potential in stressed mice. This data may have impact for patients with breast cancer experiencing extreme stress and further genomic analysis are ongoing.

Legal entity responsible for the study

School of Pharmacy and Biomolecular Sciences, University of Brighton, UK

Funding

Rising star initiative, University of Brighton.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

22P - The interplay between TP53 and mevalonate pathway in ovarian cancer (ID 4777)

Presentation Number
22P
Lecture Time
13:15 - 13:15
Speakers
  • M. Abdullah
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

TP53 gene is the most commonly mutated tumour suppressor in human malignancies. TP53 is mutated in more than 50% of all human cancers, with over 96% of high-grade serous ovarian cancer displaying changes at this locus. Mutations of TP53 gene is associated with malignant transformation and resistance to chemotherapy. In addition, previous studies have shown that ectopic expression of TP53 mutant form in breast cancer cells leads to increased transcription of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR). This enzyme regulates the synthesis of geranylgeraniol which is used to post-translationally modify small GTPase oncogenes. HMGCR is itself considered to be a metabolic oncogene. Statins, which inhibit HMGCR, are potential cancer therapeutics which can cause ovarian cancer (OC) cell apoptosis and regression of xenografts.

Methods

The level of mevalonate pathway (MP) enzymes evaluated in panel of OC cell lines using immunoblotting. in addition, MP enzymes expression were evaluate using qPCR following ectopic expression of wild-type and R248W, R175H, and R273H p53 variants in Skov-3 cells and after inhibition of TP53 expression using siRNA directed to TP53 mRNA in Ovcar-3 cells.

Results

We confirmed that the expression of HMGCR is higher in OC cell lines than in normal epithelial ovarian cells. The level of geranylgeranyl transferase I-β (GGTI-β) and Geranylgeranyl transferase II-β (GGTII-β) was significantly higher in a subset of OC cell lines. The ectopic expression of TP53 variants in Skov-3 cells, which lack endogenous p53 protein, led to significantly increased expression of HMGCR, GGTI-β, GGTII-β and Farnesyltransferase-β (FT-β) enzymes compared to cells transfected with vector. The inhibition of the pre-existing mutations in TP53 encoding R248Q in Ovcar-3 cell line significantly decreased p53 protein and also HMGCR, GGTI-β, GGTII-β and FT-β mRNA.

Conclusions

These data suggest that TP53 mutations play critical role in regulation of the activity of MP enzymes, providing a rationale for the evaluation of the pathway inhibitors such as statins and bisphosphonates in the treatment of OC.

Legal entity responsible for the study

The study was designed by AR and MIA, the experimental work was conducted by MIA and MNA.

Funding

Higher Committee for Education Development in Iraq (MIA ref D-11-296).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

23P - Epigenomic landscape of breast cancer in very young women (ID 4482)

Presentation Number
23P
Lecture Time
13:15 - 13:15
Speakers
  • S. Oltra Sanchis
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Although less frequent than in older women, breast cancer in very young women (BCVY) (≤35 years old) presents more aggressive and complex biological features. Epigenetic modifications such as miRNA regulation or DNA methylation are reported to play an important role in the onset and progression of cancer. The aim of this work is to identify the epigenetics mechanisms characteristics of BCVY that may be conferring more aggressive features to this group of patients.

Methods

We analysed methylation (Infinium MethylationEPIC BeadChip) from 26 BCVY and 15 samples from women >45 years old. Methylation differences were assessed using Wilcoxon rank sum test. We selected from The Cancer Genome Atlas (TCGA) those genes regulated by significantly different methylated sites and their expression was analysed. MiRNA expression data from TCGA, METABRIC and data previously published from our group was evaluated in a meta-analysis. We then selected those target genes which expression was more affected by miRNA deregulation. Pathway enrichment analysis was performed with most relevant genes from the epigenetic study by Enrichr.

Results

We detected a global hypomethylation profile in BCVY samples and hypermetilation of 502 specific CpG sites exclusive of this group of age. Hypomethylated CpG sites were regulating genes mainly involve in neuronal processes, extracellular matrix and cell communication. Whereas specific hypermethylation was located in genes related to immune system, NOTCH signalling, vesicular trafficking, DNA repair and senescence. MiRNA expression meta-analysis revealed a profile of 22 miRNAs significantly deregulated in BCVY. Pathway enrichment analysis of most affected target genes showed an involvement in neural processes, glucose metabolism, vesicular trafficking, DNA repair, histone and chromatin related proteins, apoptosis, cell cycle, response to DNA damage and senescence.

Conclusions

Our work highlights the presence of epigenomic profile distinctive of BCVY. Both methylation and miRNAs studies points to deregulation of pathways related to neural processes, vesicular trafficking, DNA repair and senescence. All these processes may lead to cancer development and progression, thus genes in these pathways may be potential candidates for further studies.

Legal entity responsible for the study

INCLIVA Research Institute

Funding

None

Disclosure

S. Sanchis: Funded on a FPU predoctoral Fellowship (FPU13/04976) from MINECO, Spanish Government.

rn

G. Ribas: Funded on a Miquel Servet II contract (CPII14-00013) from CarlosIII Health Institute.

rn

M.P. Chilet: Funded by private Patients Fundation LeCado. CIBERONC is an initiative of the Carlos III Health Institute.All other authors have declared no conflicts of interest.

Disclosure

S. Sanchis: Funded on a FPU predoctoral Fellowship (FPU13/04976) from MINECO, Spanish Government.

\r\n

G. Ribas: Funded on a Miquel Servet II contract (CPII14-00013) from CarlosIII Health Institute.

\r\n

M.P. Chilet: Funded by private Patients Fundation LeCado. CIBERONC is an initiative of the Carlos III Health Institute.All other authors have declared no conflicts of interest.

Disclosure

S. Sanchis: Funded on a FPU predoctoral Fellowship (FPU13/04976) from MINECO, Spanish Government.

G. Ribas: Funded on a Miquel Servet II contract (CPII14-00013) from CarlosIII Health Institute.

M.P. Chilet: Funded by private Patients Fundation LeCado. CIBERONC is an initiative of the Carlos III Health Institute.All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

24P - Evaluation of cell free circulating DNA in plasma by digital PCR for early diagnosis in Peruvian breast cancer patients (ID 5096)

Presentation Number
24P
Lecture Time
13:15 - 13:15
Speakers
  • A. Aguilar
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

New diagnostic tools can be useful and give clinical benefits, including diagnosis, prognosis, treatment and monitoring of the disease. A new non-invasive method is the study of liquid biopsies, performed in fluids containing cell-free circulating DNA (cfDNA). Analyses with digital PCR technique (dPCR) allows to establish the levels of cfDNA as well as the absolute quantification of mutant alleles with accuracy. This system scatters the sample among twenty thousand wells (microfluids on the chip), where amplification reactions occur independently and are recorded by a reader.

Methods

Peripheral blood samples were obtained from breast cancer patients and healthy controls. From each sample, the cfDNAs were extracted from plasma using the MagMAX™ Cell Free DNA Isolation Kit and dPCR was done for quantification of samples. We evaluated two genes, PUM1 and RNaseP. For amplification of fragments, master mix 1X (Applied Biosystems) and PCR detection assays of both genes were combined with 1.5 μl of plasma cfDNA, dispersed in the chips and placed in a ProFlex ™ thermocycler (Applied Biosystems) following the program pre-established by the manufacturer, with additional five cycles. Finally, quantification data were obtained with QuantStudio® 3D AnalysisSuite™ Cloud Software. Comparison of DNA concentration in copies per microliter and other statistical calculations were performed with InfoStat 2015.

Results

Significant differences were found in the values of cfDNA between patients and controls for PUM1 (p = 0.0001) and RNase P (p = 0.0003). These results allowed to establish cut-off points between groups at 78,995 and 51,154 copies/uL, respectively. These values can be considered in the classification of groups for further analysis of others samples. Statistical support for the use of markers in diagnosis was also evaluated using the ROC curve that favors the PUM1 marker, with a sensitivity of 75% and a specificity of 95.2%.

Conclusions

Based on the significant differences found between breast cancer patients and controls, cell free DNA is a good biomarker that can be used in the diagnostic of breast cancer. On the other hand, digital PCR has been established as a good tool to check cfDNA levels from plasma of breast cancer patients.

Legal entity responsible for the study

Jose Buleje

Funding

Programa Nacional para la competitividad y Productividad (Innovate Perú)-No.138-PNICP-PIAP-2015, Universidad de San Martin de Porres, Oncosalud - AUNA

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

25P - Breast cancer predisposing germline mutations identified by exome sequencing (ID 3645)

Presentation Number
25P
Lecture Time
13:15 - 13:15
Speakers
  • E. Kuligina
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

A significant portion of hereditary predisposition to breast cancer (BC) is attributed to yet unknown factors. Russian population is characterized by surprisingly strong founder effect, therefore whole exome sequencing (WES) for a limited number of these genetically homogenous patients has a potential to identify novel BC-predisposing genes.

Methods

WES was performed for 32 Russian BC cases, which demonstrated strong clinical signs of the hereditary disease (family history, BC bilaterality, young onset) and lacked germline mutations in “canonical” BC genes (BRCA1, BRCA2, CHEK2, PALB2, and NBS1/NBN).

Results

Eight patients carried potentially pathogenic mutations in BRCA1 network genes (3 truncations (BLM p.Q548*, RAD51C c.904 + 1G>A, FANCM p.S497fs) and 5 missense mutations (FANCM (p.R100W, p.Q891P), ERCC4 p.R799W, RAD54L p.R394W, RAD50 p.D515G)). The remaining 24 patients were analyzed for the presence of rare non-silent genetic variants; in total, 15437 alleles had ExAC frequency <1%. Use of ACMG-guided bioinformatic pipeline classified these variants for 64 ‘pathogenic/likely-pathogenic’, 12844 of ‘uncertain significance’, and 2529 ‘benign/likely-benign’. Prevalence of 69 top candidates was compared in 640 genetically enriched BC patients, 1200 consecutive BC, 1200 middle-aged healthy females and 460 elderly healthy women. Several likely pathogenic mutations were overrepresented in the BC groups: nonsense PZP p.R680* [9/1845, OR = 13.2]; heterozygous missense LEPREL1 p.P636S [6/1778; OR = 4.4] and ING1 p.P319L [3/1792; OR = 3.9]; homozygous missense BRCA1 p.Q356R [12/1683; OR = 5.5] and EXO1 p.G759E [9/1606, OR = 4.3]. Some potentially pathogenic variants occurred only in the index cases but were absent in other BC patients (rare ExAC alleles: HELLS p.R53C and TP53INP1 p.E27D; newly identified variations: MLH3 p.C1393F, EMSY p.G934* and ATRIP p.R760*).

Conclusions

This study revealed several alleles, which may be associated with increased BC predisposition. However, in contrast to well-known Slavic BRCA founder mutations, newly identified candidates are exceptionally rare and therefore are unlikely to be responsible for a significant share of BC morbidity. Supported by the RSF grant No 16-45-02011

Legal entity responsible for the study

Evgeny N. Imyanitov, Head of the Department of Tumor Biology in the N.N. Petrov Institute of Oncology

Funding

Russian Scientific Fund (grant No 16-45-02011).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

26P - Global transcriptome deregulation of breast cancer in very young women samples (ID 4703)

Presentation Number
26P
Lecture Time
13:15 - 13:15
Speakers
  • V. Vidal Tomas
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Breast cancer in young women (under 35 years) (BCVY) often presents distinct clinic-pathological features: more aggressive phenotype and worse prognosis than older women. Genomic and molecular alterations play a significant role in breast cancer biology. Due to the low incidence of BCVY (2-5%) these women are underrepresented in most molecular studies. This work presents a comprehensive study of the transcriptome in BCVY, focusing in the search of gene expression biomarkers characteristic of this group of patients.

Methods

We analysed the transcriptome by Clariom™ D (Affymetrix) from 31 BCVY and 11 samples from women >45 years old. Global gene expression was filtered and normalized by RMA method. After initial pre-processing we analysed expression in 3,639 mRNAs, 66,457 lncRNA and 3,271 pre-miRNA and differences were assessed using t-test. We performed a meta-analysis with gene expression data from The Cancer Genome Atlas (TCGA) for validation of results. Pathway enrichment analysis was performed by Enrichr.

Results

showed a specific transcriptomic landscape in BCVY. Clariom D study revealed 134 significant mRNA with p-value< 0.05 that pointed out towards pathways related with olfactory receptors, GPCR signalling, tight junction and cell-cell communication. After meta-analysis with TCGA gene expression data and own data, 43 genes were statistically significant and 15 of those withstood FDR correction (FDR< 0.05). Among those we found PIK3CB, HOXD10, ZNF654, TMEM204, IRX5, PF4, MAGEA2 and TSR2 deregulated in BCVY compared with older women. Pathway enrichment analyses and GO search highlight pathways related to cell-cell communication, cancer processes, chemokine and PI3K signalling pathways, cell differentiation, extracellular matrix, vesicular trafficking, neuronal processes among others.

Conclusions

We find the presence of a distinctive transcriptomic profile of the BCVY samples. Our study points to deregulation of pathways related to cell migration, proliferation and differentiation that promote cancer development and progression. Genes obtained in meta-analyse might be potential target genes for further studies in BCVY which could help to clarify the biological background for the development of the disease in this group of age.

Legal entity responsible for the study

INCLIVA Instituto de investigación

Funding

None

Disclosure

S. Sanchis: Funded on a FPU predoctoral Fellowship (FPU13/04976) from MINECO, Spanish Government. G. Ribas: Funded on a Miquel Servet II contract (CPII14-00013) from CarlosIII Health Institute. M.P. Chilet: Funded by private Patients Fundation LeCado. CIBERONC is an initiative of the Carlos III Health Institute. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

27P - Integrated system-level analyses of androgen receptor variant networks to identify novel prostate cancer-relevant genes that serve as prognostic biomarkers (ID 922)

Presentation Number
27P
Lecture Time
13:15 - 13:15
Speakers
  • F. Magani
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Castration resistant prostate cancer (CRPC) is a rapidly progressing disease state for which there is no cure. The constitutively active androgen receptor (AR) splice variant AR-V7 represents a well-established mechanism of therapeutic resistance and disease progression. This variant lacks the AR C-terminal ligand binding domain and, as such, is not inhibited by androgen deprivation therapy. Designing high-affinity drugs to target the amino terminus of AR and AR-V7 is a major challenge due to the intrinsic disorganized structure of this region. Thus, there is an imperative need to identify novel AR-V7 hub genes in PC that may serve as novel therapeutic targets.

Methods

We performed a highly robust gene expression meta-analysis on PC patient samples. We defined gene modules correlated with PC progression using a Weighted Gene-Co-expression Network Analysis (WGCNA), a powerful systems biology approach. Further, we identified AR-V7 downstream target genes using gene expression profiling and mapped the AR-V7 functional interactome for the first time using a novel high-throughput synthetic genetic array screen in yeast. Finally, we combined the results from our three independent system-level analyses with experimental data to identify hub genes that were upregulated in PC patients, upregulated by AR-V7, and that also functionally interacted with AR-V7.

Results

The identified genes not only included select genes previously linked to PC, such as members of the topoisomerase and cyclin families, but also novel genes that had not been previously linked to PC progression. The identified gene-signature expression correlated with patients’ Gleason score and had a prognostic value that predicted disease free-survival at the time of patient biopsy in large independent cohorts.

Conclusions

In sum, we show here an unbiased integrated system-level analysis of AR-V7 networks, where we combined bioinformatic analysis of patient samples and cell-based approaches to identify new candidate genes in CRPC that may serve as novel prognostic markers and future targeted therapies.

Legal entity responsible for the study

University of Miami Miller School of Medicine

Funding

Sylvester Comprehensive Cancer Center

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

28P - ODM-208, a novel CYP11A1-inhibitor as a therapeutic approach for the treatment of castration-resistant prostate cancer (ID 2858)

Presentation Number
28P
Lecture Time
13:15 - 13:15
Speakers
  • R. Oksala
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Androgen receptor (AR) plays a central role in prostate cancer and continues to be a driver in castration-resistant prostate cancer (CRPC), with increased AR expression in most cases. Approximately half of the men with CRPC respond initially to abiraterone or enzalutamide, but most relapse within 1 to 2 years. Majority of the abiraterone and enzalutamide-resistant tumors have still high AR expression and persistent AR activity. Several precursor steroids, like testosterone (T) and dihydrotestosterone activate AR, are synthesized in adrenal glands and de novo in tumours. CYP11A1 (cytochrome p450scc) is a mitochondrial enzyme catalysing the conversion of cholesterol to pregnenolone (Preg), which is the first rate-liming step in steroid hormone biosynthesis. ODM-208 is a novel, oral, non-steroidal and selective inhibitor of CYP11A1 enzyme and suppresses the synthesis of all steroid hormones and precursors.

Methods

The inhibition of CYP11A1 was measured in vitro by detecting the formation of radiolabelled isocapronic acid in a human adrenal cortex cell line (H295R), and further analysing Preg and T formation by ELISA. Inhibition of the adrenal and testicular hormone production in vivo was tested in the intact male rat assay by analysing plasma concentrations of progesterone (P), corticosterone (C) and T (with LS-MS/MS) after single oral dose of ODM-208. The tumor growth inhibition was studied by using androgen dependent VCaP cells, which were subcutaneously grafted to intact male nude mice. When tumor volumes reached on average 200 mm3, mice were castrated, and after regrowth of the tumors, the oral treatment of ODM-208 was started.

Results

ODM-208 potently inhibits CYP11A1 enzyme and formation of Preg and testosterone with low nM concentrations in vitro. In male rats, clear decreases of P, C and T concentrations can be detected already after single oral administration of ODM-208. In the murine VCaP CRPC xenograft model ODM-208 significantly inhibited tumor growth.

Conclusions

ODM-208 shows promising antitumor activity in preclinical CRPC models and suggests that ODM-208 may have the potential to be an effective treatment in CRPC. Clinical trial in patients with metastatic CRPC is planned to be started in the 2018.

Legal entity responsible for the study

Orion Corporation Orion Pharma Orion Corporation Orion Pharma

Funding

Orion Corporation Orion Pharma

Disclosure

R. Oksala, M. Karimaa, M. Ramela, R. Riikonen, P. Rummakko, G. Wohlfahrt, A. Vuorela, M.V. Mustonen, P. Kallio: Employee: Orion Corporation Orion Pharma. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

29P - Determining the role of the ETS factor ELF3 in normal and malignant prostate (ID 3214)

Presentation Number
29P
Lecture Time
13:15 - 13:15
Speakers
  • L. Archer
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Aberrations in the ETS transcription factor family members are a common feature of multiple cancers including prostate cancer (PCa), such as the TMPRSS2:ERG fusion. ELF3, also known as ESE-1, is an epithelial-specific ETS transcription factor involved in regulating cell differentiation in various tissues, however, its role in the prostate is controversial. The aim of this study was to identify the function of ELF3 in normal prostate development and to explore its role in PCa.

Methods

Three model systems were used: prostate cell lines, primary prostate epithelial cells cultured from patient tissue and paraffin-embedded human tissue sections. The function of ELF3 was investigated using knockdown via siRNA transfection and overexpression via lentivirus transduction. Western blots, immunofluorescence and immunohistochemistry were used to measure protein localisation and levels of expression. Other assays measured cell viability, colony forming ability and migration.

Results

ELF3 expression was restricted to the basal layer of the normal prostate epithelium and was not expressed in stroma. Analysis of a prostate tissue microarray indicated that whilst ELF3 is expressed in benign prostate tissue, its expression is lost in low-grade prostate tumours and re-expressed in some more advanced tumours. ELF3 knockdown resulted in decreased migration, cell viability and did not induce stem cell characteristics, whilst promoting basal cell gene expression. ELF3 overexpression increased cell migration. ELF3 was induced in primary prostate epithelial cells following treatment with the clinically approved HDAC inhibitor Vorinostat, which can promote neuroendocrine differentiation.

Conclusions

ELF3 expression correlates with the normal prostate epithelial cell differentiation hierarchy, and may have a role in advanced PCa. Analysis of total gene expression following knockdown of ELF3 will give an indication of transcriptional networks that are regulated by ELF3. In addition, a lentivirus that expresses an ELF3 mutant, which alters its localisation, will be used to assess any cytoplasmic function of ELF3. This comprehensive and clinically relevant approach will allow complete elucidation of the role of ELF3 in prostate cell differentiation and PCa.

Clinical trial identification

N/A

Legal entity responsible for the study

Norman Maitland

Funding

Prostate Cancer UK (PCUK) registered charity

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

30P - Treatment-induced hypoxia attenuates enzalutamide response and promotes resistance in pre-clinical models of prostate cancer (ID 2887)

Presentation Number
30P
Lecture Time
13:15 - 13:15
Speakers
  • P. Maxwell
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Inhibition of androgen signalling remains the therapeutic mainstay in castrate-resistant prostate cancer. Retention of active AR signalling or acquisition of splice variants have been reported as mechanisms of resistance to the anti-androgen Enzalutamide. Other non-AR dependent mechanisms of resistance have also emerged including acquisition of a hypoxic microenvironment. We propose treatment-induced hypoxia and the induction of angiogenesis may define a novel mechanism of relapse to Enzalutamide.

Methods

Preclinical experiments were conducted in LNCaP tumors and established human prostate cancer cell lines. Tumour growth, intra-tumoral hypoxia and blood vessel density were measured in vivo. AR expression, activation and target gene expression were measured in vitro. Effects of Enzalutamide on hypoxia-driven, disease-progressing pathways and genes of interest and the role of these genes in resistance to Enzalutamide was investigated.

Results

Enzalutamide promoted persistent hypoxia in LNCaP tumours in vivo, followed by increased blood vessel density and restoration of oxygen tension (>14 days). In vitro, hypoxia increased AR expression and transcriptional activity in LNCaP cells and sustained but did not further potentiate high basal AR and ARv7 activity in 22Rv1 cells. Enzalutamide failed to attenuate the concurrent hypoxia-induced HIF-1 and NF-κB signalling, resulting in up-regulation of disease-progressing genes and pathways. Administration of neutralizing antibodies to two hypoxia-regulated genes, IL-8 and VEGF prolonged Enzalutamide-mediated LNCaP tumour growth control over 28 days in vivo (p < 0.001) and re-sensitised enzalutamide-resistant LNCaP cells in vitro.

Conclusions

Enzalutamide-induced hypoxia upregulates the expression of VEGF and IL-8, whose multi-model signalling effects contribute to microenvironment-promoted resistance in prostate tumours.

Legal entity responsible for the study

David Waugh

Funding

Prostate Cancer UK

Disclosure

J. Worthington: Scientific director at Axis Bioservices. D. Waugh: Consulting/advisory role at Almac Diagnostics, Craigavon, Northern Ireland. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

32P - Prognostic impact of KRAS mutation in cell-free DNA in patients with pancreatic cancer (ID 2967)

Presentation Number
32P
Lecture Time
13:15 - 13:15
Speakers
  • M. Kim
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Cell-free DNA (cfDNA) has been known to be released from tumor cells and evaluated potential biomarkers for therapeutic responses. However, the role of cfDNA in pancreatic cancer has not been well studied. Here we selected KRAS mutation which has been known common over 95% of pancreatic ductal adenocarcinoma (PDA) and evaluated applicability as a prognostic marker through the quantitative analysis of cfDNA and KRAS mutation in the patients with PDA.

Methods

Total of 106 PDA patients were enrolled in the study. The concentration and fraction of KRAS mutation were measured by KRAS screening multiplex droplet digital PCR kit (Biorad, USA) in plasma samples.

Results

KRAS mutation was detected in 97.4% of tissue samples and the correlation with cfDNA was 0.561 with 80.5% positivity. KRAS mutation concentration and fractional abundance showed the association with poor survival in both PFS (P <.001 and P = 0.001) and OS (P = 0.003 and P = 0.006) in the entire stage groups. Specially, the impact for survival of KRAS mutation concentration and fractional abundance was obvious in PFS in resectable group (P = 0.016 and P = 0.02). When we analyzed the receiver operating characteristic (ROC) curve to determine whether KRAS mutation in cfDNA have additive benefits with well-known tumor markers CA19-9, combined with KRAS mutation concentration or KRAS fractional abundance, the value of area under the curve (AUC) was significantly higher than the value calculated as CA19-9 alone.

Conclusions

This study represents that KRAS mutation concentration and fractional abundance in cfDNA could be prognostic marker in pancreatic cancer especially in resectable group.

Legal entity responsible for the study

Sun-Young Kong

Funding

This work was supported by grants from the National Cancer Center of Korea (NCC-1510203).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

33P - Isoform-specific functions in pancreatic adenocarcinoma (ID 1257)

Presentation Number
33P
Lecture Time
13:15 - 13:15
Speakers
  • H. Arasanz
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

AKT/PKB is a protein kinase that plays a key role in cancer, which is expressed as 3 isoforms: AKT1 (PKBα), AKT2 (PKBβ) and AKT3 (PKBγ). Although these isoforms are remarkably similar, there is evidence that each isoform yields specific functions which may vary depending on the cell type. Even so, the underlying molecular pathways specifically regulated by each one of them are unknown.

Methods

To gain insight into the role of each isoform in the biology of human pancreatic adenocarcinoma cells, we have silenced each AKT isoform individually using short hairpin RNAs (shRNAs) delivered by lentiviral transduction. Cells transduced with an unspecific shRNA were used as controls. Then, high-throughput quantitative proteomic analyses were performed to evaluate the differential signaling routes altered by silencing of each AKT isoform.

Results

AKT1 silencing induced the upregulation of 57 proteins and downregulation of 58. AKT2 silencing up-regulated and down-regulated 78 and 101 respectively. AKT3 silencing resulted in the upregulation of 88 and downregulation of 93. The expression levels of 45 proteins were altered exclusively after AKT1 knockdown, while 74 proteins and 89 were specifically altered for AKT2 and AKT3 silencing, respectively. AKT1 silencing up-regulated RNA splicing, GPCR and mTOR pathways, and mitochondrial functions such as the respiratory chain, fatty acid metabolism or mitochondrial DNA synthesis. Pathways related to apoptosis and cell migration were inhibited. AKT2 silencing caused the activation of pathways related to apoptosis, splicing, protein folding and some mitochondrial functions. In contrast, other key metabolic pathways such as nucleic acid synthesis, pentose phosphate pathway, cell adhesion and PI3K signalling were down-modulated. Lastly, AKT3 silencing induced increased splicing and mitochondrial functions, regulation of gene expression and snRNA processing. In this case, the pentose phosphate pathway, cell adhesion, apoptosis, protein synthesis and nucleic acid synthesis were also inhibited.

Conclusions

AKT isoforms have specific functions in pancreatic adenocarcinoma. The individual silencing of each isoform induces a differential alteration of molecular pathways involved in main cellular processes.

Legal entity responsible for the study

D. Escors

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

34P - High chemopreventive and therapeutic efficacy of Id1 inhibition in KRAS-mutant (KM) adenocarcinoma (AD) non-small cell lung cancer (NSCLC) (ID 1932)

Presentation Number
34P
Lecture Time
13:15 - 13:15
Speakers
  • M. Roman Moreno
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Id1 is an independent prognostic factor in NSCLC-AD. Id1 silencing impairs cell viability and migration of NSCLC-AD cell lines. KRAS is the most frequently mutant gene in NSCLC with no specific therapies clinically available. Here we evaluate Id1 as a potential chemopreventive and therapeutic target in a humanized mouse model of KM NSCLC-AD.

Methods

Several human lung AD cell lines with known mutations (H1792-604, H2009, H358, H1568, H1437, H1703 and H2126) were selected for Id1 silencing using inducible short hairpin RNA (shRNA). Humanized AD xenograft mouse models were generated by subcutaneous injection of H1792-604 and H2009 cell lines (Id1 silenced or Id1 wild type) in flanks of immunodeficient mice. Id1 silencing was activated at the time of tumor cell inoculation (chemoprevention assay) or once the tumors were established (therapeutic assay).

Results

Id1 inhibition was achieved in all selected cell lines compared to their controls. In vivo, in the chemoprevention assay we observed a significant decrease in tumor volume in mice injected with Id1 silenced H1792-604 cells (60% ± 32.39) compared to the control group (356.29% ± 115.32) (p < 0.001). Moreover, mice injected with Id1 silenced H2009 cells never developed tumors compared to control mice (168.35 ± 68.71) (p < 0.001). In the therapeutic assay, the activation of inducible silencing of Id1 in established tumors induced a significant reduction of tumor volume in both xenograft models. Id1 inhibition induced a partial response in 40% of the tumors after injection of H1792-604 cells and in 100% of tumors in H2009 inoculated mice.

Conclusions

These findings encourage further evaluation of Id1 as a potential therapeutic target in KM NSCLC-AD patients.

Legal entity responsible for the study

Clínica Universidad de Navarra

Funding

Clínica Universidad de Navarra

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

36P - Synergistic effect of vismodegib and cisplatin in NSCLC models via autophagy (ID 4977)

Presentation Number
36P
Lecture Time
13:15 - 13:15
Speakers
  • F. Papaccio
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Platinum-based chemotherapy still represents the standard first-line approach for NSCLC patients, although primary or secondary resistance is frequently observed. Recently the Shh pathway has been associated with resistance to platinum-based chemotherapy in NSCLC. The aim of this work is to investigate whether a combined treatment with cisplatin and the Hedgehog-pathway inhibitor vismodegib could potentiate the anti-tumour effect and to explore possible mechanisms of this synergy.

Methods

Two Human NCSLC cell lines A549 and H460 were treated with single agent Cisplatin, single agent Vismodegib and a combination of the two drugs. MTT cytotoxicity assays were performed and the data were analysed with CompuSyn software. Experiments of apoptosis and cell cycle were done by using flow cytometer. Immunofluorescence with lysoTracker as well as western blot (WB) analysis for the LC3B protein were performed to analyse autophagy.

Results

The CompuSyn analysis showed an important synergistic effect of cisplatin + Vismodegib. Combined treatment induced a significant increase in cellular apoptosis compared with single agent cisplatin. The cell cycle analyses revealed a block in S-phase with the combination treatment. The lysoTracker immunofluorescence assay showed that cisplatin induces an increase of autophagy, while the combination with vismodegib strongly reduces it, finally reverting this effect. These findings were confirmed by WB analysis for LC3B which is significantly increased by single agent cisplatin and reduced by the combined treatment.

Conclusions

Combined treatment with cisplatin and vismodegib has a synergistic effect with an increase in cancer cell apoptosis. Autophagy has been described as a mechanism through which cancer cells escape cisplatin-induced cytotoxicity. Combining cisplatin with vismodegib leads to an inhibition of autophagy, so that it could suggest a new therapeutic approach.

Legal entity responsible for the study

Università della Campania “Luigi Vanvitelli”

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

37P - Association of the rs4567312 variant in the leptin receptor gene with plasma leptin concentrations and lung cancer incidence in the PREDIMED study (ID 4774)

Presentation Number
37P
Lecture Time
13:15 - 13:15
Speakers
  • J. Ramirez Sabio
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Many studies have found leptin related genes involved in tumorigenesis and have suggested it may play a role in the pathogenesis of lung cancer. Leptin Receptor (LEPR) is expressed in many tissues and cells, including lung mucosal cells. LEPR was reported to be associated with tumor cell proliferation and angiogenesis. Our objective has been to estimate the association between the rs4567312-LEPR gene and lung cancer incidence in a Mediterranean population.

Methods

We analyzed 1094 participants (398 men, 696 women) recruited in the PREDIMED-Valencia Study. Participants were high cardiovascular risk subjects aged 67±6 years at baseline. PREDIMED is a multicenter randomized, controlled trial aimed at assessing the effect of the Mediterranean diet (MedDiet) on cardiovascular prevention (primary outcome). Cancer incidence was a secondary outcome in this trial. Demographic, clinical, life-style, biochemical, and genetic variables were obtained. Subjects were followed-up prospectively from 2003 to 2014 (in the extended-follow-up).

Results

We detected 12 new cases of lung cancer from 2003 to 2014 (1.1% cumulative incidence). Tobacco smoking was strongly associated with lung cancer incidence (91.7% of current or former smokers in lung cancer subjects vs 41.5% in the non-cancer participants (p = 0.001). In the whole population, prevalence of the rs4567312 polymorphism was: 95.5% CC, 4.4% CT and 0.1% TT. We also detected in the whole population an association between this polymorphism and plasma leptin concentrations, 26.9±22.9 ng/mL in CC vs 18.4±16.7 ng/mL in T carriers (p = 0.013). We found a strong association between the rs4567312-LEPR polymorphism and lung cancer risk, being higher in carriers of the T-allele. This association remained statistically significant (OR = 7.61; 95% CI: 1.74-33.37 for T-carriers vs CC) even after adjustment for gender, age, tobacco smoking, dietary intervention group (MedDiet vs control diet) and leptin levels.

Conclusions

T-carriers allele in the rs4567312-LEPR polymorphism presented a higher incidence of lung cancer in this Mediterranean population even after adjustment for tobacco smoking and dietary intervention.

Clinical trial identification

Controlled-trials.com number ISRCTN35739639

Legal entity responsible for the study

Instituto de Salud Carlos III and University of Valencia

Funding

Instituto de Salud Carlos III

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

38P - Two-step microarray analysis of cell-free miRNA in plasma of lung cancer patients (ID 4662)

Presentation Number
38P
Lecture Time
13:15 - 13:15
Speakers
  • I. Zaporozhchenko
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Lung cancer (LC) is causing more than 1.3 million deaths worldwide annually. Early detection of LC is critical for survival but despite recent advancements in LC diagnostics most patients are still diagnosed at advanced stages of the disease. The situation is further complicated by high intratumor heterogeneity and general diversity of lung malignancies. Insights into cancer genetics have kindled interest in molecular cancer diagnostics. One of the lucrative sources of prospective LC biomarkers is cell-free circulating miRNAs. These small non-coding RNAs are frequently deregulated in LC. It is also known that miRNAs can travel in bodily fluids for extended periods of time, shielded from degradation by membrane vesicles or other biopolymers. Recently, specific subsets of miRNAs associated with tumor phenotypes and disease progression have been found circulating in blood of cancer patients and suggested as potential biomarkers for LC.

Methods

In the present study, we have investigated the profiles of circulating miRNAs in blood plasma of LC patients and healthy individuals (HD) in order to identify potential markers for lung cancer diagnostics. Small RNAs were isolated from blood plasma of 20 LC patients and 10 healthy individuals (HD) using protocol reported earlier (Zaporozhchenko et al, Anal Biochem, 2015). Profiles of miRNA expression were obtained using miRCURY LNA miRNA qPCR Panels Plasma/Serum (Exiqon). Ratio based normalization was applied to all miRNA’s with call rate higher than 80%.

Results

Statistical comparison using two-way ANOVA identified 241 ratios (98 individual miRNAs) with significantly different expression between LC patients and HD (p < 0.05 after Benjamini-Hochberg correction). Using LASSO penalization models and manual filtering of miRNAs associated with haemolysis, 7 miRNA ratios were identified as best predictors of cancer. Extended set of miRNAs (n = 19) was selected for further verification in an independent sample of 30 LC patients, 20 HD and 10 patients with hyper- and metaplastic endobronchitis using custom miRCURY LNA miRNA qPCR Pick & Mix Panel.

Conclusions

Based on expression in both data sets 5 ratios containing 7 miRNAs were selected for further validation in an extended cohort of LC and cancer-free individuals.

Legal entity responsible for the study

Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russian Federation

Funding

Study has been supported by Russian Foundation for Basic Research (RFBR, grant No. 14-04-01881), BOR grant VI.62.1.4, and Presidium of RAS research program ‘Molecular and Cellular Biology’ No. 6.1.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

39P - DNA methylation of the CYP1A1 and GSTP1 genes and incidence of major cancers (lung, breast and colon) in the PREDIMED-Valencia study (ID 4694)

Presentation Number
39P
Lecture Time
13:15 - 13:15
Speakers
  • J. Ramirez Sabio
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

DNA methylation is an epigenetic determinant of gene expression. Cytochrome P450 (CYP1A1) is a phase 1 xenobiotic metabolizing enzyme. Glutathione S-transferase P1 (GSTP1) detoxifies metabolites and regulates cellular chemical stress and death. Methylation changes in these genes could play a role in the incidence of cancer. Our aims were to analyze DNA-methylation of selected CpG islands in the CYP1A1 and GSTP1 genes at baseline in subjects who had incident cancer and paired controls, and to determine if DNA methylation in cancer patients changed from baseline to the time close to cancer diagnosis.

Background

DNA methylation is an epigenetic determinant of gene expression. Cytochrome P450 (CYP1A1) is a phase 1 xenobiotic metabolizing enzyme. Glutathione S-transferase P1 (GSTP1) detoxifies metabolites and regulates cellular chemical stress and death. Methylation changes in these genes could play a role in the incidence of cancer. Our aims were to analyze DNA-methylation of selected CpG islands in the CYP1A1 and GSTP1 genes at baseline in subjects who had incident cancer and paired controls, and to determine if DNA methylation in cancer patients changed from baseline to the time close to cancer diagnosis.

Methods

We followed-up 1094 subjects (aged: 67±6years) of the PREDIMED-Valencia study prospectively from 2003 to 2014. Cancer incidence was a secondary outcome in our trial. We analyzed 69 cases of incident cancer (lung, breast and colon) during this follow-up period, and 69 paired controls free of cancer. We analyzed DNA-methylation levels of the CYP1A1 and GSTP1 genes at baseline in both groups. Quantitative DNA methylation analysis was undertaken by MALDI-TOF mass spectrometry. We evaluated methylation levels on chromosome 15 (region: 74726090 -74726460 base pairs from promoter) and chromosome 11 (region: 67583556-67583896 base pairs from promoter).

Results

We detected statistically significant differences in DNA methylation levels at baseline in the CYP1A1 and GSTP1 genes between cancer cases and controls (P=0.008 for the CpG site 26-27 of the CYP1A1 gene and P=0.049 for the CpG 10-11 island at the GSTP1 gene). We detected statistically significant changes in DNA methylation prospectively in cancer patients. DNA methylation at the CpG 4 (CYP1A1 gene) was 0.020±0.034 at baseline vs 0.006±0.013 close to cancer diagnosis (P=0.044). For the GSTP1 gene, methylation of the CpG 34-39 prospectively increased from 0.327±0.046 at baseline to 0.345±0.053 (P=0.014) close to cancer diagnosis.

Conclusions

We have detected differences in DNA-methylation of selected CpG Islands of the CYP1A1 and GSTP1 genes at baseline, between future diagnosed cancer subjects and cancer-free controls. Moreover, in patients subsequently diagnosed with cancer, a change in DNA methylation was observed between baseline and close to the time of cancer diagnosis. This suggests a dynamic influence of DNA methylation that could be modulated for prevention.

Clinical trial identification

Controlled-trials.com number ISRCTN35739639

Legal entity responsible for the study

Spanish Government's Instituto de Salud Carlos III and University of Valencia

Funding

Instituto de Salud Carlos III

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

40P - Regulation of glucose transporters by protein kinases in cancer cells (ID 3699)

Presentation Number
40P
Lecture Time
13:15 - 13:15
Speakers
  • A. Henriques
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Cancer cells require increased glucose supply to sustain proliferation. One mechanism involves increased expression of glucose transporter (GLUT) genes. But insulin has revealed that protein phosphorylation is another key mechanism in glucose uptake regulation: insulin binding to responsive cells triggers a signalling cascade with phosphorylation of TBC1D4, a negative regulator of endosomal GLUT trafficking, so that more transporters are inserted into the plasma membrane. Previous work from the host lab has identified the family of WNK protein kinases and shown that WNK1 can also phosphorylate TBC1D4 and promote GLUT translocation to the cell surface. Our objective is to understand the contribution of WNK1 to glucose uptake in colorectal cancer cells. Our objective is to understand the contribution of WNK1 to glucose uptake in colorectal cancer cells.

Methods

To characterize the role of WNK1, various colorectal cell lines were first cultivated with different glucose concentrations. Levels of GLUT1 at the cell surface were compared under these conditions and the effect of depleting WNK1 expression by siRNA determined.

Results

For selected conditions, key cell cycle or apoptotic marker proteins were analysed by Western blot and revealed higher apoptotic and cell-cycle arrest phenotypes in WNK1-depleted cells cultured in low glucose medium. In order to dissect key phosphorylation events involved in GLUT1 regulation, mass spectrometry analysis revealed that WNK1 phosphorylates TBC1D4 and the functionally related TBC1D1 at unique Serine residues. The corresponding phospho-mimetic mutants are currently being tested for their ability to increase GLUT1 translocation.

Conclusions

Together, these studies will elucidate the molecular details regulating the translocation of glucose transporters in cancer cells and have the potential to identify novel therapeutic targets.

Legal entity responsible for the study

Peter Jordan

Funding

Fundação para a Ciência e Tecnologia BioISI - Biosystems and Integrative Sciences Institute

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

41P - Signal transduction pathways regulating alternative splicing of tumor-related RAC1b (ID 3659)

Presentation Number
41P
Lecture Time
13:15 - 13:15
Speakers
  • V. Gonçalves
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In colon cancer distinct genetic subtypes have been described, one of which involves overexpression of RAC1b, a variant generated by alternative splicing. Aberrant splicing is known to occur in cancer and can be caused by mutation in a gene or splicing factor but also represents a dynamic response to oncogene-induced cellular signaling and in this case it may be pharmacologically targeted. Here we explore how signaling pathways are involved in the deregulation of alternative RAC1b splicing in colorectal tumor cells.

Methods

HT29 colorectal cells represent serrated colorectal tumors with BRAF gene mutation V600E in one allele and RAC1b overexpression. Cells were transfected with shRNA vectors directed against target candidate protein kinase transcripts and their effects on RAC1b levels analyzed 24h later by Western Blot and qRT-PCR. Treatment with kinase inhibitors or anti-inflammatory drugs was performed 24h prior to cell lysis.

Results

Two kinases, SRPK1 and GSK3β, were found required to sustain RAC1b levels and both were shown to act upon the phosphorylation of splicing factor SRSF1, which binds to and promotes the inclusion of the alternative exon in RAC1b. SRPK1 knockdown or pharmacological inhibition reduced SRSF1 phosphorylation decreasing its nuclear translocation and concomitantly RAC1b splicing. The same regulatory pathway was also found to be controlled by GSK3β. Interestingly, GSK3β phosphorylation was identified to serve as target for the anti-inflammatory drug ibuprofen, which inhibits RAC1b overexpression.

Conclusions

Together, our results demonstrate that oncogenic signal transduction pathways deregulate alternative splicing and this may be drug revertable.

Legal entity responsible for the study

Peter Jordan

Funding

Fundação para a Ciência e Tecnologia

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

42P - Preserving tumor heterogeneity: A microfluidic reactor for ex vivo preservation of colorectal cancer biopsies (ID 5157)

Presentation Number
42P
Lecture Time
13:15 - 13:15
Speakers
  • G. Llamazares
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Foreseeing treatment outcome in cancer patients is still a challenge that needs to be addressed. Tumors are complex structures, where the interaction between the tumor cells and the surrounding microenvironment regulates key processes in cancer progression, such as angiogenesis, evasion and modulation of the immune system response, and invasiveness. These interactions confer tumors a high heterogeneity not only inter-patient but also intra-patient. In vitro experimental models have been developed to preserve this heterogeneity present on tumor biopsies by the use of rotary wall and perfused bioreactors. However, the complexity and size of the bioreactors prevent from visual inspection of the sample and the realization of a high-throughput screening. The present work focuses on the combination of microfabrication techniques and microfluidics to downsize classic experimental models. The developed methodology requires only microliter size sample, and allows real time optical inspection.

Methods

A microscopy slide size optically transparent microfluidic bioreactor (µbioreactor) was designed and developed to preserve high cellularity on complex samples through constant perfusion. Colorectal carcinoma (CRC) biopsies were taken after previous patient consent was obtained. CRC biopsies were perfused by cell culture media in the µbioreactor during one week. After perfusion, CRC biopsies were histologically processed, stained and characterized by immunofluorescence.

Results

High cellularity was observed in CRC biopsies after one week of perfusion. Stromal and parenchymal preservation was confirmed by both, histological staining and immunofluorescence.

Conclusions

The use of microfluidic bioreactors can be successfully used to preserve CRC biopsies, maintaining cell heterogeneity while allowing optical inspection. The use of small sample volumes (microliters) allows high throughput screening using regular biopsy samples, a key feature to achieve personalized treatments in cancer.

Legal entity responsible for the study

University of Zaragoza

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

44P - Role of ICAM-1/LFA-1 interaction in the angiogenic and desmoplasic response in liver metastasis (ID 4826)

Presentation Number
44P
Lecture Time
13:15 - 13:15
Speakers
  • A. Alonso
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The colorectal cancer is one of the most common cancers in the world being the main cause of death the metastatic spread to the liver. During metastatic progression, a stromal and tumor cell crosstalk mediated by hepatic ICAM-1 and tumor LFA-1 interaction modulate the tumor microenvironment through an inflammatory and immune response. Additionally, a matrix remodeling and angiogenic response is also associated with tumor progression. The main cell type associated to these processes is the fibroblast associated to the tumor. Thus, the aim of this work is to elucidate the effect of ICAM-1/LFA-1 interaction during the angiogenic and desmoplasic response during liver metastatic progression.

Methods

To do so, the effect of ICAM-1/LFA-1 interaction on the tumor progression and recruitment of cancer associated fibroblasts was analyzed by an experimental metastasis assay in vivo and a modified Boyden chamber migration assays in vitro, after either activation of tumor cells with sICAM-1 or blocking of ICAM-1/FA-1 interaction. In addition, the effects of LFA-1 tumor depletion on tumor migratory potential induced by tumor-activated fibroblasts derived factors were analyzed. Also, the effect of the modulation of this pathway on MMPs protein and angiogenic gene expression levels was measured by zymography and qPCR, respectively.

Results

In vivo and in vitro assays showed an increase on fibroblast and tumor cells recruitment after activation of tumor LFA-1 activation by binding with sICAM-1 which was abrogated after blocking of LFA-1. Moreover, the expression levels of MMPS and other proangiogenic factors were decrease after the blockage of ICAM-1/LFA-1 interaction. Also, the collagen deposition was increased after LFA-1 activation and diminished by LFA-1 blocking.

Conclusions

The interaction of ICAM-1 with tumor LFA-1 favors the recruitment of fibroblast within the tumor mediated by a modulation of pro-desmoplasic factors. This favors the remodeling of the tumor stroma and the angiogenic response and promotes tumor metastatic progression. Thus, LFA-1ICAM-1 interaction might be pointed out as a potential target for therapy of the metastatic disease.

Legal entity responsible for the study

Department of Cellular Biology and Histology, University of the Basque Country, School of Medicine and Nursery

Funding

Basque Government

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

45P - Role of discoidin domain receptors in extracellular matrix remodeling during tumor-host interaction in liver metastasis (ID 4904)

Presentation Number
45P
Lecture Time
13:15 - 13:15
Speakers
  • I. Romayor
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Metastasis is the main cause of death for most solid tumors. The liver is a metastasis-susceptible organ and represents the first most common site for colorectal cancer (CRC). During tumor progression, the unique hepatic microenvironment suffers a reorganization involving the interaction between the tumor, the different hepatic stromal cells and the extracellular matrix (ECM), which is under an extreme remodeling. Among the receptors involved, discoidin domain receptors (DDR-1 and -2), a class of tyrosine kinase receptors for fibrillar collagen, are emerging as new therapeutic targets in cancer treatment, including colorectal. Aim: We aim to elucidate the implication of DDRs in the prometastatic properties of the CRC cells and stromal cells in the liver.

Methods

First, the expression of DDRs on the stromal cells under tumor activated conditions and on tumor cells in the presence of tumor-activated stromal factors was assessed at protein levels. Second, this was related to the migratory potential under the same conditions. Finally, metalloproteinases expression, known to be induced by DDRs activation and involved in cell migration and ECM remodeling, was determined by western blot and zymography.

Results

DDRs expression was inversely altered in macrophages and fibroblasts after their activation by tumor derived factors. The expression of DDRs on CRC cells was decreased by factors derived from stromal cells. These DDRs deregulated expression was related to changes in the migratory capacity of tumor and stromal cells. Moreover, MMP-9 and MMP-14 expression increased in the stromal cells activated by tumor factors, while TIMP-2 expression was higher in fibroblasts but lower in macrophages. Also, the activation of CRC cells by either fibroblasts or macrophages derived factors induced a differential expression of MMP-2, MMP-9 and MMP-14.

Conclusions

The differential expression of DDRs by cells in the tumor microenvironment could redirect the expression of MMPs inducing the migratory capacity of CRC cells. In conclusion, tumor and stromal cells crosstalk may dysregulate the ECM deposition related to DDRs expression contributing to the extensive stroma remodeling in CRC metastatic diseases.

Legal entity responsible for the study

University of the Basque Country (UPV/EHU)

Funding

University of the Basque Country (UPV/EHU)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

46P - Novel role of apatinib as a multi-target RTK inhibitor in the direct suppression of hepatocellular carcinoma cells (ID 4277)

Presentation Number
46P
Lecture Time
13:15 - 13:15
Speakers
  • X. Li
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Although apatinib has been demonstrated with potential antitumor activity to multiple solid tumors, the underlying mechanism of apatinib for the treatment of hepatocellular carcinoma (HCC) remains unclear. In the present study, we explore if there is any direct suppression effect of apatinib on HCC cells and its relevant targets.

Methods

To determine the role of apatinib, we investigated its effect on viability, colony formation, apoptosis, migration of 6 HCC cell lines in vitro, and HCC progression in mice model. Using a phospho-receptor tyrosine kinase pathway array with 49 different tyrosine kinases, we screened and verified the tyrosine kinase targets involved in apatinib therapy.

Results

Apatinib treatment significantly inhibited HCC cell viability, proliferation, colony formation, migration, and enhanced cell apoptosis in a concentration-dependent manner (p <0.05). Furthermore, apatinib showed a favorable anti-tumor growth effect (71% of inhibition ratio, p <0.05) in the established human HCC xenograft mice model with good safety. RTK pathway arrays and western blots analysis demonstrated apatinib significantly down-regulated the phosphorylation levels of several tyrosine kinase receptors, especially PDGFR-α and IGF-IR, and inhibited Akt phosphorylation.

Conclusions

These novel data suggested that the apatinib may have a direct anti-HCC effects as a direct multi-target RTK inhibitor of HCC cells and a promising potentiality in HCC clinical therapies.

Legal entity responsible for the study

Xiaojin Li

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

47P - GATA6 exhibits tumor suppressive effects in hepatocellular carcinoma (ID 1330)

Presentation Number
47P
Lecture Time
13:15 - 13:15
Speakers
  • H. Tan
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

GATA6 is a transcription factor that regulates endoderm differentiation and lineage specification. Dysregulation of GATA6 expression had been reported in cancers of endoderm-derived organs such as the lungs, stomach and pancreas. The aim of this study is to determine the clinical significance of GATA6 in hepatocellular carcinoma (HCC) and characterize its potential functional roles.

Methods

GATA6 mRNA expression was assessed in 74 clinical HCC samples by quantitative polymerase chain reaction (qPCR). Correlation between GATA6 expression and clinicopathological parameters was analyzed. Stable GATA6 knockdown clones were established by a lentiviral-based approach in HCC cell line Huh7, which showed relatively high endogenous GATA6 expression. Functional effects upon GATA6 manipulation were investigated by cell proliferation, migration, invasion and tumorsphere formation assays in vitro.

Results

GATA6 expression was significantly downregulated in HCC tumor tissues when compared with the corresponding non-tumoral liver tissues (p = 0.007). A lower GATA6 expression was correlated with poorer cellular differentiation (p = 0.004). Silencing of GATA6 stimulated HCC cell proliferation, and promoted cell invasive and migratory abilities in vitro. This increase in metastatic capacity was mediated through the activation of epithelial-mesenchymal transition (EMT), as demonstrated by the loss of E-cadherin and gain of vimentin protein expression levels. Suppression of GATA6 augmented the self-renewal ability of HCC cells as demonstrated by the enhanced number and size of tumorspheres formed in both primary and secondary generations. In addition, the expression of various stemness markers such as Nanog, Oct4 and Sox2 were upregulated upon GATA6 silencing.

Conclusions

Our findings suggest that GATA6 is downregulated in HCC which may help to convert HCC cells to a more poorly differentiated state and enhance proliferation, self-renewal ability and metastatic potential.

Legal entity responsible for the study

Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

48P - The effects of 5-fluorouracil (5-FU) on TGF-β-related signaling molecules (ID 636)

Presentation Number
48P
Lecture Time
13:15 - 13:15
Speakers
  • T. Wang
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

By identifying the mechanism of therapeutic effects of the combination of interferon alpha (IFNα)-2b and 5-fluorouracil (5-FU) on advanced hepatocellular carcinoma (HCC) with portal venous invasion1, we recently found that 5-FU increased both the expression and secretion of transforming growth factor (TGF)-β in HepG2 cells. In this study, we analyzed the effects of 5-FU on TGF-β-related signaling molecules.

Methods

Hepatoma cells (HepG2 and HuH7) were treated with 5-FU, IFNα-2b, and the combination of 5-FU and IFNα-2b. The total and/or phosphorylated protein levels of TGF-β-related signaling molecules were detected by western blot analysis. Additionally, the effects of above-mentioned treatments on the epithelial–mesenchymal transition (EMT) of the cells were evaluated by performing invasion and migration assays.

Results

With respect to the TGF-β-induced apoptosis signals, 5-FU inhibited not only the phosphorylation of SMAD2, but also reduced the total protein levels of SMAD2, SMAD4, and pINK4b. Conversely, 5-FU stimulated the phosphorylation of TGF-β-induced EMT molecules, such as ERK and JNK, but not p38MAPK. Accordingly, the protein levels of E-cadherin were reduced in the cells treated with 5-FU. On the other hand, IFNα-2b did not affect the levels of TGF-β-induced EMT molecules, whereas the combination of 5-FU and IFNα-2b neutralized the effects of 5-FU on TGF-β-related signaling molecules and restored their protein levels to those observed in the control. Interestingly, the phosphorylated protein levels of SMAD2 and the total protein levels of E-cadherin and p15INK4b increased in 5-FU-stimulated HUH7 cells, but not in HepG2 cells. Furthermore, 5-FU stimulated both cell invasion and migration in HepG2 cells, whereas the combination of 5-FU and IFNα-2b abolished these effects of 5-FU.

Conclusions

Our data suggest that 5-FU induces the EMT of hepatoma cells through TGF-β, and that the higher efficacy of the combination therapy of 5-FU and IFNα-2b results from the inhibition of these effects of TGF-β. The differences observed between HepG2 and HUH7 cells in response to the stimulation with 5-FU indicate that the efficacy of the therapy may differ between patients with hepatitis B (HBV) or C virus (HCV) background.

Legal entity responsible for the study

Iwate Medical University

Funding

Japan Society for the Promotion of Science (JSPS)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

49P - Gene mutations involved in drug resistance in liver cancer cells using a new rna-seq data analysis workflow (ID 3528)

Presentation Number
49P
Lecture Time
13:15 - 13:15
Speakers
  • E. AKHAN GUZELCAN
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

According to Global Cancer Statistics (GCS) Hepatocellular carcinoma (HCC) is the 5th most common and 2nd deadliest cancer in the world. The incidence of HCC has increased over the past decades but still an effective therapy has not been developed. Sorafenib, which is the only FDA approved agent, can improve the patient survival just for a few months, therefore liver transplantation is the most efficient way of treatment up to date. In this study, we offer potential drug targets by analyzing the relationship between mutation status and drug treatment response of well-differentiated Huh7 and poorly-differentiated Mahlavu liver cancer cells.

Methods

PI3K/Akt pathway is hyperactive in ∼%40 of HCC. We determined the characterized and determined IC50 values of Sorafenib, PI3Ka and b inhibitors and their combinations. RNAseq experiment were performed with the inhibitor treated cells. The RNAseq data of each cancer cell line (as control) was compared to treated samples. Somatic mutations associated with drug resistance were comparatively identified with RNAseq data workflow wrapped in our laboratory using GATK-MuTect tool (Cibulskis, 2013). The results were further filtrated to obtain the missense mutations. The mutated genes that were identified during the chemical knockdown studies were further analyzed in patient survival data. The functional studies were performed by gene silencing.

Results

SLC39A5, FRG1, PPHLN1 and SRP9 gene mutations were found to be shared among all drug resistant cells. Mutated genes were shown to be associated with cancer perseverance and aggressiveness. In addition, we found an unknown transcript called CTC512. The functional studies demonstrated that once these genes were silenced, the cellular growth was prevented. Gene silencing showed the alteration of the cell cycle progression of drug resistant cells. The affected downstream pathways were further analyzed by western blotting.

Conclusions

Our results indicate potential target genes which are critical to be addressed due to their roles in resistance to drugs in HCC. Once the mutated genes are silenced the cancer progression is prevented. The identified genes can be considered as chemotherapeutical and disease progress targets.

Legal entity responsible for the study

Rengul Cetin Atalay, METU

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

50P - Camouflaging iRGD-EGFR anchored human cytotoxic T-lymphocyte membranes to the surface of nanoparticles combined with low-dose irradiation: New approach to enhance drug-delivery targeting in gastric cancer (ID 2578)

Presentation Number
50P
Lecture Time
13:15 - 13:15
Speakers
  • L. Zhang
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

We report a biomimetic delivery platform based on human cytotoxic T-lymphocyte membranes. In this platform, T-lymphocyte membranes were camouflaged to the surface of poly-lactic-co-glycolic acid nanoparticles, with local low-dose irradiation (LDI) as a chemoattractant. These carriers were further anchored with the recombinant protein anti-EGFR-iRGD, improving tumor accumulation, facilitating tumor uptake.

Methods

The T-lymphocyte membrane coating was verified by dynamic light scattering, transmission electron microscopy and confocal laser scanning microscopy. The particle phagocytosis study was performed using a human phagocytic cell line. In vivo NIR fluorescence imaging was performed to monitor the route of nanoparticles. EGFR expression of tumor cells was tested before and after LDI.

Results

This new platform reduced phagocytosis of macrophages by 23.99% (p = 0.002). iRGD-EGFR anchored T-lymphocyte membrane-encapsulated nanoparticles accumulated in tumor site more than unfunctionalized groups, while LDI significantly enhanced the targeting ability. LDI could up-regulate EGFR expression on tumor cells, which was important in the process of localization of iRGD-EGFR anchored T-lymphocyte membrane-encapsulated nanoparticles in tumors.

Conclusions

This new platform included both the long circulation time and tumor sites accumulation ability while LDI could significantly enhance the tumor accumulation ability.

Legal entity responsible for the study

National Natural Science Foundation of China

Funding

National Natural Science Foundation of China

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

51P - Delivery of paclitaxel-loaded erythrocytes-based nanoparticles using injectable albumin hydrogel for regional chemotherapy (ID 3604)

Presentation Number
51P
Lecture Time
13:15 - 13:15
Speakers
  • H. Qian
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Peritoneal dissemination often occurs in advanced gastric cancer (GC) patient. However, systemic chemotherapy alone has limited effect on peritoneal metastases. The purpose of this work is to fabricate a regional nanomedicine delivery system with injectable hydrogel, to investigate the sustained drug release, biocompatibility, degradation, and the therapeutic efficacy on advanced GC.

Methods

The injectable hydrogel gelling at body temperature was synthesized by one-step esterification of albumin and polyethylene glycol. The paclitaxel-loaded nanoparticles (PRNP) were prepared by encapsulating the drug in erythrocytes membrane ghost and embedded into the hydrogel (PRNP-Gel). The physical and chemical performances were characterized by dynamic light scattering, electronic microscope and SDS PAGE. The drug loading content, hemocompatibility, degradation, drug release, drug accumulation at tumor site, and anti tumor efficacy was also investigated.

Results

The PRNP-Gel suspension gelated in 15 min after subcutaneous injection. The diameter of PRNP in hydrogel was 133.1±1.6 nm, drug loading efficacy and content were 85.45±1.32% and 22.10±0.25%, respectively. 37.9% of the loaded paclitaxel was released in 196 h in vitro, demonstrating the sustained release properties of PRNP-Gel. No hemolysis was detected within the concentration up to 10 mg/mL, and the PRNP-Gel degraded completely in 200 days after injection. The IC50 of PRNP against MKN45 gastric cancer cells, determined by MTT, was 15.16 ng/mL. In vivo antitumor evaluation found that, the tumor growth inhibition of MKN45 on tumor bearing mice was 64.5% of PRNP-Gel group, which was higher than the PRNP (40.0%, P < 0.05) and Taxol (33.8%, P < 0.05). The average tumor weight was 74.9±40.1 mg, while they were 194.6±90.9 mg and 199.6±73.9 mg in PRNP and Taxol respectively (P < 0.05).

Conclusions

The biocompatible and degradable drug delivery system could release chemotherapeutics in a long-term and sustained manner, exhibited an enhanced drug accumulation at tumor site, resulting in the superior antitumor effect in vitro and in vivo. This work provided a new strategy of therapy for advanced GC.

Legal entity responsible for the study

Hanqing Qian

Funding

National Nature Science Foundation of China

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

52P - Effect of apatinib combined with 5-fluorouracil (5-FU) on proliferation, apoptosis and invasiveness of gastric cancer cells (ID 2339)

Presentation Number
52P
Lecture Time
13:15 - 13:15
Speakers
  • P. ZHAO
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

To investigate the effect of apatinib, a small-molecule tyrosine kinase inhibitor, combined with 5-FU on proliferation, apoptosis and invasiveness of human gastric cancer cells AGS, and provide experimental basis for the treatment of two drugs combination in gastric cancer in clinic.

Methods

The expression of vascular endothelial growth factor receptor 2 (VEGFR2) protein in human umbilical vein endothelial cells (HUVEC) and human gastric cancer cells were assessed by western blotting. 4-methyl-teerazolium (MTT) assay and flow cytometry were used to assess the cytotoxicity and apoptosis effects of the cells in response to control, single apatinib, single 5-FU, and apatinib combined 5-FU groups. Western blotting was used to evaluate the expression of p-Akt, proliferating cell nuclear antigen (PCNA), Caspase-3 and the invasiveness differences of the four groups were detected by wound healing assay and matrix metalloprotein-2 (MMP-2), E-cadherin gene amplification were measured by RT-PCR.

Results

AGS had the expression of VEGFR2. Compared with single drug groups, apatinib combined with 5-FU could significantly suppress the growth, proliferation and induce apoptosis of human gastric cancer cells in time and dose-dependent manners (P<0.05). Western blotting dispalyed p-Akt and PCNA expression decreased after AGS cells treated with apatinb combined 5-FU. Wound-healing assay showed the invasiveness of AGS cells was inhibited and probably through down-regulating MMP-2 and E-cadherin amplification in combined group (P<0.05).

Conclusions

Our study points that apatinib combined 5-FU could inhibit the proliferation of AGS gastric cancer cells by down-regulating the expression of p-Akt. The invasiveness of AGS cancer cell was inhibited by reduced expression of MMP-2 and E-cadherin genes, and provides a theory basis for 5-FU and apatinib combination in clinic with advanced gastric cancer patients who failed to second-line treatment but still had a good performance status.

Legal entity responsible for the study

Bangwei Cao

Funding

National Nature Science Foundation of China

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

53P - One tumour, two clones: An in vitro model of intra-tumour heterogeneity (ID 5223)

Presentation Number
53P
Lecture Time
13:15 - 13:15
Speakers
  • A. Salawu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Eradication of advanced disease remains elusive in the majority of cancers including soft tissue sarcomas (STS) despite advances in our understanding of the molecular mechanisms that drive them. Targeted treatment development to date has largely relied upon data derived from all cells within a tumour sample and/or tumour cell lines. These approaches however, do not account for inherent heterogeneity of cancer cells within a single tumour and is considered an important factor that leads to treatment failure. Understanding intra-tumour heterogeneity is therefore a priority for cancer research and appropriate tumour models with sufficient availability would greatly facilitate the identification of newer targets and factors that lead to treatment resistance. We therefore aimed to develop in vitro models of STS that reflect intra-tumour heterogeneity.

Methods

We obtained tissue from patients having surgery for STS in Sheffield Teaching Hospitals and established primary tissue cultures. Short Tandem Repeat (STR) confirmed the same origin of both clones in both cases. DNA copy number profiling and gene expression microarray analysis were used for molecular characterisation of self-immortalised primary cell lines.

Results

One leiomyosarcoma (Shef-LMS 01) and one myxofibrosarcoma cell line (Shef-MFS 01) established two morphologically-distinct tumour cell types (culture variants) in separate long term cultures. Karyotyping and growth characteristics confirm that both variants in each case are tumour cells and they have remained in culture for over 100 passages. STR profiling confirms that in each case, both clones are derived from the same tumour. DNA copy number analysis with microarray-based comparative genomic hybridisation and gene expressson analysis shows many identical somatic copy number abnormalities (SCNA) between variants, but also numerous genomic and transcriptomic differences.

Conclusions

We believe that these genomic and transcriptomic differences provide clues to clonal evolution in these tumours and may explain the development of resistance to targeted treatment. These cell lines are therefore useful for the identification of novel targets and development of effective therapies for these tumours.

Legal entity responsible for the study

Abdulazeez Salawu

Funding

Weston Park Cancer Charity and Sarcoma UK

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

54P - Docosahexaenoic acid mediates susceptible cell death through differential regulation of p62/p-eIF2alpha/NRF2 in LMP1-expressing nasopnaryngeal carcinoma cells (ID 832)

Presentation Number
54P
Lecture Time
13:15 - 13:15
Speakers
  • K. Lim
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Docosahexaenoic acid (DHA) induces apoptotic cell death through several mechanisms in cancer cells. We have previously demonstrated that DHA triggers apoptosis by increasing reactive oxygen species (ROS) accumulation and the ROS-mediated apoptosis caused by DHA is associated with Nrf2 signaling activation. Here we report that DHA-induced cell death is more susceptible through p62/p-eIF2alpha/NRF2 regulation in LMP-1-expressing nasopharygeal carcinoma (NPC) cells.

Methods

Viability of CNE-LMP1 and HONE-EBV cells was compared with CNE and HONE after DHA treatment by MTT assay. DHA-induced apoptosis was analyzed using the TUNEL assay and Western blot of cleaved form of PARP. Tissue expression of LMP-1 and p62 were observed by immunohistochemistry.

Results

Treatment of four human NPC cells (CNE, CNE-LMP1, HONE, HONE-EBV) with DHA for 24 hr resulted in a dose-dependent inhibition of cell growth. The DHA effect was due to the induction of apoptosis, given that DHA increased the cleaved form of PARP as well as the number of TUNEL-positive cells. The inhibition of CNE-LMP1 and HONE-EBV cells after DHA treatment is more susceptible. compared with CNE and HONE cells without LMP1 by MTT assay. The level of p62 and NRF2 of LMP1-NPC cells were increased after DHA pretreatment cpmpared to control NPC cells. On the other hand, the level of p-eIF2alpha produced reverse result. The activation of Nrf2 signal seems to result from decreased Nrf2 inhibitor, Kelch-like ECH-associated protein 1 (Keap1), because DHA remarkably attenuated Keap1 expression levels. Moreover, silencing Nrf2 by small interfering RNAs inhibited the cytotoxic effect of DHA, indicating that Nrf2 activation plays a positive role in the process of DHA-induced apoptosis. Increased staining for LMP1 and p62 was observed in NPC tissues when compared with the nonneplastic (chronic inflammation) tissues.

Conclusions

These results suggest that differential regulation of p62/p-eIF2alpha/NRF2 contributes to susceptible cell death by DHA in LMP-1-expressing NPC cells. Thus, utilization of DHA may represent a promising therapeutic approach for chemoprevention and treatment of human NPC.

Legal entity responsible for the study

Chungnam National University

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MEST) (2007-0054932, NRF-2015R1D1A1A01056887) and by the framework of international cooperation program This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MEST) (2007-0054932, NRF-2015R1D1A1A01056887) and by the framework of international cooperation program managed by National Research Foundation of Korea (2015K2A2A6002008)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

55P - Inhibition of the ubiquitin-conjugating enzyme E2B restores the BCNU sensitivity of cancer cells by regulating MGMT ubiquitination (ID 4310)

Presentation Number
55P
Lecture Time
13:15 - 13:15
Speakers
  • S. Hsu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

O6-Methylguanine-DNA methyltransferase (MGMT) is a DNA repair enzyme that removes the mutagenic O6-alkyl groups from guanines. 1, 3-Bis (2-chloroethyl)-1-nitrosourea (BCNU), a DNA damage reagent, is known to induce cell death of tumors and the ubiquitin dependent proteolysis of MGMT. The present study aims to enhance BCNU cytotoxicity toward cancer cells by modulating MGMT dynamics.

Methods

Human nasopharygeal carcinoma cells, including HONE-1 and TW01, and human colon carcinoma HT-29 cells were used for the BCNU treatments, siRNA knockdown, immunoprecipitation and western blot experiments. The BCNU cytotoxicity was determined using methylene blue assay. Proteins involved in MGMT ubiquitination were confirmed with immunofloresence staining and in vitro protein ubiquitination assays.

Results

We previously identified ubiquitin-conjugating enzyme E2B (UBE2B), a DNA repair enzyme with ubiquitin-conjugating abilities, as a critical regulator of the cell cycle in oral cancer cells. A novel role of UBE2B was further revealed in regulating MGMT dynamics in nasopharygeal carcinoma cells and colon carcinoma cells. Increased colocalization of UBE2B with MGMT was found in BCNU treated cancer cells. Depletion of MGMT or UBE2B in cancer cells resulted in decreased IC50 for BCNU. Lower MGMT expression levels were observed in UBE2B deficient cells. Overexpression of MGMT rescued the UBE2B-depleted cells from the cytotoxic concentrations of BCNU, suggesting that MGMT is a downstream target of UBE2B. The E3 ubiquitin ligase RAD18, that is known as a partner of UBE2B in facilitating PCNA ubiquitination, was analyzed to investigate the mechanism of the UBE2B regulation on MGMT. Interaction of RAD18 and MGMT was observed in cancer cells, and was enhanced under the BCNU treatments. Our results also showed that UBE2B and RAD18 contribute to MGMT ubiquitination under in vitro conditions.

Conclusions

Our study indicated that the UBE2B-RAD18 regulation on MGMT plays an important role in BCNU-induced cancer cell death. Thus, UBE2B inhibition may be considered as a potential strategy for cancer treatment. (This work was supported by Taiwan Ministry of Science and Technology under the grants no. NSC 103-2320-B006-036-MY3).

Legal entity responsible for the study

National Health Research Institutes, Tainan, Taiwan

Funding

Taiwan Ministry of Science and Technology

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

56P - Oral squamous cell carcinoma cells were sensitized to cetuximab by Eribulin via induction of the mesenchymal-to-epithelial transition (ID 2525)

Presentation Number
56P
Lecture Time
13:15 - 13:15
Speakers
  • H. Kitahara
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Inhibition of EGFR signalling has emerged as a new treatment strategy for oral squamous cell carcinoma (OSCC). Previously, we found that loss of EGFR expression in OSCC was associated with EMT, and might have functional implications with regard to resistance to cetuximab, a monoclonal anti-EGFR antibody. Eribulin (a microtubule inhibitor) reportedly renders breast cancer less aggressive, and less likely to metastasise, by triggering the mesenchymal-to-epithelial (MET) transition. Here, we evaluated whether eribulin-induced MET was associated with re-sensitization of resistant OSCC cell lines to cetuximab.

Methods

In vitro antiproliferative activities were determined in three human OSCC lines (OSC-20, OSC-19 and HOC313) treated with eribulin. These three human OSCC represented different EMT/MET states.

Results

Interestingly, HOC313 cells (mesenchymal phenotype) were highly sensitive to eribulin in comparison with other cell lines, and significantly enhanced the anti-proliferative effect of cetuximab in response to the drug. Eribulin also underwent a MET-associated gene switch that resulted in high EGFR expression in HOC313 cells, and abrogated a TGF-β-induced EMT gene expression signature.

Conclusions

Eribulin-dependent sensitization of OSCC to cetuximab is likely due to induction of MET. Combination therapies based on eribulin and cetuximab have potential as a novel treatment regimen in OSCC.

Legal entity responsible for the study

Kanazawa University

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

57P - In vivo study of the vaccine adjuvants prothymosin alpha and prothymosin alpha(100-109) in melanoma (ID 4204)

Presentation Number
57P
Lecture Time
13:15 - 13:15
Speakers
  • P. Samara
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The TLR agonist prothymosin α (proTα) pleiotropically stimulates immune cells via generating the appropriate cytokine milieu for their activation. The C-terminal decapeptide proTα(100-109) is considered the immunologically active moiety of proTα, as it restores in vitro the deficient immune responses of cancer patients equally well to proTα. ProTα and proTα(100-109) ligate TLR-4, signal through the TRIF/MyD88-dependent pathways, and promote the maturation of dendritic cells. The latter further stimulate TH1-type immune responses and prime tumor antigen-reactive T cell functions. We evaluated whether proTα and proTα(100-109) function correspondingly in vivo.

Methods

C57BL/6 mice were subcutaneously inoculated with the syngeneic melanoma B16.F1 cells. Upon palpable tumor formation, mice were intraperitonealy treated with 2 cycles of GM-CSF, proTα(100-109) or proTα, in conjunction with a B16.F1-specific peptide vaccine. Tumor growth and animal survival were monitored. Splenocytes from selected animals were tested for ex vivo cytotoxicity by 51Cr-release assay and CD107 expression. Excised tumors were analyzed by immunohistochemistry, while serum cytokines were quantified by flow cytometry.

Results

Both peptides therapeutically administered in melanoma-bearing mice in the presence of cancer antigens, retarded tumor growth and prolonged the survival of treated animals by 25 days. Ex vivo analysis of spleen cell cytotoxicity confirmed the in vivo induction of B16.F1-specific and non-specific anti-tumor responses. Tumors of mice treated with proTα/proΤα(100-109) did not show infiltration of smooth muscle fibers and vessels, produced less melanin, presented limited necrotic areas and were characterized by sparse T cell infiltration. Sera from the same animals contained more IFN-γ, whereas the concentration of IL-4 was marginally increased.

Conclusions

Our results show that proTα and proTα(100-109) induce TH1-biased immune responses in vivo. As both peptides are non-toxic to normal cells, their ability to orchestrate and modulate the desired anti-tumor immune responses in mice, suggests their eventual exploitation as adjuvants in anti-cancer peptide vaccines in humans.

Legal entity responsible for the study

O. Tsitsilonis

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

58P - Melanoma affects clock gene machinery of several organs in tumor-bearing mice (ID 1460)

Presentation Number
58P
Lecture Time
13:15 - 13:15
Speakers
  • L. De Assis
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The accumulation of soluble factors in the tumor microenvironment and their release into the bloodstream lead to systemic effects, resulting in cancer-associated syndromes such as cachexia. Another association observed in the latter scenario is chrono-disruption, which has been related with tumor onset and development. In fact, a positive relationship between master clock integrity and healthiness has already been disclosed; however, it is unclear whether the effects of chronodisruption in cancer are locally or systemically exerted. It is known that lung adenocarcinoma reprograms liver metabolism via a pro-inflammatory response without affecting liver clock genes. Our study evaluated whether a non-metastatic skin tumor can affect clock gene machinery of other organs.

Methods

Eight to sixteen-week old C57BL/6J male mice were inoculated subcutaneously with B16-F10 cells (or PBS, control animals), single housed at 22 ± 2 °C, kept under 12/12h light/dark cycle, and received food and water ad libitum. Two weeks after inoculation mice were euthanized 2 hours after lights on (ZT2) or 2 hours after lights off (ZT14). Skin of control animals and samples of non-tumoral adjacent skin, tumor, liver, lung, and brown adipose tissue (BAT) were collected to evaluate the expression of clock genes (Per1, Per2, Bmal1, and Nr1d1) by qPCR.

Results

No oscillatory profile of clock genes was detected in skin of control animals, tumor adjacent skin, and tumor itself of inoculated animals; Bmal1 expression was reduced in adjacent skin and tumor as compared to skin of control animals. In liver and lung tissue, Per1 and Per2 oscillated in control animals, and tumor inoculation did not affect this oscillatory profile. Temporal oscillation of Bmal1 and Nr1d1 in the liver was lost in tumor-bearing mice. In BAT, Per1 and Bmal1 oscillatory expression was also lost in tumor-bearing mice. In all organs analyzed. Bmal1 transcript was reduced in tumor-bearing mice when compared to control animals.

Conclusions

The presence of a non-metastatic tumor in the skin alters clock machinery in adjacent skin, liver, lungs, and BAT. These data bring new knowledge of how tumor macro-environment affects clock machinery, resulting in a likely chrono-disruption of the organism as a result of a localized tumor in the skin.

Legal entity responsible for the study

University of Sao Paulo (USP) and Sao Paulo Research Foundation (FAPESP)

Funding

Sao Paulo Research Foundation (FAPESP) and National Council of Technological and Scientific Development (CNPq).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

59P - Is there receptor tyrosine kinases expression on lymphocytes in patients with renal cell carcinoma? First-in-human study (ID 3178)

Presentation Number
59P
Lecture Time
13:15 - 13:15
Speakers
  • D. Khochenkov
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

To date little is known about receptor tyrosine kinases (RTK) expression on peripheral blood mononuclear cells (PBMC) and tumor infiltrating lymphocytes (TIL) in cancer patients. The aim of this study was to evaluate expression levels of major RTK: VEGFR1, VEGFR2, PDGFRα, PDGFRβ, FGFR2 in CD45+ population of PBMC and TIL isolated from patients with clear cell renal cell carcinoma (RCC).

Methods

Tumor and blood samples were obtained from 20 patients with RCC immediately after surgical resection of primary tumor. Tumors were harvested into sterile antibiotic-containing RPMI 1640 medium (Gibco). TIL isolation was performed under modified protocol [Baldan et al., 2015]. Isolated TIL and PBMC were prepared for flow cytometry. Cells were double stained with anti-CD45 FITC-conjugated mouse antibody, and with PE-conjugated mouse antibodies to VEGFR1, VEGFR2, PDGFRa, PDGFRb, FGFR2 (all Sony Biotech) and were analyzed on NovoCyte 2000R flow cytometer (ACEA Biosciences). Expression of RTK was evaluated with NovoExpress Software.

Results

Among PBMC 72.1±21.3% cells were CD45-positive. Isolated TIL contain 19.2±5.6% CD45-positive cells. PBMC and TIL express RTK. Expression levels of RTK are summarized in the table.

59P

Expression of RTKPBMCTILP
VEGFR125.6±11.4%31.0±27.2%0,699
VEGFR223.8±11.1%53.2±29.3%0,096
PDGFRa48.0±18.9%45.4±16.7%0,833
PDGFRb63.3±28.7%66.7±26.1%0,843
FGFR241.2±27.8%23.6±12.3%0,168

Conclusions

To our knowledge, this is first study that showed significant RTK expression on peripheral and RCC-infiltrating lymphocytes in RCC patients. PBMC and TIL have similar RTK expression levels.

Legal entity responsible for the study

Ethical committee, KCRB

Funding

Kidney Cancer Research Bureau

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

60P - Understanding and targeting Met signalling in bladder cancer (ID 4761)

Presentation Number
60P
Lecture Time
13:15 - 13:15
Speakers
  • W. Badreldin
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Bladder cancer affects 430,000 patients and leads to 165,000 deaths annually worldwide. With no major advances in the management of this disease in the last 2 decades, there is an urgent need to identify therapeutic targets with validated biomarkers. Overexpression of Met, a Receptor Tyrosine Kinase, was shown to correlate with poor prognosis in bladder cancer making it an attractive target. Cabozantinib, a Met inhibitor, showed clinical activity in patients with refractory bladder cancer in a clinical trial. However, little is known about how Met exactly signals in bladder cancer and there are no validated biomarkers. This study aims at unravelling Met signalling in bladder cancer.

Methods

Western blots and confocal/low light microscopy were used to assess Met signalling and its role in wound healing in Transitional Cell Carcinoma (TCC) cells. Met expression was assessed by immunohistochemistry in tissue samples (n = 64).

Results

Met is overexpressed in TCC cells and in 78% of invasive bladder cancer tissues. This was associated with a shorter median survival as compared to Low Met levels (12.97 Vs 18.05 months). Stimulation of TCC cells with Met ligand, Hepatocyte Growth Factor (HGF), triggered Met activation and downstream signalling as well as wound healing, all of which were reduced with Met pharmacological inhibitors including Cabozantinib. The PI3K downstream effector AKT was highly activated upon Met activation. Moreover, class I PI3K inhibition with GDC094 significantly inhibited HGF-dependent wound healing. Interestingly, HGF triggered rapid Met endocytosis in TCC cells. Furthermore, pharmacological inhibition of endocytosis reduced Met downstream signalling.

Conclusions

We report that Met is a major target in invasive bladder cancer. Our results further suggest that PI3K may be considered as a co-target of Met to improve patients’ outcome. It may also be developed as a biomarker to help select patients who may respond to Met targeted therapy. Finally, we report for the first time that, upon HGF stimulation, Met gets rapidly endocytosed in TCC cells. Furthermore, inhibiting endocytosis reduced Met dependent signalling. All together, our results open the way for novel strategies to target invasive bladder cancer.

Legal entity responsible for the study

Queen Mary University of London

Funding

Barts Cancer Institute, Queen Mary University of London

Disclosure

T. Powles: Honoraria: AstraZeneca, Bristol-Myers Squibb, Roche and Merck. Research Funding: AstraZeneca, Roche and Merck.

rn

L. Menard: Employee: AstraZeneca.

rn

All other authors have declared no conflicts of interest.

Disclosure

T. Powles: Honoraria: AstraZeneca, Bristol-Myers Squibb, Roche and Merck. Research Funding: AstraZeneca, Roche and Merck.

\r\n

L. Menard: Employee: AstraZeneca.

\r\n

All other authors have declared no conflicts of interest.

Disclosure

T. Powles: Honoraria: AstraZeneca, Bristol-Myers Squibb, Roche and Merck. Research Funding: AstraZeneca, Roche and Merck.

L. Menard: Employee: AstraZeneca.

All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

61P - Integrated molecular signatures of TERT promoter deregulation predict disease outcomes in non-muscle invasive bladder cancer (ID 5440)

Presentation Number
61P
Lecture Time
13:15 - 13:15
Speakers
  • R. Leão
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Recent studies showed increased interest in telomere biology bladder cancer. These studies were mainly focused on hotspot TERT promoter mutations and their contribution for telomerase activation and clinical outcomes. However, the study of TERT promoter methylation, as an additional deregulatory mechanism of TERT expression, was not studied in this disease. We previously uncovered a region in the TERT promoter region (THOR – TERT hypermethylated oncological region) which is specifically hypermethylated and associated with telomerase activation in cancer tissue. In this study we aim to establish the value of this duet (TERT promoter mutations and THORMeth) in bladder cancer recurrence and progression.

Methods

To explore the impact of TERTpMut and THORMeth on TERT expression and clinical outcomes in UBC we studied two cohorts of UBC patients, 331 FFPE samples. THORMeth status was assessed using quantitative bisulfite pyrosequencing. Sanger Sequencing and ddPCR accessed TERTpMut status. TERT expression was evaluated by ddPCR. Cox proportional hazards models were used to correlate THORMeth and TERTpMut with disease recurrence and progression.

Results

THOR is a diagnostic marker for urothelial bladder cancer. THOR hypermethylated (n = 127, 53.6%) is associated with higher levels TERT expression (p < 0.0001) and is a risk factor for disease recurrence in NMIBC (Log rank p = 0.034) and is crucial for disease progression. TERT promoter mutations are present in all stages and grades (n = 182, 76.8%) and are a risk factor for disease recurrence amongst NMIBC (HR: 3.8, p < 0.0001). Genetic and epigenetic combined signatures in the TERT promoter allow for a clinical bladder cancer classification system based on underlying telomere biology.

Conclusions

THOR is a novel biomarker for UBC, and, as TERTpMut is able to predict disease recurrence in NMIBC. TERTpMut/highTHORMeth, comprises a distinct signature that is associated with disease progression and higher TERT expression. This fact suggests a hypothetical synergism between both mechanisms and highlights the merit of evaluating TERT promoter methylation as other deregulatory mechanism in TERT expression with implications in patients’ outcomes.

Funding

Foundation for Science and Technology, Portugal, Individual Doctoral Grant SFRH/BD/102232/2014.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

62P - Dose- and regimen-dependent effects of dexamethasone on extracellular matrix of brain tissue (ID 1490)

Presentation Number
62P
Lecture Time
13:15 - 13:15
Speakers
  • A. Tsidulko
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Dexamethasone (DXM) is commonly used in the management of glioma patients to treat intracranial edema but patients often suffer from its side effects. The molecular mechanisms of these side effects are poorly studied. DXM seems to affect extracellular matrix (ECM) especially proteoglycans (PGs) known to be a major component of the ECM in brain tissue. The aim of our study was to investigate if the effects of DXM on brain tissue PGs depend on the treatment regimen or DXM dose.

Methods

Effects of different doses and regimens of DXM treatment on the brain ECM were studied using RT-PCR and IHC in the ex vivo model of organotypic brain tissue culture and in vivo experimental animal model. The ex vivo organotypic culture model was chosen instead of in vitro cell culture model as it represents the real 3D structure of the tissue and can be used to study ECM.

Results

The most expressed PGs in rat brain tissue were syndecan-1, glypican-1, decorin, biglycan and lumican. DXM treatment of organotypic hippocampus culture ex vivo led to dose-dependent suppression of brevican, perlecan and biglycan expression and increase in expression of glypican-1, NG2 and versican. In the in vivo experiments, PGs demonstrated age-specific and brain zone-specific expression patterns in normal brain of Wistar rats. The effects of DXM on cortex and hippocampus of the experimental animals were dose- and regimen-dependent. Low-dose DXM treatment led to significant decrease in expression of most PGs in cortex but 3-fold increase in syndecan-1, perlecan and brevican expression in hippocampus. Treatment with high-dose DXM resulted in 2-6-fold increase in most of PGs expression in both brain zones. Long-term treatment led to the most dramatic changes in PGs expression on both mRNA and protein levels, completely changing their expression pattern.

Conclusions

Taken together, obtained data demonstrate an importance of DXM doses/regimens during antiglioma therapy. Long-term treatment and high doses of DXM lead to the most dramatic alteration of PGs composition in brain ECM creating a favorable niche for tumor growth and relapses.

Legal entity responsible for the study

Institute of Molecular Biology and Biophysics

Funding

Russian Science Foundation (RSF grant 16-15-10243).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

63P - Clinical dysregulation of DNA repair by the polynucleotide kinase/phosphatase-XRCC4-DNA ligase IV in neurological disease (ID 4697)

Presentation Number
63P
Lecture Time
13:15 - 13:15
Speakers
  • D. Aceytuno
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

If not repaired, DNA double-strand breaks (DSBs) can lead to genomic instability and cell death or neoplastic transformation. The major DSB repair (DSBR) mechanism in higher eukaryotes is non-homologous end-joining (NHEJ). In NHEJ, polynucleotide kinase/phosphatase (PNKP) is the primary enzyme for processing abnormal 5'-OH and 3'-phosphate ends that prevent the final repair step by XRCC4/DNA Ligase IV (Lig IV). This processing step is thought to be mediated by an interaction between the PNKP-FHA domain and CK2-phosphorylated XRCC4 C-terminal tails.

Methods

See below.

Results

Our binding assays show tight binding between XRCC4/Lig IV and PNKP both with and without CK2-phosphorylation of XRCC4. Low-resolution ensemble structures of purified phosphorylated-XRCC4/Lig IV/PNKP ternary complex by small-angle X-ray scattering (SAXS) experiments also suggest a second phosphorylation-independent interaction between the PNKP and XRCC4/Lig IV. Hydrogen-deuterium exchange (HDX) experiments have identified a candidate for this secondary interaction site within a loop in the PNKP phosphatase domain. This site contains the clinically significant PNKP E326K mutation found in the severe form of the hereditary neurological disease MCSZ (microcephaly with early-onset intractable seizures and developmental delay). Activity assays show that the E326K mutation decreases both substrate binding and turnover in PNKP when bound to phosphorylated-XRCC4/Lig IV. Furthermore, UV laser microirradiation in cells show that the E326K mutation also disrupts recruitment of PNKP to DNA lesions.

Conclusions

We have identified a putative secondary interaction site that functionally contributes both to recruitment and catalysis of PNKP in NHEJ. Disruptions to PNKP in this region may result in decreased DNA double-strand break repair in cells and describe a molecular basis of MCSZ. Further, PNKP has other known clinical neurological significance and its presence on chromosome arm 19q has interesting implications in oligodendrogliomas. This interaction surface may prove an interesting target for small-molecule inhibition of DNA strand break repair toward novel radio- and chemo-sensitizing therapies in cancer treatment.

Legal entity responsible for the study

University of Alberta

Funding

Canadian Institutes of Health Research, Alberta Cancer Foundation, Structural Biology of DNA Repair Machines Consortium.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

64P - Effects of rottlerin and genistein through EF2K on proliferation, invasion and cell cycle/death in neuroblastoma cells (ID 5133)

Presentation Number
64P
Lecture Time
13:15 - 13:15
Speakers
  • M. Erdogan
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Neuroblastoma (NB) is the most common extracranial solid cancer in childhood and the most common cancer in infancy in the world. Rottlerin, a naturally occurring polyphenolic compound derived from Mallotus philipinensis, appears to have great potential in cancer therapy because of its effects on proliferation and apoptosis. Genistein is a phytoestrogen and it has been found to inhibit uncontrolled cell growth in several cancers. Recently, we learned that eukaryotic elongation factor-2 kinase (EF2K) is dramatically upregulated in many cancer cells and promotes cell survival and proliferation. Rottlerin and genistein have also showed inhibitory effects on this kinase in other solid tumours like pancreatic cancer. With this is mind, we investigated the effects of rottlerin and genistein in neuroblastoma cells.

Methods

In this study, two human neuroblastoma cancer cell lines (SH-SY5Y and Kelly) were treated with rottlerin and genistein in vitro. Cell proliferation, colony formation and invasion were assessed, and wound-healing tests, western blots (wb), cell cycle and apoptosis analysis by flow cytometry were performed.

Results

Our results showed that rottlerin and genistein treatments caused a significant reduction in cell proliferation, colony formation, and invasion/wound-healing capacity in neuroblastoma cells at concentrations of 5 µM and 30 µM, respectively (p < 0,0001). The combination of these doses also increased the level of inhibition in these analyses (p < 0,0001). Additionally, these drugs also increased the level of apoptosis and caused G1 cell cycle arrest in neuroblastoma cell lines (p < 0,0001). We showed that these treatments markedly inhibit EF2K overexpression. Our wb data suggested that EF2K may enhance tumorigenesis/metastasis through the upregulation of pro-tumorigenic/metastatic pathways in these cells and these agents may produce their anti-proliferative, anti-metastatic and apoptotic effects through EF2K downregulation.

Conclusions

In conclusion, these results indicate that rottlerin and genistein have important effects on neroblastoma cell behaviour and these effects may be caused by downregulation of EF2K.

Legal entity responsible for the study

Mumin Alper Erdogan

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

65P - Influence of emotiogenic brain structures on tumor growth in the experiment (ID 2421)

Presentation Number
65P
Lecture Time
13:15 - 13:15
Speakers
  • E. Korobeynikova
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Stress is the pathogenetic basis of many diseases. It leads to decreasing resistance of the organism, including antitumor one. Emotional stress of a cancer patient affects the quality of life and treatment outcomes by decreasing the antitumor resistance. Therefore, correction of emotional state is an urgent task. The purpose of the study was to reveal the influence of stimulation of emotiogenic brain structures on the antitumor resistance of animals with cancer.

Methods

Transplantable solid sarcoma S45 in white outbred male rats weighing 230-250 g was used as a model of tumor growth. Implantation of bipolar stimulating electrodes in the subcortical structures of the brain was performed aseptically by stereotactic coordinates. Electrodes were implanted to nucleus lateralis septi (LS) – “positive emotiogenic structure” and to Globus pallium (GP) – “negative emotiogenic structure”; the data were compared to the values in animals without electrodes (controls). The stimulation was performed daily for 2 months without changing the stimulation current.

Results

Stimulation of emotiogenic brain structures influenced the dynamics of S45 growth, and LS stimulation caused the greatest inhibition of tumor growth.

65P

StructureLSGPControls
n91010
Initial tumor V (cm3)3.0±0.53.1±1.92.2±1.2
Final tumor V (cm3)4.7±1.17.5±2.26.8±1.6
Tumor increase (%)53.3±2.8186±6.5211±6.8
Effectiveness index1.40.9
Inhibition of tumor growth (%)30.810.2

Morphological study of the thymus, spleen and lymph nodes of animals with LS showed signs of high functional activity of the organs providing a high level of resistance.

Conclusions

Electrostimulation of LS influences antitumor resistance, significantly improving it. This suggests the expediency of combining specific anticancer drugs and nonspecific effects of psychotropic drugs - anxiolytics in complex treatment of cancer.

Legal entity responsible for the study

Rostov Research Institute of Oncology

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

66P - Systematic evaluation of the immune microenvironment of neuroendocrine tumours (NET) (ID 5240)

Presentation Number
66P
Lecture Time
13:15 - 13:15
Speakers
  • C. Vesely
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Immunotherapy is currently being explored in many tumour types with encouraging results, but has not yet been evaluated in neuroendocrine tumours (NET). Our aim is to characterise the immune landscape of NET and determine which immune-modulatory pathways control the tumour infiltrating lymphocytes (TILs) in order to develop a rational approach for immunotherapy in this tumour type.

Methods

Peripheral blood and fresh tissue was obtained from consenting patients with NET, and subjected to multicolour flow-cytometry to determine the abundance of CD8+, CD4+FoxP3- effector (CD4eff) and CD4+FoxP3+ regulatory (Treg) T cell subsets and the expression of co-inhibitory and co-stimulatory checkpoint molecules on these subsets. Additionally, matched FFPE tissue was obtained for multiparametric immunohistochemistry to investigate the distribution of the immune infiltrate.

Results

Tissue from 23 NET patients including 19 mid-gut tumours (13: G1 and 6: G2) was analysed. Overall the tumours contained a higher proportion of Tregs compared with peripheral blood (8.5% vs 5%, P = 0.02) and had a CD8:Treg ratio of 18.1:24.3 respectively (P = 0.036). The co-inhibitory molecules CTLA-4 and TIM-3 showed highest expression on Tregs, while PD-1 and LAG-3 expression was similar across all T cell subsets. Co-stimulatory molecules, including ICOS, 41BB and OX-40, were also highest on Tregs, as was the recently identified co-stimulatory receptor TIGIT. Immunohistochemistry revealed that the majority of cases have <1% intratumoural CD4+ and CD8+ T cells but a higher number of peritumoural T cells from all subsets. Where present, T cells were predominantly CD8+ and intratumoural CD163+ macrophages were also identified.

Conclusions

These preliminary results provide novel insight into the immune landscape of NET, and may inform the development of targeted combination immunotherapies. Initial results suggest that checkpoint molecules, such as PD1 and LAG-3, may be potential targets in this tumour type and work is ongoing to further elucidate the immunogenic potential of NET.

Legal entity responsible for the study

University College London

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

67P - Developing a prediction model for response to lenalidomide treatment in refractory/relapsed multiple myeloma patients (ID 1641)

Presentation Number
67P
Lecture Time
13:15 - 13:15
Speakers
  • Y. Chung
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Despite improvements in treatment for Multiple myeloma (MM) achieved by novel drugs such as proteasome inhibitors (PIs) and immunomodulatory drugs (IMiDs), most patients will ultimately relapse or become refractory to their current treatment. Therefore, it is important to understand the mechanisms of therapeutic resistance in relapsed/refractory MM (RRMM) for improving treatment outcome. Recently, it was reported that serum or plasma of MM patients showed sufficiently stable miRNA signatures with prognostic impacts in MM cohorts, which can be used as minimally invasive markers for predicting and monitoring treatment outcomes. However, the expression patterns and biological implications of miRNAs are still unclear in RRMM patients receiving lenalidomide with dexamethasone (Len-dex).

Methods

We investigated the expression of serum miRNAs by genomewide miRNA array analysis and explored their predictive values in RRMM patients receiving Len-dex.

Results

We explored the associations of miRNAs with treatment outcome of Len-dex treatment and prognosis in 55 RRMMs (25 good responders and 30 poor responders) and built a prediction model for treatment response. Three miRNAs (miR-29c-3p, miR-30c-5p, and miR-331-3p) were found to be significantly down-regulated in poor responders. In survival analysis, lower expression of the three miRNAs was significantly associated with shorter time to progression (TTP) or poorer overall survival (OS). Eight clinical factors were also associated with TTP or OS. By combining the miRNA markers and clinical markers, we designed a a prediction model for response to lenalidomide treatment in RRMM patients. Our model showed better prediction power (AUC=0.855, sensitivity 84%, specificity 76%, and accuracy 81%) than international staging system (ISS) based prediction.

Conclusions

Our results suggest the potential of circulating miRNAs as minimally invasive markers for treatment response and prognosis in RRMM patients.

Legal entity responsible for the study

This study was approved by the Institutional Review Board of The Catholic University of Korea and conducted in accordance with the Declaration of Helsinki.

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

68P - Combinatorial inhibition of mTOR and exportin-1 (XPO1) represses cell survival via metabolic modulation of pro-survival pathways in mantle cell lymphomas (ID 2884)

Presentation Number
68P
Lecture Time
13:15 - 13:15
Speakers
  • K. Sekihara
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

MCL is an aggressive B-cell lymphoma with aberrant expression of several oncogenic effectors and requiring novel anticancer strategies. The nuclear transporter exportin-1 (XPO1) is highly expressed in MCL and is critical for cancer survival and proliferation. mTOR signaling is frequently activated and an important therapeutic target in MCL. In this study, we investigated the antitumor effects and molecular/metabolic changes induced by combined with dual mTOR inhibitor AZD-2014 and XPO1 inhibitor KPT-185 on MCL cells under the hypothesis that mTOR inhibition by AZD-2014 represses KPT-185 inducedupregulation of glycolysis.

Methods

Four MCL cell lines (Jeko-1, X138, JVM-2, and MINO), primary MCL cells, and normal bone marrow samples were utilized. Cell viability was evaluated by MTT assay. Cell cycle and apoptosis were determined by flow cytometric analysis. cDNA array, iTRAQ proteomic, immunoblotting and metabolome analysis using CE-TOF-MS were also performed.

Results

AZD-2014 enhanced KPT-185-induced the inhibition of cell growth and repression of cell viability in MCL cells but not in normal bone marrow cells. Different mTOR inhibitors (AZD-8055 and MLN0128) demonstrated similar effects. AZD-2014+KPT-185 decreased expression of the oncogenic mediator c-Myc andthe translational/transcriptional network regulator HSF1as detected by immunoblotting. iTRAQ proteomic analysis demonstrated thatthe combination caused repression of ribosomal biogenesis. Treatment with either AZD-2014 or KPT-185 depressed phospho-S6.CET-OF-MS metabolite assay showed that AZD-2014+KPT-185inhibited theKrebs cycle, and that AZD-2014 effectively repressed KPT-185-induced upregulation of glycolysis. cDNA array detected downregulation of NOD2which is known to trigger activation of MAP kinases and of NF-kappa-B signaling. Moreover, AZD-2014+KPT-185 activated AMPK, an energy stress marker in a cell type-dependent manner.

Conclusions

Our findings indicated that the combinatorial inhibition of mTOR and XPO1 identifies a novel synthetic lethality mechanism that could be exploited clinically, following satisfactory completion of pre-clinical in vivo studies.

Legal entity responsible for the study

Yoko Tabe

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

69P - CD200 and indoleamine 2,3-dioxygenase-1 (IDO-1) overexpresion in relapsed acute myeloid leukemia patients (ID 979)

Presentation Number
69P
Lecture Time
13:15 - 13:15
Speakers
  • A. Memarian
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Immunosuppression is one of the major causes of AML pathogenesis and progression. CD200 and IDO are immunoregulatory factors which overexpressed in some solid tumors and hematological malignancies. Distinct researches have shown CD200 and IDO expression is associated with AML progression. In the current study, we simultaneously examined the expression of these molecules, as the two important factors including in immunosuppression, in the newly diagnosed and relapse AML patients to investigate their correlation with each other.

Methods

48 PBMC samples of newly diagnosed and relapse AML patients were tested and also 32 normal subjects were employed as control. CD200 expression level was examined on cells by Flow cytometry and quantitative real time RT-PCR was used to determine the IDO-1 gene expression. Finally, data were analyzed statistically.

Results

Data showed that CD200 and IDO-1 overexpressed especially in relapsed patients. Comparison between FAB AML subgroups demonstrated no statistical differences in the case of CD200 level but expression of IDO-1 was slightly increased only in M4 subgroup in comparison to M3. Correlation analyses showed strong association between expression of CD200 and IDO-1 in all patients particularly in relapsed AML, whereas no significant correlation was found in normal subjects.

Conclusions

According to the role and overexpression of CD200 and IDO-1 in AML patients and also their two-way correlation with T-regs in disease induction and progression, simultaneous assessment of these parameters are so valuable for more exact prognosis detection. Also inhibition of all these immunoregulatory pathways could be so useful for immunotherapy outcome, especially in relapsed AML.

Legal entity responsible for the study

Tehran University of Medical Sciences

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

70P - Correlation between types of acute lymphoblastic leukemia with socio demographic factors (ID 4443)

Presentation Number
70P
Lecture Time
13:15 - 13:15
Speakers
  • M. Sen
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Acute lymphocytic leukemia (ALL) is the most predominant hematopoietic clonal disorder in children than adult. ALL classifies into two subtypes: B –ALLand T-ALL. The incidence of ALL subtype in urban areas is generally higher than rural areas. In the Western World, the predominant immunophenotype observed in ALL is B-ALL with 60-80% of total case, and T-ALL is only 15-20%. But in case of developing country like India the reverse is true. The objective of the present study is to examine and correlate T-ALL and B-ALL in leukemia patients with respect to socio-demographic factors.

Methods

During May 2015 - April 2017, total 427 ALL patients (male:female::1.9:1), age between 2-60 years attended OPD and IPD of Netaji Subhas Chandra Bose Cancer Research Institute, Kolkata, India. We have collected peripheral blood and/or bone marrow samples for immunophenotyping by FACS after taking written consent from the patients. Each sample is evaluated with a panel of monoclonal antibodies and compared the immunophenotyping data with socio-demographic factors (age, sex, economic and social status etc).

Results

The overall survival of ALL patients (with mean age 13.6 years) in 2 year is 73.6%. In our hospital, the economically weak patients (77.05%) are more abundant than economically sound patients (22.95%). Out of 427 patients, T-ALL (51.28%) is predominantly higher than B-ALL (48.71%). We found that immunophenotyping data is correlated with all the socio demographic data i.e., sex, economic and social status etc. Though disease free survival and event free survival is markedly higher in B-ALL compared to T-ALL, but we found the survival of T-ALL is also increasing.

Conclusions

Our unique findings emphasize that the detection of T-ALL and B-ALL by immunophenotypic analysis for better treatment and outcome of the patients and also trying to correlate the prevalence of T-ALL and B-ALL with economical status and also with other socio-demographic factors in study area.

Legal entity responsible for the study

Netaji Subhas Chandra Bose Cancer Research Institute

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

71P - CALR mutations and their link with cellular calcium during megakaryocyte hyperplasia in MPNs (ID 4585)

Presentation Number
71P
Lecture Time
13:15 - 13:15
Speakers
  • M. Morlan Mairal
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Megakaryocyte hyperplasia is a major characteristic of two myeloproliferative neoplasms (MPNs) known as essential thrombocythemia (ET) and Primary myelofibrosis (PMF). About 35% of ET and PMF patients harbour somatic calreticulin (CALR) mutations. CALR is a calcium (Ca2+) buffering protein within the endoplasmic reticulum (ER). Ca2+ is an important element for megakaryocyte functions; however the impact of CALR mutations in cellular Ca2+ during megakaryocyte hyperplasia remains elusive.

Methods

I-TASSER software was used to study the aminoacid charge using the 3D structure of CALR mutant. Marimo cells and overexpressing CALR mutant HEK293T and DAMI cells were used as cellular models. CALR cellular localization was addressed by flow cytometry and confocal microscopy. Basal Ca2+ levels were measured by Fluo-8 staining. Furthermore cells were treated with Fendiline and BTP-2 calcium channel blockers to manipulate cellular Ca2+.

Results

The present study shows that CALR mutations change the aminoacid charge of the Ca2+ binding region of CALR and that mutant CALR is localized outside the ER, within the cytoplasm and the cellular membrane. These results suggest that CALR mutations could be affecting the Ca2+ buffering activity within the ER. Therefore, we further analysed Ca2+ basal levels in CALR mutant cells, and our results showed that CALR mutations show lower cellular Ca2+levels. These results lead us to think that low intracellular Ca2+ levels could be the driving force of megakaryocyte hyperplasia characteristic of ET and PMF. Therefore, we induced low intracellular Ca2+ levels in leukemic blast by using Ca2+ channel blockers and our results showed that treated cells display an increase of the megakaryocyte marker CD41a in the cell surface, suggesting an induction of megakaryopoiesis in these cells.

Conclusions

These findings elucidate that low intracellular Ca2+ caused by CALR mutations could be the driving force of megakaryocyte formation in ET and PMF. This study shows the relevance to understand the role of cellular calcium during megakaryocyte formation and this could unravel the pathogeny of CALR mutant in MPNs.

Legal entity responsible for the study

University of Salford

Funding

University of Salford

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

72P - Approach based on magnetic nanocomplexes improves antitumor efficacy of dendritic cells immunotherapy in mice (ID 5507)

Presentation Number
72P
Lecture Time
13:15 - 13:15
Speakers
  • M. Inomistova
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Dendritic cell (DC)-based immunotherapy represents a promising approach for cancer treatment. However, the DC homing rate to the lymphoid tissues is poor, thus hindering the activation of antigen-specific T cells and reducing their antitumor efficacy. Here, we developed an approach based on magnetic nanoparticles (NP) to manipulate DC migration and thus elicit a more robust and efficacious antitumor response in tumor-bearing mice.

Methods

Mouse spleen DCs were loaded with nanocomplexes (NC) consisting of iron oxide NP (8x10−12 g/cell) and lyophilized tumor tissue. To investigate the presence of NP in DCs, the method of Fe2+ and Fe3+ detection by Prussian Blue Staining was used. DCs were injected intradermally into tumor-bearing mice three times in an amount of 2x105 per mouse at an interval of 3 days starting from day 7 after Lewis lung carcinoma inoculation. One group of mice that received DCs loaded with NC was exposed to a magnetic field for 1 h. The number and volume of metastases and tumor weight were assessed 28 days after tumor inoculation. At the same time, the levels of INF-γ, IL-10, TGF-ß, IL-4, FoxP3 mRNA expression in the spleen and inguinal lymph nodes were determined.

Results

The iron oxide NP showed no toxic effects on the DCs and had no effect on their viability. We found that almost all DCs are able to incorporate magnetic NP after 24h of incubation. Fewest metastases were found in the mice that received DCs loaded with NC and were exposed to a magnetic field: the number of metastases in mice from this group was 1.7 times less than in control mice. It should be noted that the volume of metastatic nodes in the lungs and the mass of the primary tumor were practically the same as in the control mice. The most pronounced decrease in FoxP3 mRNA levels in the lymph nodes, indicating a decrease in the activity of regulatory T cells, was also noted in the mice receiving DCs loaded with nanocomplexes and exposed to a magnetic field. In the mice of this group, a significant decrease in the level of IL-4 in the spleen was detected.

Conclusions

Our results suggest that an approach based on magnetic NC could be a promising strategy for improving the antitumor efficacy of DC-based immunotherapy.

Legal entity responsible for the study

National Cancer Institute of Ministry of Public Health of Ukraine

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

75P - Signs of tumor-specific immune processes in the regression of large rat tumors under the influence of low-intensity EMR EHF and magnetite nanoparticles (ID 4781)

Presentation Number
75P
Lecture Time
13:15 - 13:15
Speakers
  • G. Zhukova
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The problem of elaboration of methods for effective mobilization of immune antitumor processes remains urgent. Earlier, it was shown that regression of experimental animal tumors can be achieved under the influence of low-intensive electromagnetic radiations (EMR) and magnetite nanoparticles (NPs) (Garkavi L.H. et al, 1996; 2013).The aim of the study was to determine the possibility of regression of large transplanted tumors of white rats under the influence of low-intensity EMR of millimeter range (EHF) and magnetite NPs.

Methods

In experiments on 123 white outbred male rats (200-300g) with transplanted Pliss lymphosarcoma - low-intensity EMR EHF (42.2 GHz, 10 mW/cm2, exposure 15-30 min, low-frequency modulation) and magnetite NPs (10 ± 2 nm) in the form of the magnetic fluid AM-01 (“AM-Cube”, Ekaterinburg) were used. The EMR acted on the animal's head starting 3 days before the tumor was transplanted. NPs were injected into peritumoral zone 2-3 times a week in a single dose of 17.7 mg/kg to animals with already formed tumors. Duration of treatment was 4 weeks. We studied the dynamics of tumor size, histochemical and cytometric changes in tumor tissue. The Wilcoxon test was used to evaluate the results.

Results

When EMR EHF was used, complete regression of tumors with a size of 5-6 cm3 and a partial regression (by 30-40%) of tumors with a size of 10-13 cm3 were noted. The effect was obtained in 33% of the animals. In cases of using of magnetite NPs, tumor regression was observed in 40% of the animals, complete regression of tumors with a size of 5-30 cm3 was observed. Before the regression began, the tumor growth rates did not differ from those in the control group when using as one as the other factor. In addition, the regression was characterized by a rapid rate (5-7 days) and no signs of intoxication in animals. In the peritumoral area considerable macrophage activity and increasing number of cytotoxic T-lymphocytes were noticed.

Conclusions

The timing of the onset of regression, its dynamics and the absence of signs of intoxication during rapid elimination of large tumors indicated the deployment of antigen-presentation processes and specific killing of tumor cells by inducing apoptosis.

Legal entity responsible for the study

Rostov Research Institute of Oncology, Ministry of Public Healthcare of the Russian Federation

Funding

Rostov Research Institute of Oncology

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

76P - Stimulation of RAC1/PAK1 signalling upregulates DNA damage repair genes via the BCL6/STAT5-switch (ID 5288)

Presentation Number
76P
Lecture Time
13:15 - 13:15
Speakers
  • P. Barros
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Colorectal cancer is one of the most prevalent types of cancer worldwide. The GTPase RAC1 and its effector PAK1 have been found overexpressed or hyperactivated in this type of cancers, particularly those with more aggressive and invasive features, which is frequently correlated with resistance to chemotherapeutics and unfavourable clinical prognosis. Previously, we described a new signalling pathway in which activation of RAC1/PAK1 signalling promotes a transcriptional switch between the BCL6 repressor and the STAT5 transcriptional activator at a restricted subset of gene promoters.

Methods

Here we used a novel combinatory ChIP-Seq approach for the genome-wide identification of the BCL6/STAT5-switch target genes.

Results

Ontological enrichment analysis among the identified target genes revealed an overrepresentation of genes involved in DNA damage repair. Using the comet assay as readout for the extent of DNA damage, we show that the activation of RAC1/PAK1 signalling significantly accelerates DNA damage repair through the upregulation of pivotal genes.

Conclusions

This work highlights an additional role for the RAC1/PAK1 signalling axis that may contribute to the chemoresistant phenotype of aggressive colorectal tumours.

Legal entity responsible for the study

Paulo Matos

Funding

Fundação para a Ciência e Tecnologia.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

77P - Deciphering the regulation of the metastasis suppressor, NDRG1 in different cancer-types and its functional implications (ID 4635)

Presentation Number
77P
Lecture Time
13:15 - 13:15
Speakers
  • K. Park
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The metastasis suppressor, N-myc Downstream Regulated Gene-1 (NDRG1) inhibits metastasis in a variety of cancer-types, including cancers of the breast, colon, pancreas and prostate. Its potent anti-oncogenic effects were demonstrated in multiple in vitro and in vivo studies, making it a promising therapeutic target. However, exactly how NDRG1 is regulated in different cancer-types and how different regulatory mechanisms affect NDRG1 function remain to be elucidated. Notably, post-translational modifications (PTMs), phosphorylation and cleavage of NDRG1, have been associated with its function. Therefore, it was crucial to examine whether these PTMs occur universally or selectively in different cancer-types. Further, considering the DNA repair role suggested for nuclear NDRG1, the effects of the above PTMs on nuclear NDRG1 levels was examined.

Methods

DU145 and PC3 prostate cancer cells, PANC-1 pancreatic cancer cells, HT-29 colon cancer cells, HepG2 and Hep3B hepatocellular carcinoma (HCC) cells were utilised. Full-length (FL) and truncated (T) NDRG1 isoforms were detected using a C-terminus directed antibody. The FL isoform was detected using an N-terminus directed antibody. Ser330 or Thr346 phosphorylation (p-NDRG1) was detected using specific antibodies.

Results

For the first time, we demonstrated that phosphorylation and potential cleavage of the NDRG1 protein occurs in all the various cancer cell-types examined. Although the levels varied, both the FL and T NDRG1 and its phosphorylated form were detected in all tumour cells assessed. The FL NDRG1 isoform was predominantly found in the cell nucleus. Ser330 p-NDRG1 was also highly localised in the cell nucleus, while Thr346 p-NDRG1 was mostly cytoplasmic. These cellular distribution patterns were similar in all cancer-types tested.

Conclusions

This study demonstrates for the first time that the NDRG1 protein is phosphorylated and potentially cleaved in diverse cancer cell-types. Further, FL NDRG1 and Ser330 p-NDRG1 were highly localised to the cell nucleus. These results indicate that the N-terminus region and phosphorylation at Ser330 could be crucial for nuclear expression and the well-known anti-metastatic function of NDRG1.

Legal entity responsible for the study

The University of Sydney

Funding

The University of Sydney, Cancer Institute New South Wales National Health and Medical Research Council, Cancer Australia, Cure Cancer Australia Foundation.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

78P - Registration-based automated lesion detection and therapy evaluation of tumors in whole body PET-MR images (ID 4575)

Presentation Number
78P
Lecture Time
13:15 - 13:15
Speakers
  • H. Ahlström
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Integrated PET/MR scanners can simultaneously acquire whole body functional PET data together with morphological and functional MR data. Whole-body PET-MRI datasets contain huge amounts of spatially detailed morphological, functional and metabolic information. We propose a method, based on deformable registration to a whole-body atlas, for computer aided detection of lesions in image data from an integrated PET-MRI system.

Methods

Images were acquired using an integrated 3T PET-MRI system (Signa PET/MR, GE Healthcare). Fat and water MR images were collected using a Dixon MR Attenuation Correction (MRAC) sequence (TE 1.67ms, TR 4.05ms, voxel size: 2x2x5.2 mm). Subjects underwent a PET scan after injection of [F18]-FDG (2 MBq/kg) with 3 minutes per bed, with a 100-120 minute interval between injection and scan start. PET reconstruction was performed using GE’s fully 3D Time-of-Flight iterative reconstruction (2 iterations, 28 subsets, standard 5 mm filter, voxel size 3.125x3.125x2.78). Deformable image registration was used to spatially align subjects to a previously created morpphological and functional whole-body imaging atlas (Ekström et al., ISMRM 17), to allow voxel-wise comparisons between the imaged subjects and the atlas. Each voxel in the atlas contains mean and standard deviation of the PET uptake. Utilizing the knowledge that low ADC-values (low diffusion measured by MRI) and high FDG uptake (high metabolism measured by PET) is indicative of malignancy, suspected lesions can be detected by measuring how much the FDG uptake in each voxel deviates from “normality”, as defined by the atlas. This approach generates a voxel-wise “lesion probability map” for the imaged subject. The same registration approach can be used to quantify longitudinal changes in detected lesions, for treatment evaluation.

Results

Lesion probability maps have been generated for patients with manually identified lesions, correctly assigning high values to the regions manually identified as suspected lesions.

Conclusions

The proposed method is promising for lesion detection in whole body PET-MRI images. Future work includes quantitative verification of the accuracy of the detected regions, comparing against manual detection by a radiologist.

Legal entity responsible for the study

Uppsala University

Funding

Antaros Medical

Disclosure

H. Ahlström, J. Kullberg: Co-founder and owner of Antaros Medical.

rn

All other authors have declared no conflicts of interest.

Disclosure

H. Ahlström, J. Kullberg: Co-founder and owner of Antaros Medical.

\r\n

All other authors have declared no conflicts of interest.

Disclosure

H. Ahlström, J. Kullberg: Co-founder and owner of Antaros Medical.

All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

79P - 1,3,5 s-triazine containing analogues a prime Src family inhibitor: Design synthesis docking, anticarcinoma and angiogenic inhibition efficacy on cancer grafted CAM (ID 3078)

Presentation Number
79P
Lecture Time
13:15 - 13:15
Speakers
  • P. Pathak
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The importance of angiogenesis for solid tumor growth is well recognized and evident by the vast differential of research devoted to the subject for over thirty years. It is a complex process directed by growth factors, receptors, extracellular matrix (ECM)-to-cell and cellto- cell interactions. Tyrosine kinase (TKs) is the protein enzymes catalyze the transfer of a phosphate group from an ATP molecule to a tyrosine residue of the target protein, thus leading to signal transduction. Cytoplasmic TKs such as Src, Abl and Lck have been to date discovered and characterized and found that inhibition signalling pathway. A special Src protein like FAK is a non-receptor protein-tyrosine kinase which have vital role in various cellular function like cytoskeleton reorganization, migration, adhesion, spreading, configuration and destruction of FA, cell protrusions, progression, proliferation and apoptosis. The functional ateration of Src signal may be the reason for cancer and metastasis. So this can predict that Src and their signal inhibition can utilized in cancer therapy. Triazine containing hybrid analogues act as a prime skeleton to inhibit Src family TKs like FAK so in present project we constructed 1,3,5-triazine containing analogues (TCA) as an effective cancer induced angiogensis inhibitor.

Methods

TCA analogues constructed accordingly similarity field positioning and pattern through forge V10. Further more in-silico simulation was done using autodock for most prominent analogues. The analogues derived via multifactorial synthetic protocol. The activity evaluation proceeded via in-vitro assay against MCF-7 (Breast cancer) cell line and further in-ova antiangiogenic potency evaluated against cancer induced chick chorioallantoic membrane.

Results

The newly designed and constructed TCA heterocycles expressed more than 56% of similar field point pattern and intra atomic alignments. The prominent pattern showed by the analogue 8d (chloro-anilino) and 8k (bromo-anilino). In-silico docking on hydrophobic site of Src family protein (PDB: 4BRX) revealed that analogue 8d have binding with CYS502, TR503, GLU506, ILE428, ASN551 while analogue 8k shoed interaction with THR503, GLU506, ILE428, LEU567, ASN551 amino acid residue which was similar like vendatinib. Biological evaluation showed that analogues 8d and 8k have great tendency to inhibit cancer induced angiogenesis with marginal toxicity profile.

Conclusions

We have developed a significant series of anticancer analogues and propound the site of binding on the surface of Src family TKs receptor FAK.

Legal entity responsible for the study

N/A

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

81P - A new isoquinoline alkaloid bersavine as a possible anticancer agent (ID 3457)

Presentation Number
81P
Lecture Time
13:15 - 13:15
Speakers
  • K. Habartová
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Plants have had crucial role in the folklore of ancient cultures for over 5000 years. In addition to the use as food or spices, plants have also been utilized as medicine. Two remaining living traditions, the traditional Indian and Eastern medicine, have contributed most to the current state of knowledge related to medicinal plants. In their folklore, herbal medicines were prepared e.g. as teas or tinctures. These products are used as complementary treatment alongside conventional drugs till today. Another trend begun in the early 19th cent., which involved the isolation of active compounds from plants. This tendency led to the discovery of the analgesic alkaloids morphine and codeine or the cardiac glycoside digitoxin. Recently, a new alkaloid bersavine was isolated from Berberis vulgaris, along berbamine and berberine.

Methods

The dried root bark from Berberis vulgaris was minced and extracted with EtOH. The extract was evaporated, dissolved in HCl and extracted with nonpolar solvent. After the second extraction column chromatography on Al2O3 was performed. Subsequently was executed preparative TLC, which reveal a yet unknown alkaloid, later identified by MS and NMR analysis and named bersavine. Effect of bersavine on viability and proliferation was evaluated by WST assay and Trypan blue staining. Next was analysed its impact on cell cycle and apoptosis using the flow cytometry, activity of caspases and Western blot analysis of regulatory proteins was implemented.

Results

Bersavine's effect on cancer cells was first evaluated on panel of 9 different cell lines. Cancer cell lines MOLT-4, Jurkat, HT-29, HeLa and MCF-7 appear to be the most sensitive to the effect of bersavine. Follow-up experiments revealed that bersavine reduced cell viability and proliferation in a dose dependent manner within 24 h of treatment. Moreover, the reduction of cell viability was even more pronounce 48 h following the treatment. The decrease in cell viability was caused by the induction of apoptosis and activation of caspases.

Conclusions

All acquired results suggest that bersavine has very promising activity and it would be worthwhile to subject it to further evaluation.

Legal entity responsible for the study

Charles University, Faculty of Medicine in Hradec Kralove

Funding

Grant Agency of Charles University (Project No. 932616) and SVV-260397/2017.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

82P - Tactics of surgical treatment of tumors of the sacrum (ID 4797)

Presentation Number
82P
Lecture Time
13:15 - 13:15
Speakers
  • I. Khujanazarov
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Tumor lesions of the sacrum are relatively rare and account for 1-7% of all spinal tumors. Tumors of this localization are usually detected when the tumor reaches a significant size and causes gross neurologic disorders and impaired pelvic organs. Radicality of removal of tumors of the sacrum depends on the involvement of the cauda equina, pelvic organs and vascular structures in the process.

Methods

The study involved 13 pateintd TMA clinic on the basis of the Department of Traumatology, Orthopedics, Neurosurgery with GPH No2 from 2011 to 2016. There were 6 women, 7 men. Age category ranged from 17 to 50 years. 1-stage: Holographic selective angiography by Seldinger's method of small pelvic vessels with subsequent embolization of “feeding” tumor of vessels. In addition, the anatomical features of the arteries are determined taking into account the localization of the tumor process, which is important in the operation. 2-stage: After preoperative embolization, three patients underwent hemisakrumectomy with VS3-VS5, and three patients underwent VS1-VS3 sarcomectomy. In this case, patients were additionally stabilized by TPF systems by Lumbo-Pelviofixation.

Results

In 2 cases during surgery, the tumor was intimately soldered to the roots of the horse tail and their isolation led to traumatization of the horse's tail, resulting in a postoperative delay in urine and stool. These violations were resolved within 2 months. In 4 patients with neurinoma S1, S2, S3 spine, an involuntary resection of these roots was performed. In these cases complications from the pelvic organs were not observed due to the presence of a cross innervation. In 2 patients, because of the duration of the operation, suppuration of the operating wound was noted with subsequent secondary healing. There were no lethal outcomes among 13 patients during follow-up.

Conclusions

The tactics of surgical treatment of sacral tumors, including the preliminary embolization of “feeding” arteries with subsequent radical removal of volume formation, reduces the risk of intraoperative complications, and also allows to remove the tumor totally, which in turn prevents its recurrence.

Legal entity responsible for the study

Traumatology Department

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

83P - The impact of co-culture of NSCLC tumor cells and fibroblasts on drug response (ID 1090)

Presentation Number
83P
Lecture Time
13:15 - 13:15
Speakers
  • S. Abreu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The role of stromal cells and the tumor microenvironment has been described to modulate cancer development and tumor drug sensitivity, in part due to the interaction with fibroblasts. Therefore, it is critical to incorporate this feature in our in vitro model and to evaluate its potential impact in early stages of drug development.

Methods

Non Small Cell Lung Cancer (NSCLC) tumor cell aggregates were microencapsulated in alginate capsules, alone or in combination with fibroblasts (immortalized normal and cancer-associated fibroblasts – NFs and CAFs, and human dermal fibroblasts - hDFs), cultured during four weeks; and tumor growth and drug response, both in vitro and in vivo, were evaluated.

Results

Microencapsulation of H1650 and H1437 spheroids in mono- or co-cultures with fibroblasts resulted in viable cultures with tumor aggregate increasing continuously during culture time. However, tumor growth in in vitro co-cultures was dependent on the source of fibroblast and cell line used. When challenged with drugs, co-cultures with fibroblasts in our in vitro 3D model presented, in general, lower sensitivity to therapy after 3 weeks of treatment. H1437+hDFs co-cultures showed less sensitivity to volasertib treatment, with higher DNA concentration (2-fold higher versus mono-cultures) and higher resazurin reduction activity (35% versus 22% in mono-cultures). H1650+NFs co-cultures also demonstrated lower sensitivity to erlotinib and docetaxel treatment, with higher resazurin reduction activity (71% versus 29% in mono-cultures) and higher viable area of aggregates, respectively. Mono and co-cultures can also be injected in mice for the generation of xenografts. Evaluation of tumor growth based on the local of injection, fibroblast source and drug response was compared. In agreement with the in vitro results, only co-culture of H1437+hDFs injected in the lungs significantly enhanced in vivo tumor growth. However, co-culture of H1650 with fibroblasts did not result in altered tumor growth in vivo.

Conclusions

Altogether, we established a 3D model with co-culture of NSCLC tumor cell aggregates and fibroblasts that, depending on the pair used, presented reduced sensitivity to standard of care drugs, better reflecting the clinical observations.

Clinical trial identification

N/A

Legal entity responsible for the study

iBET/ITQB-UNL; AbbVie and Oncotest

Funding

Innovative Medicines Initiative Joint Undertaking (IMI grant agreement n° 115188), resources composed of financial contribution from EU – FP7 and EFPIA companies in kind contribution. iNOVA4Health – UID/Multi/04462/2013, a program financially supported by Fundação para a Ciência e Tecnologia/Ministério da Educação e Ciência, through national funds and co-funded by FEDER under the PT2020 Partnership Agreement is also acknowledged. SA and MFE are recipients of PhD fellowships from FCT (PD/BD/105768/2014 and SFRH/BD/52208/2013, respectively).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

BIOMARKERS (ID 5672)

Lecture Time
13:15 - 13:15
Speakers
  • S. Singh
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15
Poster display session Poster Display session

88P - PALB2 reversion mutations in breast, prostate, and ovarian carcinomas (ID 4874)

Presentation Number
88P
Lecture Time
13:15 - 13:15
Speakers
  • L. Gay
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Like BRCA1/2, deleterious mutations in PALB2 underlie deficiencies in homologous recombination-based DNA repair (HRD) and can underlie sensitivity to platinum (Pt) therapies and PARP inhibitors (PARPi). BRCA1/2 reversion mutations have been widely reported to cause therapy resistance. We report analogous reversion mutations in PALB2 for breast, ovarian, and prostate carcinomas (Ca).

Methods

Comprehensive genomic profiling (CGP) of DNA (≥50 ng) extracted from 114,200 samples, both solid tumors and heme malignancies, was performed using hybridization-captured, adaptor ligation-based libraries (mean coverage depth >600X) for up to 315 cancer-related genes. Samples were evaluated for substitutions, indels, copy number changes and rearrangements.

Results

PALB2 mutations were found in ∼1-3% of breast, ovarian and prostate carcinomas. Of 744 samples (0.7% of total) with likely deleterious PALB2 mutations, 7 (0.9%) harbored multiple alterations with at least one predicted to be a reversion. Of these samples, 4 were breast Ca, 1 a prostate acinar adenocarcinoma, 1 a high-grade ovarian serous Ca, and 1 an ovarian carcinosarcoma. For 3 samples the availability of multiple tests indicated the acquisition of the predicted reversion mutation(s) over time. Several reversion mutation types were observed: missense (1), compensatory frameshifts (3), overlapping small indels (2), and a deletion likely to disrupt splicing (1) that may lead to skipping of exon 4 and an in-frame deletion excising a frameshift mutation. Multiple reversion mutations were observed in 2 cases. Only 1/7 cases also showed disruption of BRCA1/2.

Conclusions

In addition to the commonly sequenced genes BRCA1/2, PALB2 mutation can lead to homologous recombination deficiency. As with BRCA, the acquisition of additional mutations predicted to restore at least some PALB2 function and thus potentially confer resistance to therapies dependent on HRD, can be observed in tumors such as breast, ovarian, and prostate carcinomas. CGP is a valuable tool to identify clinically significant, albeit rare, primary PALB2 mutations in BRCA-negative tumors as well as acquired secondary resistance mutations in patients who progress on Pt and PARP inhibitor based therapies.

Legal entity responsible for the study

Foundation Medicine

Funding

Foundation Medicine

Disclosure

L.M. Gay, S. Daniel, J. Suh, S. Ramkissoon, J-A. Vergilio, E. Severson, P.J. Stephens, J.S. Ross, J.A. Elvin: Employee of and stockholder in Foundation Medicine, Inc.

Collapse
Poster display session Poster Display session

89P - Pan-cancer genomic analysis of MSI-H tumors reveals commonly altered pathways (ID 2255)

Presentation Number
89P
Lecture Time
13:15 - 13:15
Speakers
  • S. Trabucco
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Microsatellite instability (MSI) is a hallmark of mismatch repair (MMR) deficiency and can be attributed to alterations in MMR-related genes including MSH2, MLH1, MSH6, and PMS2. Although alterations in PI3K pathway genes have been reported in MSI-High (MSI-H) colorectal carcinoma (CRC), a comprehensive enrichment analysis of the genomic landscape in MSI-H and MSI-stable (MSS) populations across tumor types is lacking. To better understand the molecular signatures of MSI and investigate new avenues for therapeutic opportunities, we sought to define the genomic landscape of MSI-H tumors across cancer types.

Methods

Comprehensive genomic profiling of 395 cancer-related genes, including MSI status, was performed on ∼70,000 tumors. To identify potential driver alterations enriched in MSI-H tumors, variants in regions likely to be affected by polymerase slippage were excluded.

Results

As expected, alterations in MSH2, MHL1, MSH6, and PMS2 as well as MMR deficiency variants were enriched in MSI-H specimens regardless of tumor type. We confirmed that variants in PI3K genes were enriched in MSI-H tumors in CRC. Importantly, this was observed across all MSI-H tumors, with 57% of pan-solid MSI-H tumors harboring a PI3K pathway variant compared to 24% of MSS tumors. WNT pathway variants were also enriched specifically in MSI-H tumors, except for CRC, in which frequent APC variants in MSS resulted in WNT enrichment in MSS tumors. Together, 84% of MSI-H tumors have at least one PI3K or WNT pathway variant (compared to 48% of MSS samples). Finally, although ERBB2 alterations occur in both MSS and MSI-H tumors, we found that ERBB2 amplifications occur nearly exclusively in MSS tumors, while ERBB2 missense mutations are enriched in MSI-H tumors.

Conclusions

The genomic landscapes of MSI-H and MSS tumors suggest that they acquire alterations in distinct pathways. MSI-H tumors appear to share signaling pathway alterations across diseases, suggesting that MSI-H tumors may be more molecularly similar to one another than they are to MSS tumors of the same disease histology. These data may provide new avenues for exploration of targeted therapies in MSI-H tumors.

Legal entity responsible for the study

Foundation Medicine Inc.

Funding

Foundation Medicine Inc.

Disclosure

S.E. Trabucco: Current employee at Foundation Medicine. S.L. Maund, P.S. Hegde, S-M.A. Huang: Current employee and has ownership interest in Genentech. R. Hartmaier, K. Gowen, KJ. Sun, G.M. Frampton, P.J. Stephens: Current employee and has ownership interest in Foundation Medicine.

Collapse
Poster display session Poster Display session

90P - Molecular feature and clinical use development of gene expression profile “TP53 signature” in early stage breast cancer (ID 4537)

Presentation Number
90P
Lecture Time
13:15 - 13:15
Speakers
  • S. Yamaguchi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

There have been reported many gene expression profile which can predict prognosis of early stage breast cancer. We have reported the TP53 signature which can predict dysfunction caused by TP53 gene mutation in transcriptome level, and the status defined by TP53 signature can predict more accurate prognosis of early stage breast cancer compare to the status defined by TP53 DNA sequence. Recently, TP53 signature was reported as robust predictor of early stage breast cancer by meta-analysis (BMC Cancer,2015). The aim of this study is to make clear the molecular feature of poor prognosis group diagnosed by TP53 signature and to make easy and quick diagnostic kit which can be used in clinical situation.

Methods

We have done RNA-seq analysis using Hiseq2500 (Illumina) and reanalyzed TCGA data of breast cancer to make clear molecular feature of poor prognosis group defined by TP53 signature. We made simple diagnostic kit using nCounter (Nanostring technology). Using this simple diagnostic kit, we diagnosed 234 breast cancer sample as TP53 signature mutant or TP53 signature wild, and we proved robust prediction power of TP53 signature for early stage breast cancer. We used RNA-seq data to compare prediction power of TP53 signature to Mammaprint, OncotypeDX, TP53 structural mutation.

Results

TP53 signature mutant group have structural mutation in genes, including BRCA1, BRCA2, Rb1 except for TP53, which function is gene repair. In addition, TP53 signature mutant group shows high expression of PD-L1, high mutation burden and high copy number alternation. Analysis of 190 stage I-II breast cancer patients shows TP53 signature by simple diagnostic kit using nCounter has strong prediction power compare to Mammaprint, OncotypeDX, TP53 structural mutation, and clinical factors.

Conclusions

We have developed TP53 signature as diagnostic system for early stage breast cancer which is useful in clinical situation. Poor prognosis group diagnosed by TP53 signature shows molecular features which overlap good response marker of immune-check point inhibitors.

Legal entity responsible for the study

Japan

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

91P - Potential biomarkers of response to DKK1 blockade with DKN-01 in combination with paclitaxel in advanced esophagogastric cancer (EGC) patients (pts) (ID 4895)

Presentation Number
91P
Lecture Time
13:15 - 13:15
Speakers
  • D. Duda
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Overexpression of DKK1, a modulator of canonical WNT signaling, is frequently seen in malignant tumors and is often associated with worse survival. Preclinical studies have shown that DKK1 can augment tumorigenesis by promoting angiogenesis as well as enhancing tumor-associated immunosuppression. In this Phase I trial, DKN-01 (a humanized IgG4 monoclonal antibody against DKK1) showed encouraging early efficacy signals when combined with paclitaxel (DP) in EGC pts (Strickler et al., GI ASCO 2017). We performed correlative studies of plasma biomarkers of angiogenesis and inflammation in these pts.

Methods

Blood samples were collected from 34 patients treated with DP at baseline, and then weekly for 4 weeks (w). Plasma biomarkers were measured by multiplexed array for angiogenesis (bFGF, PIGF/PGF, sVEGFR1, sTIE-2, VEGF, VEGF-C, and VEGF-D) and inflammation (IFN-g, TNF-a, IL-1b, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70 and IL-13) (Meso-Scale Discovery) and by ELISA for HGF and SDF-1a (R&D Systems). Biomarker changes were evaluated by Wilcoxon Sign-Rank test, and correlations with response using Kendall’s test values and with survival using two-sided Wald test in the Cox regression.

Results

Six of 34 EGC pts in this subset analysis (18%) had a PR and 11/34 (32%) showed SD with preliminary median PFS and OS of 13.7w and 28.4w, respectively. DP treatment induced significant and sustained increases in plasma IFN-g, IL-8, VEGF-D and decreases in IL-10 (all p < 0.05). Transient increases in bFGF and PlGF and decreases in sTie2 were also observed. IFN-γ, IL-10, and VEGF-D correlated with overall response and change in target lesion size. Finally, OS was poor in pts with high IL-6, IL-8 and TNF-a (HR > 1 all time-points) and prolonged in pts with greater increases in IL-2 and VEGF-D after treatment.

Conclusions

Prospective plasma biomarker analyses showed that DP treatment changed biomarkers of systemic immunity and angiogenesis in EGC pts, and indicated potential associations between inflammation biomarkers and outcomes. These hypothesis-generating results will inform future prospective investigation of these plasma biomarkers as well as paired evaluation of tumor biopsies for this combination regimen.

Clinical trial identification

Clinical trial information: NCT02013154

Legal entity responsible for the study

Leap Therapeutics, Inc.

Funding

Leap Therapeutics, Inc.

Disclosure

D.G. Duda: Consultant fees from Bayer and research funding from Leap Therapeutics, Inc, Merrimack, Bristol-Myers Squibb and Bayer. C. Sirard: Employee of Leap Therapeutics, Inc. (salary and stock options for compensation).

Collapse
Poster display session Poster Display session

92P - Low expression of miR-20a-5p predicts benefit to bevacizumab in metastatic breast cancer patients treated within the TANIA trial (ID 3161)

Presentation Number
92P
Lecture Time
13:15 - 13:15
Speakers
  • S. Gampenrieder
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Biomarkers predicting response to a bevacizumab containing therapy in metastatic breast cancer (MBC) are of urgent need. MicroRNAs (miRNAs) are involved in regulation of angiogenesis and development of treatment resistance and could therefore provide predictive information.

Methods

Profiling of 754 miRNAs was performed in FFPE tumor samples from 58 MBC patients who received bevacizumab-containing first-line treatment (learning set). Based on median PFS patients were divided into responders (R) and non-responders (NR). Differentially expressed miRNAs between R and NR were selected and validated in a cohort of 57 patients treated with first-line chemotherapy without bevacizumab (control set), to exclude miRNAs providing prognostic information only. In the learning set a multivariate analysis including clinical and pathological information was performed. MiRNAs significantly associated with PFS were further validated in 203 patients treated within the TANIA phase III trial randomizing between chemotherapy plus bevacizumab and chemotherapy alone for two consecutive treatment lines in patients pretreated with bevacizumab in first-line.

Results

Low expression of five miRNAs (miR-9-5p, miR-20a-5p, miR-21-5p, miR-210-3p, and miR-224-5p) was significantly associated with longer PFS in the learning set. For miR-20a-5p (P = 0.035) and miR-21-5p (P = 0.004) this association remained significant in multivariate analysis. In the control set no correlation between expression of those five miRNAs and PFS was seen. In tumor samples from the TANIA trial, low expression of miR-20a-5p was also significantly associated with longer second-line PFS and longer OS in the bevacizumab arm (HR 0.60, 95%CI 0.37-0.89; P = 0.012 and HR 0.54; 95%CI 0.32-0.83; P = 0.007, respectively) but not in the chemotherapy only arm (HR 0.73, 95%CI 0.48-1.09; P = 0.119 and HR 1.01 95%CI 0.63-1.62; P = 0.964, respectively). For miR-21-5p no significant association with PFS or OS in both treatment arms was observed.

Conclusions

MiR-20a-5p expression in breast cancer tissue showed a promising predictive value for identifying patients deriving greater benefit from bevacizumab-containing therapy.

Legal entity responsible for the study

Richard Greil for the translational research project, Roche for the TANIA trial

Funding

Roche

Disclosure

S.P.P. Gampenrieder, G. Rinnerthaler, A. Egle, R. Greil: Advisory role, speakers’ honoraria, travel grants, research funding (no personal payment) from Roche. C. Monzo Fuentes: Travel grants from Roche. C. Hufnagl, C. Hauser-Kronberger: Travel grants, research funding (no personal payment) from Roche. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

93P - Characterization of a novel tumor-suppressor gene CHL1 at 3p26.3 in esophageal squamous cell carcinoma (ID 1054)

Presentation Number
93P
Lecture Time
13:15 - 13:15
Speakers
  • H. Wang
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The major histologic subtype of esophageal cancer, esophageal squamous cell carcinoma (ESCC), is one of the most common cancers and has been ranked as the sixth leading cause of cancer-related deaths in the world. Using human genome U133 Plus 2.0 GeneChip, we identified gene down-regulation of Cell adhesion molecule L1 like (CHL1) located at 3p26.3 in ESCC. We analysed its down-regulated expression, biological effects and prognostic significance in ESCC.

Methods

To determine whether the down-regulation of CHL1 was associated with aberrant methylation. Methylation-specific PCR (MSP) was performed in ESCCs and their corresponding non-tumor tissues, as well as ESCC cell lines. Loss of heterozygosity status of CHL1 was evaluated by fluorescence in situ hybridization (FISH). The effects of CHL1 re-expression or knockdown were determined in proliferation, invasion and metastasis assay. CHL1 target genes and related pathways were identified by protein mass spectrometry, co-immunoprecipitation (Co-IP), immunofluorescence (IF) and western-blot. Clinical impact of CHL1 down-regulated expression was assessed in 287 patients with ESCC.

Results

The results showed that down-regulation of CHL1 was significantly associated with allele loss (14/21) and promoter methylation (19/21; P<0.05) in 39 pairs of ESCC and their corresponding non-tumor tissues by using MSP and FISH. Biofunctional investigation of CHL1 revealed that CHL1 significantly decreased cell proliferation, G1-S cell cycle transition, invasion/migration abilities, and promoted xenograft tumor growth as well as lymph node metastasis in vivo. The anti-proliferation effect by CHL1 was mediated through inducing cell cycle arrest at G1/S checkpoint by down-regulation of p21 and p53 and up-regulation of cyclin D1; the inhibiting metastasis role was by suppressing Epithelial-to-Mesenchymal Transition and F-actin formation which was the result of recruitment Merlin to cell surface expression by CHL1. After a median follow-up of 48.23 months, multivariate analysis revealed that patients with CHL1 protein down-expression had a significant decrease in overall survival. Kaplan-Meier survival curves showed that CHL1 down-regulated expression was significantly associated with shorter survival in patients with ESCC.

Conclusions

CHL1 plays a pivotal tumor suppressive role in ESCC; its down-regulated expression is an independent prognostic factor of patient with ESCC.

Legal entity responsible for the study

Henan Cancer Hospital

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

94P - The role of vitamin D receptor polymorphisms in predicting response to therapy in non-muscle invasive bladder carcinoma (ID 595)

Presentation Number
94P
Lecture Time
13:15 - 13:15
Speakers
  • K. Loh
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Clinicopathological factors predicting for response to Bacillus-Calmette Guerin (BCG) treatment for non-muscle invasive bladder carcinoma (NMIBC) are well defined but there is a paucity of data on genetic factors. Vitamin D has been found to have immunomodulatory effects in pre-clinical bladder cancer studies. Various single nucleotide polymorphisms of the Vitamin D Receptor (VDR) gene has also been found to be associated with response to treatment for mycobacterium. In this study, we evaluated the predictive role of 3 VDR single nucleotide polymorphisms (SNP) in patients with NMIBC in assessing BCG immunotherapy outcome.

Methods

Peripheral blood DNA was prospectively obtained from 140 evaluable EORTC intermediate to high risk NMIBC patients, who underwent post-transurethral resection intravesical regimes of BCG or BCG with interferon alpha. 3 VDR SNPs commonly implicated in susceptibility to tuberculosis infections were evaluated using high resolution melt (HRM) analysis followed by DNA sequencing. Kaplan-Meier together with Log-Rank test and Cox regression methods were used to analyze the data.

Results

Genotype frequencies were similar between the NMIBC patients and controls in accordance to the Hardy Weinberg equilibrium. Mean follow-up time was 91.9 months. Overall mean time to recurrence and progression was 25.8 months and 47.0 months respectively. Kaplan-Meier analysis indicate that individuals carrying the VDR genotype rs1544410 A/G were significantly associated with lower recurrence-free survival rates after BCG therapy (p = 0.007). The VDR rs1544410 “A” allele frequency was found to be higher in patients with bladder cancer recurrences (p = 0.01). No association of VDR genotypes with progression-free survival was found.

Conclusions

Our findings suggest that polymorphisms in the VDR gene correlate with response to BCG therapy in NMIBC patients and further work should be performed to evaluate their utility as predictive markers of response to BCG immunotherapy.

Legal entity responsible for the study

National Healthcare group Domain Specific Review Board

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

95P - Gene signatures as potential predictive markers of response to neoadjuvant chemotherapy in ER+/HER2+ breast cancer patients (ID 3537)

Presentation Number
95P
Lecture Time
13:15 - 13:15
Speakers
  • E. Risi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Chemotherapy (CT) combined with anti-HER2 drugs (H) is the treatment of choice for HER2+ early breast cancer (BC) patients (pts). However HER2+ tumors are clinically and biologically heterogeneous, and treatment response largely varies according to ER status. Predictive biomarkers are urgently needed in this context. We have recently developed a meta-dataset of clinical trials of neoadjuvant CT +/− H (trastuzumab, lapatinib, or both) in HER2+ BC pts annotated for gene expression, hormone receptor status and pathological complete response (pCR) rates. We have shown that a gene-signature (GS) of RB-1 loss-of-function (RBsig) seems to be predictive of response to neoadjuvant CT +/− H in ER+/HER2+ BC pts in this meta-dataset. Here we report the results of additional analyses aimed to evaluate RBsig’s predictive value against 10 previously developed GSs in the subset of ER+/HER2+ BC pts.

Methods

The association of RBsig with pCR was evaluated in comparison with previously described GSs of: low ER signaling, p53 mutation, high PI3K pathway signaling, high expression of HER2 amplicon genes, PAM 50 and 5 immune-related GSs. For each GS, samples were classified as High or Low group using a previously described classifier. Odds Ratio (OR) performance was calculated using the ROCR (v. 1.0) package in R and plotted by forest plot using the survcomp (v. 1.24) package.

Results

RBsig and the HER2 amplicon GS were best at predicting response to neoadjuvant CT +/− H (211 pts; p:0.017). In the subgroup of pts treated with CT alone (n = 94), only the PI3K pathway GS was significantly associated with pCR (p:0.026). In pts treated with CT + H (n = 117) only the HER2 amplicon GS significantly correlated with pCR (p:0.042). RBsig showed a similar trend in both these subgroups (p: 0.078 and 0.104, respectively) The immune GSs and PAM50 were not associated with pCR, independent of treatment received.

Conclusions

RBsig and HER2 amplicon GS are strongly associated with pCR in ER+/HER2+ tumors unselected for treatment. These results support the potential use of such GSs as predictive markers of response to CT +/−H in ER+/HER2+ BC pts. Validation studies are warranted.

Legal entity responsible for the study

Angelo Di Leo

Funding

None

Disclosure

A. Di Leo: Consultant/Advisory Board: Novartis, Pfizer, Lilly. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

96P - Predictive biomarkers for adjuvant therapy in gastric adenocarcinoma (ID 3779)

Presentation Number
96P
Lecture Time
13:15 - 13:15
Speakers
  • E. Pectasides
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Gastric cancer is a common and lethal malignancy, killing over 700,000 people worldwide. Recurrence rates are high even in early stage disease, with 5-year overall survival being < 50% for stage II disease and above. Current guidelines for adjuvant therapy include 5-fluorouracil (5-FU) chemotherapy in combination with radiation. The aim of our study is to develop immunohistochemical biomarkers to predict response to adjuvant chemoradiation in patients with resected gastric cancer.

Methods

A tissue microarray composed of 100 specimens from primary resected gastric cancer cases was constructed. All patients received 5-FU based chemotherapy, and 92% of patients received radiation. Tumors underwent immunohistochemical staining for 11 proteins (HER2, EGFR, p-AKT, PTEN, MTOR, VEGFA, IGF1R, MLH1, MSH2, MSH6, PMS2), and H-scores were calculated. Primary endpoints were progression-free survival (PFS) and overall survival (OS). Survival analysis was performed by Kaplan-Meier method with log-rank test for assessing statistical significance. Multivariate analysis was performed with Cox regression.

Results

Mean follow-up time for the cohort was 39.4 months. Ninety-one of 100 cases had sufficient tissue for biomarker analysis. Interestingly, low expression of MSH2 and MSH6 was significantly associated with shorter PFS, while there was a trend towards worse OS for patients with low MSH2 expression. In multivariate analysis, adjusting for well-characterized prognostic variables, both MSH2 and MSH6 retained their predictive significance. In addition, nuclear expression of the tumor suppressor PTEN was associated with longer OS. No other biomarkers were significantly associated with PFS or OS.

Conclusions

These results indicate that low expression of MSH2 and MSH6 predicts for poorer outcomes in patients with resected gastric adenocarcinoma, independent of other predictive markers.

Legal entity responsible for the study

Hellenic Cooperative Oncology Group

Funding

None

Disclosure

G. Fountzilas: Honoraria: AstraZeneca, Consulting or advisory role: Pfizer, Sanofi, Roche Stock ownership (an immediate family member): ARIAD. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

97P - Large-scale DNA organization is a prognostic marker of breast cancer survival (ID 3898)

Presentation Number
97P
Lecture Time
13:15 - 13:15
Speakers
  • M. Guillaud
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Breast cancer is the leading cause of cancer-related deaths among women worldwide. Current clinicopathological parameters only partially encompass and predict biological diversity and therefore limit our ability to make informed treatment decisions and predict outcomes. The successful future of oncology will rely on our ability to correctly select patients who would benefit from chemotherapy or benefit from treatment intensification, and spare the rest from unnecessary exposure to toxic and expensive therapies. Tumour biology and prognostic markers may be the key to achieving the above goal. We investigated whether changes in Large scale DNA Organization (LDO) of tumour epithelial nuclei is an indicator of the aggressiveness of the tumour.

Methods

We tested our algorithm on a set of 172 TMA cores samples, coming from 95 breast cancer patients. Thirty five patients died of breast cancer and 60 were still alive 0 years after surgery. This TMA slide was stained with Feulgen-thionin and imaged using an high-resolution Imaging system. Automated segmentation of cell nuclei followed by manual selection of intact, in-focus nuclei resulted in an average of 50 cell nuclei per sample. Approximately 60 features measuring Large-scale DNA organization were calculated.

Results

Forward stepwise Linear Discriminant analysis selected 6 features that, combined linearly, gave the best discrimination between nuclei from alive patients specimens and nuclei from deceased patients specimens. Patient LDO score was defined as the percentage of cell nuclei with a high cell LDO. LDO algorithm correctly classified 82.1% patients, with a specificity of 79% and a sensitivity of 88%. Furthermore, individuals with a high LDO score had a 9x fold increase in relative risk of death. In the multivariate Cox regression model, LDO, Node status and Tumor Grade were all significant predictors of cancer death.

Conclusions

This data suggests that LDO could be used to identify patients more likely to have an aggressive disease and thus select a candidate for more aggressive or novel adjuvant therapies.

Legal entity responsible for the study

Martial Guillaud

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

98P - Telomere associated variables and their potential in CLL prognosis (ID 5129)

Presentation Number
98P
Lecture Time
13:15 - 13:15
Speakers
  • N. De Pedro
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The molecular mechanisms that determine disease progression and evolution in CLL are not completely known. Telomeres are usually short in CLL and their attrition may contribute to disease evolution. In addition, telomerase activity (TA) levels have also been associated with prognosis and response to treatment. In order to integrate telomere associated variables (TAV) in CLL disease management further studies with robust methodology are required.

Methods

Purified peripheral CD19 (+) B-cells from 19 healthy donors and 42 CLLs in different stages of disease were obtained from the National Bank of DNA (Salamanca, Spain). Samples were tested to determine full telomere length (TL) distribution- including percentage of short telomeres by a high throughput quantitative fluorescence in situ hybridization (HT-Q-FISH) technique. TA by Quantitative Telomere Repeat Amplification Protocol (Q-TRAP) was also quantified. Full statistical analysis of the results in the context of the clinical history of the patients was performed.

Results

Data from the pilot, retrospective study stablished strong correlations between key CLL variables and the severity of the disease. Overall, TL was shorter and TA presented higher values compared to normal age-matched subjects. Interestingly longer TL was observed for all CLL patients with somatic hyper-mutation (SHM) that in turn, was associated with better prognosis. Concomitantly, TA was elevated in those patients with no SHM and was linked to poor response to treatment and negative prognosis. The percentage of short telomeres was significantly higher for Binet C/Rai III and IV cases.

Conclusions

The use of reliable technologies to measure TAV should be integrated during early diagnostic in CLL to enhance the ability to predict disease evolution. This will require larger, prospective, longitudinal clinical studies.

Legal entity responsible for the study

Life Length S.L.

Funding

Life Length S.L.

Disclosure

N. De Pedro, M. Diez, I. Garcia, R. González, B. Garcia, L. Esteban, L. Otero, P. Najarro, J.C. Estrada, J. García: Employee of Life Length. M. Chiesa: Former employee of Life Length.

Collapse
Poster display session Poster Display session

99P - Micro-RNA profile in advanced metastatic breast cancer as a predictive tool for response to bevacizumab-paclitaxel (ID 5174)

Presentation Number
99P
Lecture Time
13:15 - 13:15
Speakers
  • M. Mendiola
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Bevacizumab-containing therapy improves progression-free survival (PFS) in human epidermal growth factor receptor (HER) 2-negative metastatic breast cancer (mBC), but its use has been questioned due to the lack of benefit in overall survival (OS). To date, biomarkers to predict its positive effect are not available. Interestingly, miRNAs have emerged as regulators of most processes, forming tight interconnected feedback loops with genes under their regulation. Currently, different analyses, such as microarray, are performed in order to identify miRNA biomarkers, using formalin-fixed and paraffin-embedded (FFPE) tissue samples.

Methods

In the present study we recorded clinical data from 57 mBC patients, and selected two (4 + 4) PFS extreme groups for the analysis of the miRnome. Three miRNAs were used for normalization (U6, 191-5p and 103-a-3p). miRNAs for model construction were selected by differential expression between the two groups. Candidates were measured in the remaining 49 cases, and stepwise based Akaike criterion was used for profile generation. Additionally, integrative miRNA and mRNA analyses were done to reveal markers and pathways with potential clinical impact.

Results

We selected two groups of patients with extreme differences on PFS (2.48 ± 1.85 vs 35.43 ±8.03 months) for their miRnome study. The expression profiles of miRNAs in both groups were highly correlated, except for 13 miRNAs where statistical differences arised. These miRNAs were selected as candidates for profile generation on the 49 additional cases, and a combination of five of them (miR-362-3p, miR-150b-5p, miR-671-3p, miR-744-3p and miR-941) was able to accurately discriminate two PFS groups. Additionally, Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses on miRNA possible target genes revealed interesting pathways to explore in these patients, such as cellular adhesion.

Conclusions

By combining experimental approaches and computational biology, we have identified candidate markers of outcome for bevacizumab-containing therapy. The five miRNAs included in the prognostic profile and cellular adhesion related genes should be explored as potential biomarkers.

Legal entity responsible for the study

Fundación para la Investigación del Hospital Universitario La Paz

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

100P - Biomarkers of immune therapy in CUP (ID 3269)

Presentation Number
100P
Lecture Time
13:15 - 13:15
Speakers
  • J. Xiu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Carcinoma of unknown primary (CUP) accounts for approximately 3% of all malignancies. Identification of common cancer pathway alterations (hallmarks of cancer) in diverse cancer lineages offers a rationale for search for biomarkers of targeted therapies in patients with CUP. Avoiding immune destruction is a more recently recognized common cancer characteristic and biomarkers associated with immune check-point blockade were explored in this study.

Methods

392 cases of CUP were tested with NextSeq platform with a 592-gene panel. Tumor mutational load (TML) was calculated using only somatic nonsynonymous missense mutations; microsatellite instability (MSI) was evaluated with NGS by direct analysis of known MSI loci in the target regions of the sequenced genes. ArcherDx FusionPlex Assay was used to detect gene fusions and 52 gene targets were analyzed for 156 tumors. IHC was used to detect tumor expression of PD-L1 (SP142 antibody) and PD-1 TILs (NAT105 antibody) All tests were done in a CLIA-certified lab.

Results

Average patient’s age was 62.4 years; 52% were female. TML high was seen in 12.2% (48/392) tumors using a cutoff of 17 mutations/Mb. MSI-high was detected in 10/392 (2.6%) of tumors. A total of 70 different genes were found mutated with the incidence ranging from 0.3% to 54%; the most frequent were TP53 (53.5%), KRAS (21.5%) and ARID1A (14.6%). Additional notable targetable mutations include PIK3CA (13.1%), CDKN2A (8.1%), PTEN (4.5%), BRAF (4.1%), ATM (3.3%), NOTCH1 (2.4%) and ERBB2 (1.5%). Targetable gene fusions identified included FGFR2 fusions (N = 2), RET (N = 1), RAF1 (N = 1).Tumors with fusions identified carry a lower TML (average 5.9) than the complete cohort (11.7, p < 0.001) with no MSI-high seen in this subgroup. Tumor expression of PD-L1 was seen in 22.5% (82/365) tumors while PD-1 expression on tumor infiltrating lymphocytes seen in 58.7% (37/63). The highest frequency of gene amplification seen were CCND1 (4.7%), FGF3 (3.4%), FGF4(3.4%), FGF19(3.4%), Her2 (3.1%), MYC (2.9%) and AKT2 (2.4%). Of note, CD274 (PD-L1) was rarely amplified (1.4%).

Conclusions

Using a multiplex testing approach, 28% of CUPs had biomarkers (TML-H, MSI-H and/or PD-L1) of response to the immune check-point blockade were identified, making CUP one of the most likely candidate to benefit from immune checkpoint inhibitors.

Legal entity responsible for the study

Joanne Xiu

Funding

None

Disclosure

J. Xiu, Z. Gatalica: Employee of Caris Life Sciences.

Collapse
Poster display session Poster Display session

101P - A circulating TH2 cytokines profile predicts survival in patients with resectable pancreatic adenocarcinoma (ID 2022)

Presentation Number
101P
Lecture Time
13:15 - 13:15
Speakers
  • D. Melisi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Surgery is the only potentially curative option for patients with pancreatic ductal adenocarcinoma (PDAC), but metastatic relapse remains common. We hypothesized that the expression levels of inflammatory cytokines could predict recurrence of PDAC, thus allowing to select patients who most likely could benefit from surgical resection.

Methods

We prospectively collected plasma at diagnosis from two-hundred eighty-seven patients with pancreatic resectable neoplasms. The expression levels of 23 cytokines were measured in ninety patients with PDAC by using a multiplex analyte profiling assay.

Results

Levels higher than cutoff identified of the TH2 cytokines interleukin (IL)4, IL5, IL6, of macrophage inflammatory protein (MIP)1a, granulocyte-macrophage colony-stimulating factor (GM-CSF), and monocyte chemoattractant protein (MCP)1, and of IL17a, IFN-γ- induced protein (IP)10, and IL1b were significantly associated with a shorter median OS. In particular, levels of IL4 and IP10 higher than cutoff identified, and level of TH1 cytokines TNFa and INFg, and of IL9 and IL1Ra lower than cutoff identified were significantly associated with a shorter DFS. In the multivariate analysis, high IP10 was confirmed as negatively associated with OS (HR = 3.097, P=0.014) and IL4 and TNFa remain negatively (HR = 2.75, P=0.002) and positively (HR = 0.224, P=0.049) associated with DFS, respectively. Simultaneous expression of low IL4 and high TNFa identified patients with best prognosis (HR = 0.313, P<0.0001).

Conclusions

We demonstrated that, among a series of cytokines, IL4 is the most significant independent prognostic factor for DFS in resectable PDAC patients, and it could be useful to select patients with high risk of early recurrence who may avoid an unnecessary resection.

Legal entity responsible for the study

University of Verona

Funding

Associazione Italiana per la Ricerca sul Cancro (AIRC)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

102P - Analytic validation of a next generation sequencing assay to identify tumor mutational burden from blood (bTMB) to support investigation of an anti-PD-L1 agent, atezolizumab, in a first line non-small cell lung cancer trial (BFAST) (ID 2948)

Presentation Number
102P
Lecture Time
13:15 - 13:15
Speakers
  • D. Fabrizio
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Recent data suggest that analysis of tumor mutational burden (TMB), a measure of tumor neo-antigenicity derived from tissue biopsies, has shown clinical utility in predicting outcomes for patients treated with anti-PDL1/PD1 therapies across a range of tumor types. Unfortunately, such analyses require quality tumor tissue that in many cases is not available for patients diagnosed in the metastatic setting. As such, there exists a significant unmet medical need for orthogonal diagnostic approaches that enable the analysis of TMB in patient samples without requiring tumor tissue. Herein, we describe the development of an assay to identify TMB from the circulating tumor DNA derived from blood (bTMB), and the analytical validation (AV) that supports its application in a phase III clinical trial in 1L non-small cell lung cancer comparing the anti-PD-L1 agent, atezolizumab, against standard of care platinum-based doublet chemotherapy (BFAST).

Methods

The bTMB assay delivers a count of somatic base substitutions down to 0.5% allele frequency across 394 genes from as little as 1% tumor content in a cell free DNA (cfDNA) sample. AV focused on establishing accuracy and precision, as well as the limit of circulating tumor DNA required to make precise and reliable bTMB calls. Accuracy of the two different bTMB cutoffs being evaluated in BFAST was established against an orthogonally validated TMB platform. Precision was evaluated by comparing the reproducibility of bTMB calls across replicate samples.

Results

The average PPA, NPA and PPV across both bTMB cutoffs was 95%, 100% and 100%, respectively. The average precision was 96%, with a coefficient of variation of 17% across all replicates. The assay limit of detection was defined as 1% tumor content in at least 20 ng of cfDNA.

Conclusions

We have developed and analytically validated a blood-based assay to determine bTMB with high accuracy and precision from as little as 1% tumor content in 20 ng of cfDNA. Clinical validation of bTMB will be established in a prospective, randomized phase III clinical trial, BFAST, with a primary endpoint of progression free survival.

Legal entity responsible for the study

Foundation Medicine, Inc.

Funding

Genentech, Inc.

Disclosure

D. Fabrizio, C. Malboeuf, D. Lieber, S. Zhong, J. He, E. White, M. Coyne, J. Silterra, T. Brennan, J. Ma, M. Kennedy, D. Lipson, G. Otto: Employee and stockholder of Foundation Medicine. E. Schleifman, S.M. Paul, Y. Li, D.S. Shames, C.A. Cummings, E. Peters, M. Kowanetz: Employee and stockholder at Genentech.

Collapse
Poster display session Poster Display session

103P - Comparison of continuous measures across diagnostic PD-L1 assays using image analysis (ID 4712)

Presentation Number
103P
Lecture Time
13:15 - 13:15
Speakers
  • T. Wiestler
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Tumour programmed cell death ligand-1 (PD-L1) expression is a key biomarker in identifying patients who may have an enhanced response to non-small cell lung cancer treatment using anti-PD-1 (e.g. nivolumab and pembrolizumab) or anti-PD-L1 (e.g. atezolizumab and durvalumab). Each treatment is currently used in conjunction with an individual PD-L1 diagnostic immunohistochemistry (IHC) assay and it is unclear whether immunolabelling parameters determined by pathologists are comparable across assays. We extended previous studies (Ratcliffe et al Clin Cancer Res 2017; Ratcliffe et al ASCO-SITC 2017 [abstr 7]) by performing image analysis (IA) with a customised PD-L1 scoring solution to permit a quantitative comparison of the 4 PD-L1 IHC assays.

Methods

We developed an IA scoring algorithm that enabled us to quantify the percentage of positive tumour cells on a whole slide image for 4 PD-L1 assays (Ventana SP263, Ventana SP142, Dako 28-8, Dako 22C3). The analysis was applied to 473 commercially available NSCLC cases (180 cases with SP142). We co-registered the consecutive slides per case and harmonised tumour and exclusion annotations to ensure that readouts of identical areas were compared per case.

Results

In agreement with previous reports, IA results showed concordance between 3 assays, whereas the SP142 assay was discordant. Moreover, high correlation was observed between IA results and pathologist ratings. This correlation could be further improved by matching the information the pathologist received to the same information used in the IA solution: blinding against the assay, scoring on digital scans and masking of non-comparable image regions. The remaining differences represent the differing sensitivity profiles of the assay protocols.

Conclusions

The results of our objective IA suggest differences in sensitivity between the analysed assays. Importantly, despite the observed differences, we confirm previous findings indicating concordance between 22C3, 28-8 and SP263. In addition, our analysis provides a continuous distribution of PD-L1 measurements allowing deeper characterisation of the samples. Tobias Wiestler and Moritz Widmaier are joint first authors.

Clinical trial identification

NA

Legal entity responsible for the study

AstraZeneca PLC

Funding

AstraZeneca

Disclosure

T. Wiestler: Full-time employee Definiens, Stock/shareholder: Definiens/AstraZeneca. M. Widmaier: Full-time employee Definiens. J. Walker, C. Barker, M. Scott: Employee and shareholder AstraZeneca. F. Sekhavati, A. Budco: Employee Definiens. K. Schneider: Full-time employee Definiens AG. K. Steele: Employee MedImmune, shareholder AstraZeneca, Patents/Royalties MedImmune/AstraZeneca. M.C. Rebelatto: Employee AstraZeneca/MedImmune; Stockholder AstraZeneca/MedImmune.

Collapse
Poster display session Poster Display session

104P - Pharmacokinetics (PK) and pharmacodynamics (PD) of a novel carcinoembryonic antigen (CEA) T-cell bispecific antibody (CEA-CD3 TCB) for the treatment of CEA-positive solid tumors (ID 3855)

Presentation Number
104P
Lecture Time
13:15 - 13:15
Speakers
  • I. Melero
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

CEA-CD3 TCB (RG7802, RO6958688) is a novel T-cell bispecific antibody targeting CEA on tumor cells and CD3e on T cells. In mouse models, CEA-CD3 TCB displays potent anti-tumor activity, leads to increased intratumoral T cell infiltration and activation and up-regulates the PD-L1/PD-1 pathway.

Methods

Biodistribution was assessed in mice using SPECT/CT. Patient (pt) samples were from 2 dose-escalation studies in CEA-positive solid tumors. Study 1 (S1): single agent weekly (qw) (0.052-600 mg IV; n = 80), and Study 2 (S2): CEA-CD3 TCB qw (5-160 mg IV) plus atezolizumab 1200 mg q3w (n = 46). Analytics: [CEA-CD3 TCB]—bifunctional PK assay; antidrug antibodies—ELISA; immunophenotyping in peripheral blood (PB)—flow cytometry (FCM), in baseline (BSL) and on-treatment (OT; week 7) biopsies by immunohistochemistry and FCM; plasma cytokines—multiplex assay; PD-L1—SP142 assay.

Results

In mice, CEA-CD3 TCB preferentially accumulated in CEA-positive tumors. CEA-CD3 TCB showed near-linear PK in both studies (S1: 35; S2: 28). In pts with matched BSL and OT biopsies, 7/10 CRC pts treated with ≥ 60 mg of CEA-CD3 TCB in S1 had > 2.4-fold increase in CD8 T cells, which did not correlate with RECIST response. In S2, 2/2 CRC pts receiving ≥ 80 mg of CEA-CD3 TCB (with RECIST reduction ≥ 25%), showed > 8-fold increase in CD8/Ki67 T cells. SUVmax decrease (FDG-PET) correlated with BSL levels of CD4-OX40 and CD8-PD1 in S1 and CD8-OX40 in S2. In PB at week 4, a > 4-fold expansion of activated CD8 T cells (HLA-DR/Ki67) but not CD4, was detected in most pts at doses ≥ 60 mg (S1: 24; S2: 9). In most pts, increases in IL-6 were seen after the first TCB infusion and in fewer cases after the second/third infusion in both studies (S1: 62; S2: 33).

Conclusions

On-treatment increases in intratumoral CD8 T cells consistent with the mechanism of action and support that CEA-CD3 TCB is the first tumor-targeted T cell bispecific showing biological activity. The activation level of intratumoral T cells at BSL could be a predictive biomarker of response. In preclinical models, tumor targeting has been demonstrated. Updated data will be presented. Clinical data are reported separately.

Clinical trial identification

NCT02324257

Legal entity responsible for the study

F Hoffmann-La Roche Ltd.

Funding

F Hoffmann-La Roche Ltd.

Disclosure

I. Melero: Advisory board: Bristo-Myers Squibb, Roche-Genentech, AstraZeneca, Lilly, Merck Serono, Bayer, Genmab, Alligator, Bioncotech, Tusk Grants from: Roche-Genetech, Bristo-Myers Squibb, Bioncotech. N.H. Segal: Consulting and research funds from Genentech/Roche. J. Saro: Employee of Roche and stock holder of Roche. A. Marabelle: PI: Roche, Bristo-Myers Squibb, Merck, Pfizer, Lytix pharma, Eisai, AstraZeneca/Medimmune Scientific Consulting: Roche, Pierre Fabre, Onxeo, EISAI, Bayer, Genticel, Rigontec, Daichii Sankyo, Imaxio, Sanofi, BioNTech. J.M. Cleary: Research funding to the institution from Merrimack Pharmaceuticals, Taiho Oncology, Merck, Roche, Abbvie, Precision Biologics, and Bristo-Myers Squibb. H.I. Hurwitz: Honoraria: Roche and Lilly. Consultant: Roche, Bristo-Myers Squibb, Lilly, Novartis, Incyte, TRACON Pharma, Acceleron Pharma, GlaxoSmithKline, OncoMed. Institutional Funding: Roche, GlaxoSmithKline, Novartis, TRACON Pharma, Bristo-Myers Squibb, Regeneron, Lilly, Macrogeneics, NCI. C. Jamois, S. Bouseida, F. Sandoval, V. Karanikas: Roche employee. E. Andersson: RICM participation in the Roche Connect program. M. Bacac: Employed by Roche and own stock options. T. Nayak: Roche stocks. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

105P - Baseline gut microbiota in metastatic melanoma patients treated with ipilimumab: Relation with clinical response and colitis (ID 4743)

Presentation Number
105P
Lecture Time
13:15 - 13:15
Speakers
  • N. Chaput
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

It is now demonstrated that gut microbiota composition has a determining influence not only on inflammatory bowel diseases but also more broadly on the immunological status of healthy individuals as well as in patients with cancer. We explored the potential role of baseline gut microbiota in anti-tumor response and in intestinal toxicity of patients with metastatic melanoma treated with anti-CTLA4 mAb ipilimumab. Moreover we explored how the composition of gut microbiota could influence not only local gut-immunity but also distant sites such as anti-tumor immunity.

Methods

Fecal microbiota compositions were prospectively assessed at baseline and before each ipilimumab infusion, using 16S rRNA gene sequencing. Patients were further clustered based on microbiota patterns. Peripheral blood lymphocytes immunophenotypes were studied in parallel.

Results

A distinct baseline gut microbiota composition was associated with both clinical response and colitis. As compared to patients whose baseline microbiota was driven by Bacteroides (Cluster B), patients whose baseline microbiota was enriched with Firmicutes (Cluster A) had longer progression-free survival and overall survival. Most of the baseline colitis-associated phylotypes were related to Firmicutes, whereas no-colitis related phylotypes were assigned to Bacteroides. A low proportion of peripheral blood regulatory T cells was associated with Cluster A, long-term clinical benefit and colitis. Ipilimumab led to a higher Inducible T-cell COStimulator induction on CD4+ T cells and to a higher increase in serum CD25 in patients who belonged to Cluster A.

Conclusions

Baseline gut microbiota enriched with Firmicutes is associated with beneficial clinical response to ipilimumab and more frequent occurrence of ipilimumab-induced colitis.

Clinical trial identification

GOLD study: SC12-018; ID-RCB-2012-A01496-37

Legal entity responsible for the study

Coordination: Franck Carbonnel (AP-HP)

Funding

This study was funded by academic groups only: 1- Gustave Roussy Cancer Campus 2- Fondation Gustave Roussy, 3- Direction Générale de l’Offre de Soins (DGOS; GOLD TRANSLA 12-174), 4- Institut National du Cancer (INCa; GOLD 2012-062 N° Cancéropôle: 2012-1-RT-14-IGR-01) 5- SIRIC SOCRATE (INCa DGOS INSERM 6043) 6- MMO program: ANR-10IBHU-0001.

Disclosure

N. Chaput: Grants from Cytune Pharma. E. Soularue: Medical Board for Novartis. F. Carbonnel: Advisory Board for Vifor, enterome, MSD (oncology), Ferring Lecture fees from Abbvie, Janssen, Mayoly Spindler, Hospira, Pfizer, Takeda Research Grants: Alpha Wassermann, Mayoly Spindler. C. Robert: Occasional consultant for Amgen, MSD, Merck, Novartis, Roche, Bristol-Myers Squibb. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

106P - The association between PD-L1 expression, EGFR mutation and ALK translocation in a series of 982 lung cancers (ID 5091)

Presentation Number
106P
Lecture Time
13:15 - 13:15
Speakers
  • M. Evans
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

PD-L1 expression testing is mandatory prior to pembrolizumab prescription in non-small cell lung cancer. Pembrolizumab was made available in the UK through the Early Access to Medicines Scheme (EAMS) in May 2016 and was NICE-approved in December 2016. Our Molecular Pathology Diagnostic Service has been offering PD-L1 testing using Dako’s 22C3 IHC assay in parallel with EGFR and ALK testing. We present here data on the relationships between PD-L1 expression, and EGFR and ALK status in a series of 982 tumours.

Methods

PD-L1 expression testing was performed using the aforementioned assay on the 4800 Dako stainer. EGFR mutation testing was performed using the Therascreen kit on the RGQ platform and using COBAS kit (both screening for exon 19 deletions, L858R, G719X, L861Q, S768I, exon 20 insertions and T790M); ALK translocation was assessed using the D5F3 Ventana antibody on XT platform. PD-L1 was considered positive when more than 1% of tumour cells showed membranous staining.

Results

Of the 982 tumours, 492 were positive for PD-L1 (50.1%), 85 bore EGFR mutations (8.7%) and 14 bore ALK translocations (1.4%). There was no significant difference in PD-L1 expression rate with EGFR mutation status. However, whereas 39.3% tumours with a classical EGFR mutation were PD-L1 positive, 86.4% with a rare EGFR mutation (G719X, L861Q, S768I, exon 20 insertions) were PD-L1 positive (p = 0.0006). PD-L1 positivity rate was 52.3% and 85.7% in ALK non-rearranged and rearranged tumours, respectively (p = 0.01).

Conclusions

Our data show that the presence of classical or of rare EGFR mutations is associated with different degrees of PD-L1 expression, which may have future therapeutic implications. Our data also show that the presence of an ALK translocation is positively associated with PD-L1 expression.

Clinical trial identification

N/A

Legal entity responsible for the study

N/A

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

107P - Association of tumor and stroma PD-1, PD-L1, CD3, CD4 and CD8 expression with response to nivolumab treatment in NSCLC patients (ID 4793)

Presentation Number
107P
Lecture Time
13:15 - 13:15
Speakers
  • S. Sahba
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

PD-L1 immunohistochemistry (IHC) correlates only moderately with response to nivolumab treatment. Characterizing PD-1, PD-L1 and T-cell markers in both tumor and stroma might improve the predictive value of tissue IHC as predictive biomarker in this setting.

Methods

From 08-2015 to 12-2016 patients with stage IV NSCLC treated with nivolumab were registered and prospectively followed. A histological tumor biopsy, obtained before start of nivolumab, was required. Tumor PD-L1 expression and immune cell (IC) PD-L1, PD-1, CD3, CD4 and CD8 expression in tumor and stroma was assessed using IHC on serial sections. IC infiltration was scored semi-quantitatively indicating no, very low, low, intermediate, or high infiltration. Presence of CD4+ macrophages in between tumor cells was used to aid assessment of tumor PD-L1 expression. Nivolumab was dosed 3 mg/kg Q2W and response assessment was done by CT every six weeks.

Results

Overall response rate of pts (n = 65) was 23% and quantifiable (≥1%) tumor PD-L1 expression was found in 25% of pts, versus <1% expression in 75% of pts. Univariate analyses showed a significant correlation between the levels of tumor PD-L1 expression (0%, 1-50%, >50%) and associated response rates of 17%, 22% and 57% respectively. Stromal IC expression of PD-L1, CD3, CD4 and CD8 also correlated with response (p < 0.05 for all markers). CD8+ tumor IC infiltration and stromal PD-1+ staining did not correlate with response. In the subgroup of n = 47 pts with negative (<1%) tumor PD-L1 expression, pts with either high CD3 or high PD-L1 stromal IC expression (n = 45) showed a remarkably high response rate of 59% (p = 0.009).

Conclusions

A clear positive correlation was found between PD-L1 expression and response. The distribution of PD-L1 expression was lower compared to historical data. Availability of CD4 IHC that identified PD-L1 positive macrophages is the explanation for lower percentage of tumor PD-L1 positive samples. Stromal PD-L1, CD3, CD4 and CD8 IC expression were all predictive for treatment response. In tumor PD-L1 negative pts, high stromal PD-L1 and/or CD3 IC expression selected pts with a remarkably high response rate.

Legal entity responsible for the study

A. J. de Langen

Funding

VU University Medical Center, Department of Pulmonology

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

108P - Characterisation of heterogeneity in microsatellite instable (MSI) tumours associated with distinct cell types and immune phenotypes (ID 4742)

Presentation Number
108P
Lecture Time
13:15 - 13:15
Speakers
  • E. Fontana
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In the immunotherapy era, a better understanding of heterogeneity in MSI cancers is required. We evaluated gene expression profiles of MSI colorectal (CRC), gastric (GC) and endometrial cancer (EC) samples with our cell-of-origin signature (CRCassigner), which is able to classify samples in differentiated (goblet-like; CMS3), differentiating (transit-amplifying – TA; CMS2) and less differentiated/mesenchymal (stem-like; CMS4 and inflammatory; CMS1) subtypes to identify whether MSI transcriptional heterogeneity exists.

Methods

Normalised RNAseq/microarrays gene expression profiles and microsatellite status were downloaded from TCGA. CRCassigner classification of samples was performed using Pearson correlation. Samples with low classification confidence were classified as “mixed” subtype. Gene selection enrichment analysis (using published immune markers) and differential protein expression analysis (of PDL1 from Cancer Proteome Atlas data) was performed between inflammatory and goblet-like MSI samples.

Results

The majority of MSI-H in all the 3 cancer types expressed the inflammatory profile. While in MSI-H CRC only two subtypes were present (inflammatory - 91% and goblet-like - 9%), 5 subtypes in MSI-H GC (inflammatory - 45%, goblet-like - 24%, stem-like - 21%, TA - 6%, enterocyte - 3%) and 4 subtypes in EC (inflammatory - 36%, stem-like - 36%, goblet-like - 14%, TA - 14%) were present. Inflammatory MSI tumours from all the three cancer types were significantly (p < 0.05) enriched for genes associated with checkpoint inhibition (PDL1), MHC Class I, Type I interferon response and macrophages compared to goblet-like MSI tumours. On the other hand, goblet-like MSI cancer showed enrichment of B-cells.

Conclusions

MSI tumours are heterogeneous and can be stratified by virtue of differentiation states (or cell-of-origin) and different immune phenotypes. With further studies, this heterogeneity may help select MSI cancer patients for immune checkpoint combination therapies.

Legal entity responsible for the study

Anguraj Sadanandam

Funding

NIHR Biomedical Research Centre at the Royal Marsden Hospital and Institute of Cancer Research, London, UK; Cancer Research UK.

Disclosure

A. Sadanandam: Entitled to a share of royalties received by the licensor for a patent patent number PCT/IB2013/060416. Received research funding from Bristol-Myers Squibb for pancreatic cancer. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

109P - Clinical implication of PLR and PD-L1 in breast cancer patients (ID 1362)

Presentation Number
109P
Lecture Time
13:15 - 13:15
Speakers
  • S. Wang
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Anti-tumor action of the host immune systems and cancer-immune interaction are associated with tumor prognosis. Programmed death ligand-1 (PD-L1) expression was found as an unfavorable prognostic factor in breast cancer in our previous study. Besides, PD-L1 and biomarkers of the host immunity which included Neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR) have been reported in predicting prognosis of biliary tract cancer. However, combination of NLR/PLR and PD-L1 in breast cancer has not been reported. The aim of this study is to evaluate the prognostic role of NLR/PLR and PD-L1 in breast cancer.

Methods

A total of 870 patients with breast cancer treated in Sun Yat-Sen University Cancer Center from 2000 to 2012 with known PD-L1 status were included. Clinicopathological data and pretreatment complete blood count were retrospectively collected. X-tile was used to generate the optimal cut-off value of NLR and PLR. Kaplan-Meier and univariate Cox proportional hazards model analyses were used to compare the survival of patients between different groups.

Results

High PLR group achieved worse result than low PLR group in OS and DFS (5-year OS rate: 82.6% vs 88.8%, p = 0.010; 5-year DFS rate: 78.7% vs 85.6%, p = 0.003). High PLR was associated with shorter DFS (adjusted HR = 1.540, 95%CI: 1.124-2.110, p = 0.007), while high PLR was not an independent factor for OS (adjusted HR = 1.001, 95%CI: 0.999-1.003, p = 0.488). NLR was not associated with patients’ survival outcome. And we found patients with PD-L1 expression and high PLR had the worst prognosis. The 5-year DFS rates were 68.4%, and 85.8% in high PLR+PD-L1 (+) group and low PLR+PD-L1 (-) group respectively (p = 0.002). The 5-year OS rates were 73.4% and 90.1%, respectively (p < 0.001).

Conclusions

High PLR are associated with poor DFS in breast cancer patients. PD-L1 expression combined with high PLR was associated with an aggressive clinical outcome. Further studies are needed to evaluate the predictive value of combination of PD-L1 and peripheral blood immune markers.

Legal entity responsible for the study

Shusen Wang

Funding

National Natural Science Foundation of China (81502302); Science and Technology Program of Guangdong Province (2014A020212384; 2016A020215079)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

110P - Prognostic and predictive value of lymphovascular invasion and lymph node status among breast cancer subtypes (ID 5019)

Presentation Number
110P
Lecture Time
13:15 - 13:15
Speakers
  • G. Liao
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Breast cancer subtype (BCS) and lymphovascular invasion (LVI) have both been independently demonstrated as prognostic factors. The objective of this investigation was to evaluate the prognostic power of LVI and lymph node status among BCSs.

Methods

From an institutional database, 2017 women with a histopathologically confirmed diagnosis of breast cancer treated between January 2006 and December 2014 were consecutively selected for participation in this study.

Results

Of the 2017 patients with breast cancer in the BCS groups, the highest OS and RFS rates were observed in luminal A subtype (93.6% vs. 95.1%, respectively) and the lowest were observed in TN subtype (85.3% vs. 83.0%, respectively). There were significant differences in OS according to the LVI status between the luminal A, luminal B and luminal HER2 subtypes. There were also a significant difference in the RFS rate of the luminal A, luminal B, luminal HER2 and HER2 subgroup. Therefore, we inferred that there were stronger links with LVI and BCS with regard to OS and RFS rates. Kaplan-Meier analysis showed that there were significant differences in the OS and RFS rates according to the LVI status among the BCS groups. There were significant differences in OS according to the LVI status in the distribution of the luminal A, luminal B, luminal HER2, and TN subtypes. There were also significant differences in the RFS rates among the luminal B, luminal HER2, and HER2 subtypes. On multivariate analysis, after controlling for age, tumor size was independently associated with LVI and lymph node status among all BCS groups. There were significant differences in OS according to the status of lymph node-negative and LVI-positive in the luminal HER2 subtype, as well as lymph node-positive and LVI-positive in the TN subtype. There were also significant differences in RFS according to the status of lymph node-negative and LVI-positive in the luminal A subgroup.

Conclusions

LVI and lymph node status were important prognostic factor for OS and RFS among all BCSs. In lymph node-negative breast cancer, luminal HER2 had greater predictive value for OS, whereas luminal A displayed greater predictability for RFS. In lymph node-positive breast cancer, the TN subtype had greater predictive value for OS.

Clinical trial identification

nil

Legal entity responsible for the study

Tri-Services General Hospital, National Defense Medical Center

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

111P - PD-L1 expression in TNBC: A predictive biomarker of response to neoadjuvant chemotherapy? (ID 2570)

Presentation Number
111P
Lecture Time
13:15 - 13:15
Speakers
  • A. Botticelli
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Immune system plays an important role in tumor surveillance and escape. Recently tumor infiltrating lymphocytes (TILs) have been proposed as a predictive biomarker for clinical outcome and pathological response (pR) after neoadjuvant (neoadj) chemotherapy (CT) in breast cancer. PD-L1 is expressed in about 20% of TNBC, suggesting the possibility of being a therapeutic target for this subtype of cancers. Here we studied the association between PD-L1 expression and pR in TNBC.

Methods

We enrolled 54 pts who had received neoadj CT (EC for 4 cycles followed by Paclitaxel q21 for 4 cycles) between Jan 2008 and Dec 2016 at Policlinico Umberto I and San Giovanni Hospital of Rome. We performed IHC for CD20, CD3, CD4, CD8, CD68, N-CAM and PD-L1 (Ventana SP142 clone) in basal paraffin-embedded biopsies. PD-L1 expression on tumor cells was evaluated both qualitatively (membrane staining intensity 0 to 3+) and quantitatively (% of positive cells.). The percentage of TILs positive for PD-L1 was also recorded. Statistical analysis was performed with T di Student test and χ2 test.

Results

We enrolled 54 pts (median age: 50 y; range 28-75) affected by TNBC: 51 ductal (94.4%), 2 metaplastic (3.7%), 1 lobular (1.9%). The clinical stage before neoadj CT was as follow: 12.9% cT1 (7 pts), 72.2% cT2 (39 pts), 3.7% cT3 (2 pts), 1.85% cT4 (1 pt) and 5.5% cTx (3 pts). 23 pts were cN + (42.5%). After neoadj CT 30 pts underwent mastectomy (55%) and 24 conservative surgery (45%). 19 pts (35%) showed pCR. No significant associations were found between pR and cT, cN, age, histotype and KI-67. In 64.8% of basal biopsies (35 pts) PD-L1 was not detected on tumor cells and in 18.5% (10 pts) it was absent in the immune infiltrate. PD-L1 expression was detected in > 25% of tumor cells in 4 pts, all of which showed pCR (p = 0.024). No associations between intensity of membrane staining and pR were detected (p = 0.7). The immune infiltrate was characterized mostly by the presence of CD3+ CD8+. No statistically significant associations between and PD-L1 expression on immune infiltrate were detected.

Conclusions

Basal PD-L1 expression on cancer cells was associated with a better pR in TNBC undergoing neoadj CT. The introduction of anti PD-1/PD-L1 therapy in this setting of pts could lead to interesting results.

Legal entity responsible for the study

Sapienza University of Rome

Funding

None

Disclosure

P. Marchetti: Advisory board and meeting with Pfizer, Roche, Novartis, MSD, Bristol-Myers Squibb, Ipsen, AstraZeneca, Boehringer Ingelheim. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

112P - Pathological evaluation of tumor infiltrating lymphocytes and the benefit of nivolumab in advanced non-small cell lung cancer (NSCLC) (ID 5095)

Presentation Number
112P
Lecture Time
13:15 - 13:15
Speakers
  • I. Gataa
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Assessment of tumor infiltrating lymphocytes (TIL) by pathologists using Hematoxylin-Eosin (H&E), has been described as a prognostic factor in resected NSCLC. We aimed to correlate TIL to the benefit from nivolumab in patients (pts) with treated advanced NSCLC.

Methods

Patients with advanced NSCLC treated with nivolumab, with biopsy available for evaluation, were included between November 2012 and February 2017 in two cancer centers. Patients characteristics and outcome were collected. The percentage of tumor infiltrating lymphocytes in the stroma was evaluated using H&E staining from archival pretreatment tumor tissue samples. Primary endpoint was to correlate TIL density with progression free survival (PFS).

Results

Out of ninety-eight patients included. 60 (61%) pts were male, with median age of 61 years and 85 (89%) were smokers. Sixty three (73%) pts were PS 0-1. Sixty tumors (61%) were adenocarcinoma, 29 (30%) squamous and 9 (10%) other histologies. Among 83 tumors with known molecular profile, 22 (27%) were KRAS mutated 7 (8%) EGFR mutated, 1(1%) ALK positive. The median treatment line was 3 (2-4). The median follow up was 8 months (m)(95%CI[6-19]). The median PFS was 2 m (95%CI[1-5]). The ORR was for 16%. The median TIL density was 5% (2-15). TIL density ≥5% correlated with PFS in univariate and multivariate analysis (HR: 0.48 [0.28-0.82] p = 0.007 and HR:0.31 [0.14-0.68] p = 0.004 respectively). TIL density ≥5% was also associated with better ORR (OR = 3.5, 95%CI [1.06-11.7], p = 0.04).

Conclusions

Pathological assessment of TIL allows an easy evaluation of immune infiltration in NSCLC and independently correlates PFS in NSCLC pts treated with nivolumab. Results from validation cohorts and combination with other morphological and immunohistochemical parameters will be reported.

Legal entity responsible for the study

Ithar Gataa

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

113P - Could a systemic inflammation response index (SIRI) predict overall survival (OS) in metastatic pancreatic cancer (PC)? (ID 3978)

Presentation Number
113P
Lecture Time
13:15 - 13:15
Speakers
  • V. Pacheco Barcia
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Cancer-associated inflammation is a key molecular feature of PC and may affect the clinical course. The aim of this study was to evaluate the prognostic relevance of SIRI based on peripheral neutrophil, monocyte, and lymphocyte counts in metastatic PC and its association with the metastatic site.

Methods

Retrospective analysis of the medical records of patients with pathologically confirmed metastatic PC between January 2011 and December 2016. Patients were classified as having liver metastases (LM) or extrahepatic metastases alone (EM). Associations with overall survival (OS) were analyzed using Cox proportional models.

Results

A total of 37 patients were included (47 men; median age 63). Median TTP was 4 months and median OS was 6 months. 29 patients (78%) had LM and 8 (22%) EM. 33 patients (89%) received CT: 13 (40%) Gemcitabine (GEM) plus Nab-Paclitaxel, 9 (27%) GEM in monotherapy, 7 (21%) GEM plus Erlotinib and 4 (12%) an Oxaliplatine doublet. Mean Ca 19 9 levels in patients with LM were 199349 and with EM 9107. Univariate analysis identified SIRI scores ≥1,9 as significant risk factor for OS. Age, sex and high CA 19,9 levels had no prognostic significance for OS in all groups. Patients with LM showed a higher SIRI than those with EM (p = 0,03). Patients with SIRI scores < 1,9 (55%) compared to those who had SIRI scores ≥1,9 (45%) had a longer OS (p = 0,01). LM were significantly associated with shorter OS (hazard ratio [HR] 2.79; 95% confidence interval [CI] 1.36-5.34; p = 0,002) but not those with EM (HR 1.83; CI 0.71-4.72; p = 0,2). An SIRI ≥1,9 resulted in a shorter OS compared to an SIRI <1,9 (HR 2.13; CI 1.10–4.10; p = 0.024).

Conclusions

SIRI is associated with survival in patients with metastatic PC. In patients with LM, unfavourable SIRI may be associated with higher tumor burden. In our experience, a baseline SIRI ≥1.9 duplicates the risk of mortality and this finding may allow better risk stratification.

Legal entity responsible for the study

Medical Oncology Department, Hospital Universitario de La Princesa

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

114P - Molecular imaging with 18F-fluoroestradiol (18F-FES) to assess intra-patient heterogeneity in metastatic breast cancer (MBC): A European TRANSCAN program (ID 1919)

Presentation Number
114P
Lecture Time
13:15 - 13:15
Speakers
  • A. Gennari
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Endocrine responsiveness in Estrogen Receptor positive (ER+) MBC is based on the level of ER expression on the primary tumor and/or metastatic lesion. In this study, nested in the ET-FES JTC 2011 TRANSCAN project, we used molecular imaging with 18F-FES to explore intra-patient heterogeneity in ER expression at different metastatic sites and to identify patients who are not likely to benefit from ET.

Methods

ER+ patients at first relapse underwent at baseline a 18F-FES PET/CT plus a 18F-fluoro-2-deoxy-D-glucose (FDG) PET/CT. Patients were classified into 4 18F-FES/FDG subgroups based on the proportion of FDG avid metastatic tumor load with high 18F-FES uptake (Gebhart Ann Oncol 2016). Subgroup A was considered positive (100% of concordance); subgroups B and C were considered partially positive or partially negative, with different degrees of 18F-FES uptake; subgroup D was considered negative (100% of discordance). Patients with global SUV ≥2 received first line ET while those with SUV <2 were randomized to ET or to chemotherapy. HR for progression was calculated comparing patients with concordant 18F-FES/FDG lesions (group A) with all the other patients.

Results

So far, 80 patients have been enrolled in the ET-FES trial and 79 are included in the present analysis. At baseline evaluation, 53 patien4ts (67.1%) were classified as 18F-FDG/18F-FES positive (A); 16 patients (20.3%) showed some degree of intra-patient heterogeneity (11 group B and 5 group C); 10 patients (12.6%) were classified as D with all lesions being 18F-FES negative. In the 64 patients with a response evaluation available at time of analysis, 26 have shown progression. ET was administered in 40 patients in group A and in 24 patients in groups B + C + D. The use of ET alone in partially positive (B) or partially negative (C) or negative (D) patients was associated with a 79% absolute increase in the risk of progression (HR 1.79, p 0.2) compared to patients in group A.

Conclusions

Pretreatment ER biomarker imaging at different metastatic sites with 18F-FES PET/CT indicate the presence of a significant intra-patient heterogeneity in MBC and represents a promising tool to select patients who are unlikely to benefit from ET alone.

Clinical trial identification

EUDRACT 2013-000-287-29

Legal entity responsible for the study

E.O. Ospedali Galliera, Genoa, Italy

Funding

TRANSCAN JTC 2011

Disclosure

C. Brambati: Employer Advanced Accelerator Applications. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

115P - Evaluation of a predictive radiomics signature for response to immune checkpoint inhibitors (ICIs) (ID 1102)

Presentation Number
115P
Lecture Time
13:15 - 13:15
Speakers
  • A. Prawira
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Radiomics (RAD) uses advanced image processing techniques to extract a large set of quantitative texture and geometric features from tumor regions of interest, and subject these to a supervised machine learning protocol to train a classifier, which we exploit to develop a predictive signature of response to ICIs. We previously developed a lesion-based predictive RAD classifier of response for recurrent/metastatic squamous cell carcinoma of the head and neck (RM SCCHN) pts to ICIs based on RAD features extracted from their CT images (Prawira, ESMO 2016).

Methods

INSPIRE (NCT02644369) is an investigator-initiated phase II study evaluating biomarkers for pembrolizumab (anti-PD1 monoclonal antibody) in multiple cohorts of pts with advanced solid tumors. The primary endpoint of this project is to validate the previously developed RAD classifier from RM SCCHN pts, with pts from INSPIRE. Texture feature algorithm generation and accuracy determination were as previously described. Cross validation accuracy values were generated for combinations of 3 parameters: fraction, cost, and gamma, yielding a 3 dimensional (3D) accuracy space.

Results

Eighty lesions from 23 pts were available for analysis: median age 59, 22% males. Best response: 12 progressive disease, 3 partial response, 8 stable disease (median duration 18 weeks). Primary site: SCCHN/2, triple negative breast cancer/4, high-grade serous ovarian cancer/11, malignant melanoma/2, other advanced cancers/4. Twentyseven lesions were excluded as RECIST 1.1 responses were not yet available. Fiftythree target lesions were contoured. Per lesion RECIST 1.1 radiological outcome: 17 R, 36 NR. Cross validation in the 3D space yielded a set of ROC curves with an accuracy of 71.4% (AUC 0.41, p = 0.7) with 11.2% sensitivity and 99.9% specificity, where specificity corresponds to the proportion of NR tumors classified correctly, and sensitivity to the proportion of R tumors classified correctly.

Conclusions

Heterogeneous histologies and low pt numbers may account for the negative result in this study, suggesting that RAD may be histology-specific. Further validation in a large independent cohort of RM SCCHN pts treated with pembrolizumab is planned.

Clinical trial identification

INSPIRE (NCT02644369)

Legal entity responsible for the study

Princess Margaret Cancer Centre, Drug Development Program

Funding

Merck

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

116P - Clinical and pre-clinical biomarkers of Regorafenib (REG) efficacy in metastatic colorectal cancer (mCRC) in a phase II trial (ID 4693)

Presentation Number
116P
Lecture Time
13:15 - 13:15
Speakers
  • K. Khan
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

REG demonstrated efficacy in pre-treated mCRC pts. Lack of predictive biomarkers, potential toxicities and cost/effectiveness concerns highlight the unmet need for better patient selection.

Methods

RAS mutant mCRC pts with biopsiable metastatic deposits were enrolled in this phase II trial. Tissue biopsies (6-12 cores) were obtained at baseline (BL), after 2 months if stable disease (SD) and at disease progression (PD). Dynamic contrast enhanced (DCE) MRI was acquired pre and at day 15 post-treatment. Median values of volume transfer constant (Ktrans), enhancing fraction (EF) and their product, KEF [Ktrans*EF/100] were generated. Circulating tumour (ct) DNA was collected monthly until PD and tested for clonal RAS mutations by digital droplet PCR. PDOs derived from responders and non-responders pts were implanted orthotopically in the liver of mice and treated with REG for 5 days. Changes in tumour and fractional blood volume (fBV) were monitored by oxygen-enhanced MRI.

Results

mCRC pts (n = 27) with paired MRI scans were analysed; a single target lesion per pt was chosen (25 liver and 2 pelvic metastases). Median KEF decrease was 58.2%. In the 23 analysable pts (4 received ≤1 cycle of treatment due to toxicities),>70% drop in KEF(8/27) was associated with higher disease control rate (DCR) measured by RECIST 1.1 at 2 months (m) (p = 0.05), progression free survival (PFS) [HR = 0.24 (0.07-0.86), p = 0.03], 6-m PFS (43.8% VS 0%) and overall survival (OS) [HR 0.08 (0.01-0.63), p = 0.02]. In all pts with DCR, PFS was found to be 5.6 vs. 4.2 m [HR 0.30 (95% CI 0.06-1.49), p = 0.140) and OS was 15.2 vs. 5.8 m [HR 0.11 (95% CI 0.01-1.06), p = 0.057]. KEF drop correlated with CD-31 reduction in sequential tissue biopsies (p = 0.04). RAS mutant clones decay in ctDNA after 8 weeks of treatment was associated with better PFS [HR 0.21 (0.06 - 0.71), p = 0.01] and OS [HR 0.28 (0.07 - 1.04), p = 0.06]. PDOs xeno-transplants treated with REG compared to controls had significant lower tumour fBV (4.5 VS 10.6, p = 0.03) and lower microvascular density measured by CD31 staining (4.3 VS 8.9, p = 0.02).

Conclusions

Combining DCE-MRI and ctDNA predicts depth and duration of anti-angiogenic response to REG with potential health economic implications.

Clinical trial identification

clinical trials.gov number NCT03010722

Legal entity responsible for the study

The Royal Marsden NHS Foundation Trust

Funding

Bayer Oncology Group

Disclosure

K. Khan: Advisory board for Bayer Oncology Group. D. Cunningham: Research funding from: Roche, Amgen, Celgene, Sanofi, Merck Serono, Novartis, AstraZeneca, Bayer, Merrimack and MedImmune. I. Chau: Advisory roles with Merck Serono, Roche, Sanofi Oncology, Bristol-Myers Squibb, Eli-Lilly, Novartis, Gilead Science. Research funding from Merck-Serono, Novartis, Roche and Sanofi Oncology, and honoraria from Roche, Sanofi-Oncology, Eli-Lilly, Taiho. C. Peckitt: Advisory roles with Sanofi. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

117P - Anti-HER2 therapy efficacy in HER2-negative metastatic breast cancer with HER2-amplified circulating tumor cells: results of the CirCe T-DM1 trial (ID 5522)

Presentation Number
117P
Lecture Time
13:15 - 13:15
Speakers
  • F. Bidard
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Changes of HER2 status has been reported in circulating tumor cells (CTC) isolated from preclinical models and metastatic breast cancer (MBC) patients. The prospective multicentric phase II “CirCe T-DM1” trial was set up to assess whether HER2-amplified CTC are detectable in HER2-negative MBC and whether these cancers would respond to anti-HER2 therapy.

Methods

HER2-amplified CTC were screened in HER2-negative (HER2-/ER- and HER2-/ER+) MBC patients starting a 3rd line or 4th line of systemic therapy. CTC were detected by CellSearch® (Janssen Diagnostics) and FISH was performed on isolated CTCs. HER2-amplification was defined by a HER2/CEP17 ratio ≥2.2. Patients with ≥1 HER2-amplified CTC, measurable disease and adequate organ function were eligible. After stratification according to amplified CTC count (< vs ≥ 3), patients received single agent T-DM1. The primary endpoint of the study was the response rate by RECIST criteria.

Results

From 11/2013 to 08/2016, 155 MBC patients were screened. 11 (9.2%) and 3 patients (2.5%) had 1-2 and ≥3 HER2-amplified CTCs respectively (minimal HER2/CEP17 ratio: 2.5). In the 14 patients with HER2-amplified CTC, the fraction of HER2-amplified CTCs among all detected CTCs was low (median 1.6%, range [0.3%-35.3%]), and presence of HER2-amplified CTCs was not associated with any patients’ characteristics. 11 patients were treated with single agent T-DM1. Partial response was confirmed in one patient with 1 HER2-amplified CTC (among 9 CTC detected); median PFS was 4.9 months (range: 1.8-10.1).

Conclusions

This study shows that CTCs with a true HER2-amplification can be detected in advanced HER2-negative MBC, mostly as a minor CTCs subset. Although one confirmed response was observed in our study, the overall low response rate to specific anti-HER2 therapy does not support the clinical utility of such strategy in that setting.

Clinical trial identification

NCT01975142

Legal entity responsible for the study

Institut Curie

Funding

Roche

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

118P - Nationwide external quality assessment (EQA) of EGFR testing in circulating tumor DNA: The French experience (ID 3171)

Presentation Number
118P
Lecture Time
13:15 - 13:15
Speakers
  • M. Denis
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Detection of EGFR mutation in circulating tumor DNA (ctDNA), a powerful blood-based biomarker with multiple clinical applications for lung cancer patient, is technically challenging because it requires sensitivity and specificity. In order to evaluate the performance of the french laboratories performing this assay, we set up a national EQA scheme for circulating tumor DNA.

Methods

Artificial samples were prepared by spiking DNA extracted from control FFPE sections containing specific mutations (Horizon Diagnostics, from 25 to 350 copies/mL of plasma, as determined by digital PCR) in normal plasma (Clinisciences). Aliquots (2 ml) of 10 different samples were sent in dry ice to 43 laboratories. DNA extraction and EGFR testing were performed according to local practice. Laboratories were requested to search for exon 19 deletions, p.L858R, p.G719X and p.T790M mutations. Data were collected on a web questionnaire within one month and compared to the expected results.

Results

We collected 30 complete sets of data. DNA was extracted using the QIAmp® circulating DNA kit (Qiagen; n = 13), the Cobas® cfDNA sample preparation kit (Roche; n = 9) or the Maxwell® system (Promega; n = 7). The most widely used methods were the Cobas® EGFR Mutation Test v2 (Roche; n = 10), digital PCR (n = 8) and NGS (n = 6). Among the 10 labs using the Cobas®, 9 obtained the 10 expected genotypes. This number dropped to 3 (out of 6 labs) for NGS, and 2 (out of 8 labs) for dPCR, because of false negative results, false positive results, and not contributive tests.

Conclusions

Digital PCR and NGS are known to be highly sensitive techniques. However, the results of this EQA suggest that in routine clinical practice, ctDNA analysis requires technical skill or/and validated bioinformatic pipeline to reach high sensitivy and specificity. Under the specific conditions of this scheme, the Cobas® method appeared to be the most robust approach. This external control will allow the laboratories to evaluate their practice and improve their process. A similar approach targeting other genes (BRAF, KRAS and NRAS) is being developed, and additional EQA schemes will be set up at the nationwide level. Supported by a grant from AstraZeneca.

Legal entity responsible for the study

Gen&Tiss

Funding

AstraZeneca

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

119P - IL-8 as a pharmacodynamic biomarker for TGF-β2 antisense (trabedersen) therapy: Results of a phase II trial (ID 5478)

Presentation Number
119P
Lecture Time
13:15 - 13:15
Speakers
  • L. Hwang
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In a Phase I/II study of Trabedersen (OT-101), a phosphorothioate antisense specific for human TGF- β2 mRNA, patients with advanced pancreatic cancer (PAC) treated in the 2nd-line and beyond exhibited improved overall survival (OS) when OT-101 was followed with chemotherapy. Here, we examined the association between plasma levels of cyto-/chemokines and OS outcomes to identify potential biomarkers for improved OS.

Methods

37 PAC patients were treated with continuous IV infusion in escalating doses of 2 treatment schedules (7-days-on, 7-days-off and 4-days-on, 10-days-off). Plasma levels of 31 cyto-/chemokines were measured at 8 separate time points over 3 cycles of OT-101 treatment (140 mg/m2/day, 4-days-on, 10-days-off). Standardized maximum levels of individual cyto-/chemokines on Day 2 were subtracted from levels on Day 5 of each cycle of treatment and correlated with log10 transformed OS values. Feedback interactions with PK parameters were also investigated utilizing an ANCOVA model.

Results

A median OS of 14.5 months and 2.6 months was observed for 2nd-line patients treated with and without subsequent chemotherapies, respectively (p = 0.0009). Increasing difference of IL-8 levels at Day 2 vs Day 5 was positively correlated with OS (R2 = 0.58, P = 0.0066). Stratifying for patients with and without chemo, R2 increased to 0.99 and 0.77 respectively. Similar results were observed for IL-15, with R2 = 0.93 in patients with chemo and R2 = 0.50 in those without. ANCOVA models for two PK parameters exhibited significant model fits (F3,7 = 7.89, P = 0.012 for Simulated Vz Mean; F3,7 = 8.18, P = 0.011 for Simulated Cl Mean) and the interaction effects resulted in lower p-values for the correlation of OS vs IL-8 levels.

Conclusions

Increasing peak levels of IL-8 and IL-15 response on Day 2 of OT-101 treatment correlated with OS in PAC patients. This correlation with OS was evident regardless of subsequent chemotherapy or not indicating spikes in IL-8 and IL-15 as potential biomarkers for OT-101.

Legal entity responsible for the study

Oncotelic Inc.

Funding

Oncotelic Inc.

Disclosure

L. Hwang, W. Wang, S. Qazi, K. Ng, O. D’cruz, V. Trieu: Employee

Collapse
Poster display session Poster Display session

120P - Predictive assay for anti-angiogenic agents (AADx) identifies molecular subgroups of RASwt mCRC with differential efficacy of FOLFIRI plus bevacizumab in the FIRE-3 (AIO KRK-0306) trial (ID 3385)

Presentation Number
120P
Lecture Time
13:15 - 13:15
Speakers
  • S. Stintzing
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The FIRE-3 trial compared 1st-line therapy of FOLFIRI plus either cetuximab or bevacizumab in 592 KRAS exon 2 wildtype (wt) mCRC patients. The subgroup of extended RAS wt patients consisted of 400 patients. The AADx molecular assay has previously been shown to identify a poor prognosis angiogeneic subgroup across multiple cancer types including colorectal cancer. Both bevacizumab (through inhibition of VEGFR-activation) and cetuximab (through inhibition of EGFR-signaling) would be expected to have anti-angiogenic effects in colorectal cancer. The predictive role of AADx in FOLFIRI plus bevacizumab or cetuximab treated in colorectal cancer patients remains unclear.

Methods

Transcriptional profiling of 501 formalin fixed paraffin embedded pre-treatment samples from the ITT population was performed using the Almac Diagnostics XcelTM array. Patients were classified by the AADx assay as ANGIO ON or OFF based on a predefined score. ORRs were compared using Fischeŕs exact test. Progression free survival (PFS) and Overall survival (OS) times were compared using Kaplan-Meier estimation and log-rank tests. Hazard ratios (HR) were estimated according to the Cox proportional hazard method.

Results

The AADX assay was successfully applied to 438 out of 501 specimens available from the study population (n = 752). Of those, 315 had a RAS wt tumor and 123 a RAS mutant. The correlation between RAS status and AADx score with respect to ORR; PFS, and OS were complex (Table). The addition of cetuximab to FOLFIRI was significantly superior to the addition bevacizumab in “ANGIO ON” tumors most likely reflecting the strong link between EGFR-signaling and angiogenesis in colorectal cancer.

120P

RAS wild-typeAADx scoreORRp ORPFS monthsp HROS monthsp HR
FOLFIRI + CetuximabANGIO ON65.2%0.69 0.8710.60.72 1.0729.80.88 1.04
ANGIO OFF68.3%10.630.6
FOLFIRI + BevacizumabANGIO ON46.3%0.0038 0.399.10.0002 0.5221.20.0062 0.59
ANGIO OFF69.1%13.129.1
RAS MutantAADx scoreORRp ORPFS monthsp HROS monthsp HR
FOLFIRI + CetuximabANGIO ON38.6%0.93 0.948.20.49 1.2323.00.81 0.92
ANGIO OFF40.0%7.719.2
FOLFIRI + BevacizumabANGIO ON58.8%0.13 2.111.20.35 1.3023.10.02 2.00
ANGIO OFF40.0%10.218.5

Conclusions

Here, we present data demonstrating that it possible to define subgroups in the group of wt mCRC patients within the FIRE-3 trial that responded differently to the addition of cetuximab or bevacizumab.

Clinical trial identification

NCT00433927

Legal entity responsible for the study

Klinikum der Universität München

Funding

ALMAC Inc.

Disclosure

S. Stintzing: Honoraria for talks and advisory board from: Amgen, Bayer, Lilly, Merck, Sanofi, Roche. B. Price, A. McCavigan, S. Walker, P. Harkin, R. Kennedy, L. Knight: ALMAC Diagnostics. V. Heinemann: Honoraria for talks and advisory boards by: Amgen, Servier, Sirtex, Merck, Roche, Bayer, Lilly, Sanofi. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

121P - Influence of HIF-2alpha deregulation and overexpression of VEGF ligands on the response to aflibercept: Identification of predictive biomarkers (ID 2134)

Presentation Number
121P
Lecture Time
13:15 - 13:15
Speakers
  • A. LARSEN
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Angiogenesis inhibitors are widely used for treatment of metastastic colorectal cancer. However, no predictive biomarkers are currently available for patient selection. Besides the tumor-associated endothelial cells, VEGF-signaling inhibitors target CRC cells that express VEGF as well as functional VEGFR1 receptors thereby mediating both paracrine and autocrine VEGF-signaling. The aims of this work is i) to establish the direct influence of aflibercept (Zaltrap®) that inhibits all three VEGFR1 ligands (VEGF-A, VEGF-B and PlGF) on CRC cells, ii) to identify tumor phenotypes associated with resistance to aflibercept in vitro and, iii) to extend these findings to human xenograft models.

Methods

A panel of 12 well-characterized CRC cell lines was used to establish the influence of VEGFR1 stimulation (VEGF-A, VEGF-B, PlGF) and inhibition (aflibercept) on VEGF-mediated tumor cell migration. Expression of VEGF ligands and receptors was determined by qRT-PCR and ELISA assays. The in vivo influence of aflibercept was determined in human xenograft models and complemented by IHC and Western blot analysis.

Results

Aflibercept inhibited the migration of most CRC cells under both normoxia and hypoxia including the highly sensitive HCT-116 cells. In contrast, LS174T cells did not respond to either aflibercept or to purified VEGF ligands. These cells expressed high levels of VEGF ligands and HIF2alpha, even under normoxia. Accordingly, aflibercept showed pronounced in vivo activity toward HCT-116 xenografts with 75% tumor growth inhibition but only 40% tumor growth inhibition toward LS174T tumors, compared to the corresponding vehicle controls. A similar trend was observed for bevacizumab with 41% tumor growth inhibition for HCT-116 and 32% inhibition for LS174T xenografts. IHC analysis of LS174T xenografts confirmed the strong expression of HIF2alpha and VEGF ligands as well as a modest inhibitory effect on the tumor-associated endothelia cells.

Conclusions

We here report that aflibercept has direct antimigratory effects on most CRC cells. Strong expression of HIF-2alpha and VEGF ligands was accompanied by aflibercept resistance in vitro as in vivo.

Legal entity responsible for the study

INSERM U938 and Université Pierre et Marie Curie (UPMC), Sorbonne Universités, Paris, France

Funding

Sanofi-Genzyme

Disclosure

A.K. Larsen: This work was supported in part by Sanofi Genzyme. E. Dochy: Employed by Sanofi-Genzyme. A. de Gramont: This research is in part supported by Sanofi. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

122P - Cell-free (cf)DNA analysis identifies PIK3CA/AKT1 mutations associated with greater PFS improvement from the addition of ipatasertib (IPAT) to paclitaxel (P) in triple-negative breast cancer (TNBC) (ID 2329)

Presentation Number
122P
Lecture Time
13:15 - 13:15
Speakers
  • M. Wongchenko
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The oral Akt inhibitor IPAT is being evaluated in cancers with a high prevalence of PI3K/Akt pathway activation. In the placebo-controlled randomised phase II LOTUS trial (NCT02162719), adding IPAT to P as first-line therapy for metastatic TNBC improved PFS in unselected patients (stratified hazard ratio [HR] 0.60 [90% CI 0.40–0.91]), with a more pronounced effect in patients with PIK3CA/AKT1/PTEN-altered tumours [Dent, ASCO 2017]. cfDNA sequencing was performed to assess the utility of blood samples for detecting tumour mutations in TNBC.

Methods

Pre-treatment plasma and tumour samples were evaluated for genetic alterations using Foundation Medicine’s FoundationACT® and FoundationOne® next-generation sequencing assays, respectively. Samples from 88 patients were evaluable by cfDNA, 72 of whom also had evaluable tumour samples (52 primary tumour). To test the prognostic effect of cfDNA mutational burden, patients were classified as having high or low variant allele fraction (VAF) in cfDNA using the median as a cutoff.

Results

In 81 patients (92%), at least one mutation was detectable by cfDNA sequencing. Concordance with tissue sequencing was 75%. High VAF was associated with shorter PFS than low VAF in both the IPAT + P arm (HR 2.39 [90% CI 1.19–4.70]) and the placebo + P arm (HR 2.68 [90% CI 1.46–5.11]). High VAF was also associated with the presence of > 1 metastatic site but not tumour volume per RECIST v1.1. In 22 patients (25%), an activating PIK3CA (n = 16) or AKT1 (n = 6) mutation was detected in cfDNA; concordance with tissue sequencing was 100%. PFS improvement with the addition of IPAT to P was more pronounced in patients with detectable PIK3CA/AKT1 mutations (HR 0.15 [90% CI 0.03–0.50]) than in those without a detectable mutation (HR 0.82 [90% CI 0.50–1.31]).

Conclusions

These results highlight the potential role of cfDNA in evaluating patient prognosis as well as identifying genetic markers associated with improved treatment outcomes. Furthermore, they support the occurrence of PIK3CA and AKT1 mutations as early genetic events present in primary tissue samples that are maintained during metastasis.

Clinical trial identification

NCT02162719

Legal entity responsible for the study

F Hoffmann-La Roche Ltd.

Funding

F Hoffmann-La Roche Ltd

Disclosure

M. Wongchenko: Employee of Genentech, Inc. and holds shares in Roche and Ariad Pharmaceuticals. R. Dent: Honoraria for consultancy/advisory boards/speaker engagements from Pfizer, Roche, Eisai, Merck, and AstraZeneca. S-B. Kim: Research funding from Novartis, Sanofi-Aventis, Kyowa-Kirin Inc, and Dongkook Pharma Co., Ltd. C. Saura: Honoraria for consulting/advisory roles from Puma Biotechnology, Pfizer, and Roche and research funding (to her institution) from Genentech and AstraZeneca. M. Oliveira: Honoraria for consulting/advisory roles from Puma Biotechnology and Genentech/Roche and research funding (to the institution) from Genentech and AstraZeneca. J. Baselga: Compensation for a leadership role from Infinity Pharmaceuticals, stock or ownership interest in PMV Pharma, Juno Therapeutics, Infinity Pharmaceuticals, and GRAIL, and honoraria for consulting/advisory role for Eli Lilly, Novartis, and GRAIL. A.V. Kapp, W.Y. Chan, S.M. Singel, D.J. Maslyar, S. Gendreau: Employee of Genentech, Inc. and holds stock in Roche.

Collapse
Poster display session Poster Display session

123P - Genotyping circulating tumor DNA identifies metastatic colorectal cancer (mCRC) patients highly sensitive to Sym004 (ID 3619)

Presentation Number
123P
Lecture Time
13:15 - 13:15
Speakers
  • C. Montagut Viladot
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Acquired resistance of mCRC patients (pts) to anti-EGFR monoclonal antibodies (mAbs) is frequently due to mutations in RAS/BRAF and EGFR extracellular domain (ECD), and amplifications in MET/ERBB2. Studies suggest that some anti-EGFR mAb refractory pts retain tumor EGFR-dependency potentially targetable by more efficacious agents such as Sym004 – a mixture of two non-overlapping mAb targeting EGFR for degradation. A Phase 2b trial of Sym004 in 254 mCRC pts tissue RAS wt that relapsed on anti-EGFR blockade was recently completed.

Methods

Baseline circulating tumor (ct)DNA profiles (Guardant360, Guardant Health, N = 193 pts) were obtained from blood samples collected from pts in the Sym004 Phase2b trial. Serial blood samples during treatment were analyzed for EGFR-ECD mutation dynamics.

Results

High mutant allele frequency (MAF >20%) of KRAS/NRAS was found in 5% of pts and BRAF V600E and EGFR-ECD mutations in 6.7% and 25% of pts, respectively. At least 1 of these mechanisms of resistance was identified in 32% of the pts. Mutations in KRAS, NRAS, and BRAF V600E, and amplifications of ERBB2 and MET were more likely to co-occur with EGFR-ECD mutations, demonstrating genomic complexity developed as a result of EGFR blockade. Comparative analysis of MAFs of driver genes indicated that EGFR-ECD mutations are subclonal events. The absence of any EGFR-ECD/BRAF/high RAS MAF mutation at baseline was associated with a significantly improved OS in the Sym004 treated pts as compared to control arm with investigator’s choice chemotherapy. Sym004 has previously been demonstrated to retain in vitro efficacy in EGFR-ECD mutated cancer cells, and ctDNA monitoring demonstrated decrease in EGFR-ECD in Sym004 treated pts. This suggests that EGFR-ECD mutated cells are targeted by Sym004, but this activity does not translate into clinically meaningful OS benefit, likely due to other co-occuring resistance mechanisms.

Conclusions

Comprehensive liquid biomarker profiling of 193 mCRC pts captured high intrapatient heterogeneity following anti-EGFR therapy, and identified a highly Sym004 sensitive mCRC pts subset with a RAS/RAF/EGFR-ECD wild type profile. Our data indicates heterogeneous responses of different subclones to targeted agents in mCRC.

Legal entity responsible for the study

Medical Oncology Department, Hospital del Mar, Barcelona, Spain

Funding

Funded by Symphogen

Disclosure

C. Montagut Viladot: Advisory Board for Amgen, Bayer, Merck Serono, Sanofi, Symphogen. T. Tuxen Poulsen, M. Kragh, K. Koefoed, C. Ding, J. Clausell-Tormos, T. Lindsted, M.W. Pedersen, P. Nadler, I.D. Horak: Full time employee at Symphogen. S. Kopetz: Advisory boards for Amgen, Merrimack, Bayer, Sanofi, Array BioPharma, Genentech, MolecularMatch, Symphogen, Guardant Health, EMD Serono, Merck. J. Tabernero: Advisory boards for Amgen, Bayer, Boehringer Ingelheim, Celgene, Chugai, Genentech, Lilly, MSD, Merck Serono, Novartis, Pfizer, Roche, Sanofi, Symphogen, Taiho, and Takeda. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

124P - Role of circulating miRNAs as prognostic biomarkers in resected early-stage non-small cell lung cancer (ID 3439)

Presentation Number
124P
Lecture Time
13:15 - 13:15
Speakers
  • P. Ulivi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Non-small cell lung cancer (NSCLC) is the primary cause of cancer-related death, with a 5-year survival rate < 16% mainly because of disseminated disease, even in fully resected early-stage disease. Biomarkers identifying patients at a higher risk of relapse would be useful and circulating microRNAs (miRNAs) are a promising option in this setting.

Methods

We analyzed a case series of 182 patients with resected early stage (IA-IIIA) NSCLC (99 adenocarcinoma [ADC] and 83 squamous cell carcinoma [SCC]). Peripheral blood samples were collected from each patient before surgical resection and serum was obtained after centrifugation and stored at -80 °C until miRNA extraction. A panel of 84 circulating miRNAs was analyzed by Real Time PCR. Data were normalized using an external spike-in (cel-miR-39) and the mean of the two most stable endogenous housekeeping genes chosen separately for ADC and SCC samples. miRNA expression was analyzed in relation to disease-free survival (DFS) by the Cox regression model. Results are reported as hazard ratios (HRs) and 95% confidence intervals (CIs).

Results

In ADC patients, stage was significantly associated with DFS (HR stage II-IIIA vs stage I = 4.94, 95% CI [2.71 - 9.02]). Multiple statistical methods were used to evaluate miRNA expression data. In univariate analysis, two miRNAs (miR-222-3p and miR-22-3p) were significantly associated with DFS (p = 0.033 and p = 0.041, respectively). However, this significance was not maintained after adjusting for multiple testing. In SCC patients, disease stage was significantly related to DFS (HR stage II-IIIA vs stage I = 3.31, 95% CI [1.74 - 6.33]). Five miRNAs (let-7a-5p, miR-126-3p, miR-26a-5p, miR-130b-3p, miR-21-5p) were significantly associated with DFS even after adjusting for multiple testing (False Discovery Rate q-value <0.001).

Conclusions

Pre-surgery circulating levels of let-7a-5p, miR-126-3p, miR-26a-5p, miR-130b-3p and miR-21-5p would appear to be significantly correlated with prognosis in resected early-stage SCC.

Clinical trial identification

Not applicable

Legal entity responsible for the study

IRST-IRCCS

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

125P - Targeted methylation sequencing of plasma cell-free DNA identifies patients with advanced breast, colorectal, non-small cell lung cancer, melanoma with poor outcomes (ID 3339)

Presentation Number
125P
Lecture Time
13:15 - 13:15
Speakers
  • A. Jassowicz
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Plasma cell-free DNA (cfDNA) sampling from patients with advanced cancers offers a minimally invasive source of tumor DNA for molecular testing, including methylation profiling. Tumor sequencing at disease progression offers insights into cancer biology, but is often not done because of logistical and other challenges associated with tumor biopsies.

Methods

Plasma samples were collected from patients with advanced breast, colorectal, non-small cell lung cancer (NSCLC) and melanoma at the time of disease progression and/or on therapy. Up to 30 ng (median 18ng) of plasma cfDNA was tested with the pan-cancer methylation panel to target a total of 10,888 CpG sites using an Illumina HiSeq2500 sequencer to calculate methylation scores. Methylation scores were correlated with cancer types and clinical outcomes.

Results

Of 69 plasma cfDNA samples collected from patients (colorectal cancer, 28; NSCLC, 18, breast cancer, 12; melanoma, 11) at disease progression, methylation scores were consistent with the presence of cancer in 84% (58/69); while in 79% (46/58) of plasma cfDNA samples, methylation scores accurately classified the underlying cancer type. High methylation scores in plasma cfDNA samples collected at disease progression compared to low methylation scores were associated with shorter survival irrespective of the tumor type (3.9 vs 10.4 months, P < 0.001, confirmed on multivariable analysis). In addition, high methylation scores in plasma cfDNA samples collected at progression vs low methylation scores were associated with shorter time to treatment failure (TTF) on systemic therapy (1.6 vs. 2.8 months, P = 0.007). There was a positive correlation between RECIST response (%) to systemic therapy and methylation scores (r 0.32, P = 0.03). Methylations score before therapy (median 87.28) were higher than methylation scores on therapy (median 9.49, P = 0.001).

Conclusions

Methylation scores from plasma cfDNA collected at disease progression from patients with advanced cancers accurately classify underlying cancer types and are associated with survival, TTF and RECIST responses.

Clinical trial identification

MD Anderson Protocols laboratory protocols LAB10-0334 and PA13-0956

Legal entity responsible for the study

MD Anderson Cancer Center

Funding

Sidney Kimmel Foundation for Cancer Research (Filip Janku); Sheikh Khalifa Al Nahyan Ben Zayed Institute for Personalized Cancer Therapy (Filip Janku); National Center for Advancing Translational Sciences (grant no. UL1 TR000371); National Institutes of Health through MD Anderson’s Cancer Center Support Grant (P30 CA016672); Illumina.

Disclosure

L. Liu, J. Toung, R. Vijayaraghavan, R. Zhang, H. Kang: Employee and stock ownership Illumina, Inc. F. Janku: Scientific Advisory Boards: Illumina, Guardant Health; Paid consulting: Trovagene; Stocks: Trovagene. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

126P - Single-cell combined mutation and gene expression studies of circulating tumor-associated cells in non-small cell lung cancer (ID 4319)

Presentation Number
126P
Lecture Time
13:15 - 13:15
Speakers
  • J. Mong
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Circulating tumor cells (CTC) in the bloodstream not only provide us a sampling of the tumor population but are also keys to the metastatic process. Most CTC enrichment devices are coupled to downstream marker analysis by immunocytochemistry that define CTC as CD45-negative and epithelial marker (e.g. EPCAM)-positive cells. Our group recently reported a new class of tumor-derived cells in circulation, the circulating tumor-endothelial cluster, in colorectal cancer. This demonstrated that tumor-associated cells in circulation hitherto considered malignant may instead and unexpectedly provide key insights to the tumor microenvironment. To effectively classify the circulating cell milieu in lung cancer, we used a single-cell technique RNA and mutation analysis (scrm-PCR) we have described previously to characterize DNA and RNA from the same cell.

Methods

We enriched for CTC from whole blood of non-small cell lung cancer (NSCLC) patients using silicon microsieves with uniformly spaced 8 to 10uM circular pores. We manually selected captured single cells or cell clusters for characterization. 50 single cell or cell clusters from 5 NSCLC patients and 2 healthy subjects were probed for a curated panel of 27 RNA markers and hotspot DNA mutations in EGFR, TP53 and KRAS by scrm-PCR.

Results

Classic epithelial markers such as EPCAM are largely unexpressed in CD45-negative cells in NSCLC. DNA mutations were undetected in all isolated cells except for 1 cell. CD45-negative cell clusters in NSCLC exhibit a different phenotypic profile as compared to our report of tumor-endothelial in colorectal cancer, failing to express classic endothelial markers such as VWF, MCAM and CDH5. We identified important morphological features correlating with biological phenotypic classification of these cells, allowing us to establish a map of the circulating cell milieu of NSCLC.

Conclusions

This is the first systematic same-cell combined mutation and expression analysis of circulating cells in NSCLC patients. With this novel approach, we report the presence of predominantly tumor-associated cells rather than malignant cells in circulation, and highlight the potential of these cells as biomarkers of NSCLC and its tumor microenvironment.

Legal entity responsible for the study

Institute of Bioengineering and Nanotechnology

Funding

Agency for Science, Technology and Research (A*STAR)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

127P - EGFR copy number aberrations detected in cfDNA from advanced NSCLC patients (ID 5024)

Presentation Number
127P
Lecture Time
13:15 - 13:15
Speakers
  • G. O'Kane
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Copy number aberrations (CNA) in the epidermal growth factor receptor (EGFR) represent a mechanism of tyrosine kinase activation and oncogenic signaling in advanced non-small cell lung cancers (NSCLC). High copy number gains (CNG) may predict TKI sensitivity but when acquired are postulated to reflect resistance. Treatment with monoclonal antibodies may have a role and therefore CNA integration in molecular diagnostics is potentially important.

Methods

Cell free (cf) DNA was from extracted from NSCLC patients undergoing treatment with EFGR-TKIs. A validated Liquid Biopsy Sequencing (LB-Seq) method for hybrid capture followed by ultra deep sequencing (> 20,000X) evaluated coding exons of KRAS, NRAS, BRAF, PIK3CA, and EGFR (18 kb). Subsequent filtering of mutation calls using a novel algorithm enabled detection of tumor-derived fragments at concentrations down to 0.2%. EGFR CNAs were assessed using a modified version of the VisCap algorithm. Mutation calls were compared to tissue biospy results for EGFR mutations.

Results

Targeted sequencing has been performed on 12 samples to date: 10 patients with classical EGFR mutations in L858R and del19, 1 with an exon 20 insertion and 1 with an exon 18 G718X mutation. All patients had progressed on at least one EGFR-TKI. % mutant reads ranged from 0.25% to 33%. EGFR CNGs were detected in 3 patients: 1 patient with T790M+ve disease confirmed in both tissue and cfDNA was found to have a co-occurring BRAF mutation (p.33_34insGA). The remaining 2 patients’ tissue samples tested negative for T790M; T790M was detected in cfDNA in 1 patient, who is currently receiving osimertinib, and in the other patient rapid progression on gefitinib occurred with an EGFR-G719X mutation. Copy number losses were detected in 2 patients. 2 of 6 patients with EGFR-T790M positive tissue were confirmed in cfDNA, the remaining 4 negative results were verified by ddPCR. An intronic variant of unknown significance (c.747 + 9C>T), not covered in tissue testing, was also captured in cfDNA.

Conclusions

Copy number aberrations in EGFR can be detected from targeted sequencing of cfDNA in patients with EGFR mutated NSCLC. Longitudinal evaluation in patients receiving EGFR-TKIs may provide insights into mechanisms of resistance.

Legal entity responsible for the study

Natasha Leighl

Funding

None

Disclosure

N.B. Leighl: Research funding (institution): Novartis. Travel/honoraria: AstraZeneca, Merck Sharp Dohme, Pfizer, Bristol-Myers Squibb. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

128P - Detection of esophageal cancer patients using circulating serum microRNA from the result of comprehensive analysis (ID 1051)

Presentation Number
128P
Lecture Time
13:15 - 13:15
Speakers
  • K. Kato
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Recent studies have reported that serum microRNAs (miRNAs) are potentially useful biomarkers for diagnosis and treatment of cancer. However, its utility for detecting esophageal squamous cell carcinoma (ESCC) has not been investigated yet. The aims of this study are to identify circulating serum miRNAs for ESCC detection.

Methods

We comprehensively analyzed serum miRNA expression profiles of 595 patients with ESCC, and 5051 non-cancer individuals using microarray (3D-Gene, Toray). Serum of non-cancer individuals was obtained from Biobank of NCGG and Health check up clinic (Yokohama Minoru clinic). Serum of ESCC patients was collected before starting any treatment such as radiotherapy, surgery and chemotherapy. Analyzed samples were randomly divided into discovery and validation sets. Serum miRNA levels were compared between ESCC and non-ESCC patients. Fisher’s linear discriminant analysis was performed to construct the discriminant model for ESCC detection. Measured values of each miRNAs were extrapolated into the discriminant formulas. We performed ROC analysis to evaluate the diagnostic ability of these formulas in each validation cohort.

Results

In discovery set, 300 patients of ESCC was compared to 300 individuals of non-cancer control. We picked up 3 miRNAs, named miR-A, miR-B and miR-C, for ESCC diagnosis. Their AUC for detection of ESCC was 0.967, 0.873 and 0.650, respectively in ROC analysis. We constructed the discriminant model, called EC index, using these 3 miRNAs in discovery set. In validation set which contain 295 pts of ESCC and 4751 person of non-cancer control, EC index showed the AUC, sensitivity and specificity of the discriminant formula was 0.99, 0.98 and 0.95, respectively.

Conclusions

We identified novel serum miRNAs for ESCC detection. Our discriminant using these miRNAs can diagnose ESCC.

Clinical trial identification

NCCH2016-249

Legal entity responsible for the study

National Cancer Center

Funding

Japan Agency for Medical Research and Development

Disclosure

K. Kato: Research fund: ONO, MSD, Shionogi. J. Kawauchi, S. Takizawa: Employee of Tray company. T. Ochiya: Grant/Research funding from: Kyowa Medex, Kewpie Corporation, Takeda, Rohto Pharmaceutical Co., Ltd., Japan Atherosclerosis Research Foundation, Inter Stem, BioMimetics Sympathies. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

129P - First prospective multicenter real-world RAS mutation comparison between OncoBEAM-based liquid biopsy and tissue analysis (ID 2707)

Presentation Number
129P
Lecture Time
13:15 - 13:15
Speakers
  • J. Garcia Foncillas
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Liquid biopsy is a powerful tool to refine the management of cancer patients by offering a minimally-invasive alternative to tumor tissue testing and rapid evaluation of overall tumor burden mutational status. To support its clinical adoption, a rigorous real-world evaluation of this method in routine clinical practice is required. OncoBEAM RAS is the only liquid biopsy assay to attain CE-IVD status for plasma RAS mutation analysis in routine colorectal cancer (CRC) patient care. The goal of the present study was to evaluate the aggregate performance of OncoBEAM RAS in10 hospital labs where the technology is installed in Spain.

Methods

Blood samples were collected in Streck cell-free DNA BCT® or EDTA tubes from metastatic CRC patients and circulating cell-free DNA from plasma was examined for RAS mutations using the OncoBEAM platform at each hospital laboratory. Results were then compared to those obtained from DNA extracted from tissue from the same patient.

Results

The overall percentage agreement (concordance) of results from plasma and tissue RAS mutation testing of 230 patients was 90.4% (208/230); 95% CI = 0.86-0.94, with positive percent agreement of 86.9% (113/130) and negative percent agreement of 95.0% (95/100). Re-analysis of tissue from all discordant cases by BEAMing revealed 2 false negative local tumor tissue RAS results. Plasma false negative results were observed more frequently in patients presenting with peritoneal and/or lung metastases only. The prevalence of RAS mutations in plasma (51.3%) and tissue (56.5%) were in accord with the expected occurrence of RAS mutations in mCRC patients.

Conclusions

In this first prospective real-world RAS mutation performance comparison study across a network of hospital laboratories certified to perform OncoBEAM testing in routine clinical practice, a high overall agreement was observed between results obtained from plasma and tissue samples. These results are comparable to those obtained in retrospective studies. Overall, these findings indicate that plasma OncoBEAM RAS testing is a viable solution for rapid delivery of RAS mutation status to determine mCRC patient eligibility for anti-EGFR therapy.

Legal entity responsible for the study

University Hospital “Fundacion Jimenez Diaz”, Autonomous University of Madrid

Funding

Sysmex, Merck

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

130P - EGFR plasma testing in routine practice by real-time PCR in lung cancer patients: Experience of 262 patients (ID 2716)

Presentation Number
130P
Lecture Time
13:15 - 13:15
Speakers
  • P. Taniere
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

EGFR mutation testing in lung cancer prior to tyrosine kinase inhibitor (TKI) therapy can be performed in tissue or plasma in practice. Both techniques are complementary; however, plasma testing can represent a surrogate for re-sampling a tumour in patients progressing under TKI prior to prescription of osimertinib, but clinical sensitivity of the test remains to be determined. We present our twelve-month experience of integrating EGFR plasma testing into our service with a series of 262 cases. We currently receive over 50 cases per month.

Methods

EGFR mutation testing for both tissue and plasma is performed using cobas EGFR Mutation Test v2, which covers 29 deletions in exon 19, T790M, L858R, G719X, S768I, L861Q and 5 insertions in exon 20. For plasma testing, DNA is extracted using the cobas cfDNA sample preparation kit. All samples were submitted in Paxgene ccfDNA tubes.

Results

Of the 262 cases submitted for testing, five failed (1.9%). 123 mutations, including 42 T790M, were detected. Turnaround time was two days. Clinical sensitivity is very difficult to assess because of the uncertainty of the presence of circulating tumour DNA at all. All types of primary mutation were detected: 70 exon 19 deletions, 42 L858R, 2 G719X, 5 L861Q, 3 combined S768I and G719X, and 1 singlet T790M. Clinical details were not available for all patients. A sensitising mutation was found in 51 of 92 patients (55.5%) under TKI therapy, where this was indicated on the request form; among these, an associated T790M mutation was found in 19 (37%) of patients. Several patients underwent multiple tests while they received TKI therapy; in two patients, the secondary mutation was detected prior to clinical progression.

Conclusions

We show here that EGFR plasma testing is perfectly suitable for clinical practice; it is highly specific and cost-effective due to rapid turnaround times, but the low sensitivity renders it complementary to tissue testing rather than a true surrogate.

Clinical trial identification

N/A

Legal entity responsible for the study

N/A

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

131P - Circulating tumor cells as liquid biopsy for castration resistant prostate cancer patients treated with cabazitaxel (ID 3371)

Presentation Number
131P
Lecture Time
13:15 - 13:15
Speakers
  • S. Salvi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Cabazitaxel (CBZ) has been shown to improve overall survival (OS) in metastatic castration resistant prostate cancer (mCRPC) patients (pts) and to overcome resistance to docetaxel (DOC). Circulating tumor cells (CTCs) can be useful tool for precision medicine. CTCs expression profiling before CBZ treatment could establish novel predictive biomarkers.

Methods

We prospectively enrolled 28 pts with mCRPC treated with CBZ 25 mg/mq q21 after DOC and abiraterone or enzalutamide. CTCs enrichment was assessed with Adna Test EMT/STEM. Expression analyses of AKR1C3, AKT2, ALDH1, AR, ARV7, EPCAM, FOLH1, MDK, PARP, MRP1, PIK3CA, POU5F1, PSCA, TUBB3, VIM, ACT, HPRT1 were analyzed using real time PCR. CTCs positive pts were defined when at least one marker among AKT2, AR, AR-V7, EPCAM, FOLH1, PSCA, PIK3CA was expressed. Progressive disease was defined according to PCWG2 criteria. Main endpoint was the correlation between CTCs expression profiling and outcome.

Results

Of these 28 pts, 18 (64%) had detectable CTCs before the starting of CBZ and 10 (36%) had undetectable CTCs. Detection of CTCs was associated with poor OS. However, no difference was observed for progression free survival (PFS). No correlation between CTCs assessment and PSA response rate was found. In addition, we subdivided pts according to median value of CTCs expression markers. Nine (50%) pts with ≥3 markers had a significant worse PFS compared to pts with <3 markers. Pts with ≥3 markers, reflecting heterogeneity of disease, had also a poor OS (Table).

131P

CTCsPtsPFS
OS
Median (months)HR (95% CI)pMedian (months)HR (95% CI)p
CTCs +18/285.81.31 (0.58- 3.04)p=0.52209.77.03 (1.16- 11.00)p=0.0279
CTCs -10/287.0
≥3 markers9/182.53.59 (2.07- 22.01)p=0.00393.95.92 (3.86- 52.21)p=0.0003
<3 markers9/1810.116.6

Conclusions

We prospectively confirmed a prognostic role for CTCs in mCRPC pts treated with CBZ and we also firstly showed the utility to characterize CTCs expression markers thanks to its potential predictive role. The identification of markers expressed on CTCs may also provide additional therapeutic targets. More details on the prognostic impact of these biomarkers and AR status and mutations are under analysis and will be presented at meeting.

Clinical trial identification

IRSTB030

Legal entity responsible for the study

Ugo De Giorgi

Funding

Sanofi

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

132P - Biomarker analysis using circulating tumor DNA in patients treated with sorafenib for advanced hepatocellular carcinoma (ID 4380)

Presentation Number
132P
Lecture Time
13:15 - 13:15
Speakers
  • S. Park
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

We aimed to investigate potential biomarkers in patients treated with sorafenib for advanced hepatocellular carcinoma (HCC) using circulating tumour DNA (ctDNA).

Methods

155 patients who had started sorafenib between March 2014 and November 2016 were identified from a prospective biomarker cohort of Asan Medical Center, Korea. We quantified the concentration of ctDNA extracted from blood samples of each patient collected before sorafenib treatment and measured the copy numbers of vascular endothelial growth factor-A (VEGFA) in ctDNA. We also applied low depth whole genome sequencing from ctDNA to find copy number aberrations in HCC and employed Q-score, defined as a standard deviation regarding Z-scores of sequenced reads on each chromosome.

Results

Among 155 patients, 124 were finally included in the analysis. 82 patients achieved partial response, stable disease or non-CR/non-PD with sorafenib treatment (non-PD group) whereas 42 exhibited progressive disease (PD group). The PD group had significantly higher levels of ctDNA concentrations than the non-PD group (153.3 vs. 109.3 ng/mL; p = 0.038). Q-score of PD group was also higher than that of non-PD group but there was a borderline significant difference between two groups (6.10 vs. 3.80; p = 0.058). VEGFA copy number, which was available for only 41 patients, did not differ between PD (n = 16) and non-PD (n = 25) groups (2.56 vs. 2.48; p = 0.467). Divided into two groups based on the median value (119.7 ng/mL) of ctDNA concentrations, patients with high ctDNA had significantly shorter time to progression (TTP) (median, 2.3 vs. 4.1 months; p = 0.025) and overall survival (OS) (median, 4.5 vs. 14.8 months; p < 0.001) than those with low ctDNA. Similarly, patients with higher Q-score than median value of 4.12 had significantly worse TTP (median, 2.7 vs. 4.0 months; p = 0.012) and OS (median, 5.2 vs. 17.3 months; p < 0.001) compared to those with lower Q-score. After adjusting confounding factors by multivariate Cox regression analysis, the concentration of ctDNA and Q-score remained independent prognostic factors associated with both TTP (p = 0.026 and 0.042, respectively) and OS (p < 0.001 and p = 0.001, respectively).

Conclusions

Our results showed that ctDNA level and copy number aberrations represented by Q-score could be potential prognostic biomarkers in HCC patients treated with sorafenib.

Legal entity responsible for the study

Department of Oncology, Asan Medical Center, Seoul, Republic of Korea

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

133P - Heparinase enables reliable quantification of circulating tumor DNA from heparin plasma samples by droplet digital PCR (ID 4938)

Presentation Number
133P
Lecture Time
13:15 - 13:15
Speakers
  • D. Sefrioui
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Circulating tumor DNA has been highlighted as a potential “liquid biopsy”, which can be used to identify prognostic and predictive alterations in oncology. Heparin is often used as plasma anticoagulant source for tumor marker analysis such as CA19.9 or CA15.3 but corresponds also to an inhibitory of PCR not enabling ctDNA detection. We aimed to evaluate the impact of heparinase addition on heparinized plasma samples to recovery the possibility of ctDNA analysis on samples initially dedicated for tumor markers analysis.

Methods

Plasma samples were collected in heparinized (n = 194) and EDTA (n = 8) tubes from hormone receptor-positive metastatic breast cancer (HR+MBC) patients resistant to aromatase inhibitor (AI) treatment (n = 144) and from newly diagnosed pancreatic adenocarcinoma (PA) patients (n = 50). ESR1 and KRAS mutations were used as targets for ctDNA detection in HR+MBC and PA patients, respectively. ctDNA was detected by droplet digital PCR after an amplification step either without or with heparinase (H- and H+ respectively). PCR efficiency and ctDNA detection rate were compared between H- and H+ subgroups as well as with EDTA subgroup.

Results

Heparinase addition improved significantly PCR efficiency for 91/144 HR+MBC and 26/50 PA patients enabling ctDNA detection in 22/91 (24%) and 13/26 (50%) of these patients. Moreover, heparinase condition did not quantitatively and qualitatively alter the ctDNA detection for patients without heparin inhibition of PCR and comparable results for ctDNA detection were obtained between H+ and EDTA subgroups.

Conclusions

Heparinase addition allows removing the heparin inhibition on cfDNA amplification and to detect and quantify accurately ctDNA levels by dPCR in the heparinized plasma samples.

Legal entity responsible for the study

Rouen University Hospital

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

134P - Circulating tumour DNA (ctDNA) in the clinical management of patients (pts) with advanced non-small cell lung cancer (NSCLC): A single centre experience (ID 1742)

Presentation Number
134P
Lecture Time
13:15 - 13:15
Speakers
  • M. Uccello
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Circulating tumour DNA (ctDNA) is an emerging non-invasive method to guide personalised medicine in patients (pts) with advanced non-small cell lung cancer (NSCLC). The aim of this project was to assess the utility of ctDNA in routine clinical practice where tumour biopsy (Bx) was contraindicated or insufficient for genotyping.

Methods

A 73-gene cancer panel test using next generation sequencing for ctDNA profiling (Guardant360®) was offered at our institution to stage IV NSCLC pts. The reports obtained were discussed in a multidisciplinary Genomics Review Board (GRB) to assess potentially actionable genomic alterations and enrolment into clinical trials. All pts consented for prospective collection of demographic, clinical, and biomolecular data.

Results

Thirty advanced lung cancer pts were offered ctDNA testing. Histology included adeno- in 27 (90%) pts, squamous cell- in 2 (7%) pts, and small cell carcinoma in 1 (3%) pt. As assessed by baseline tumour Bx, EGFR status was mutant in 15 (50%) pts, wild type in 8 (27%) pts, and unknown in 7 (23%) pts. Activating EGFR mutations (mut) showed on original Bx at diagnosis were detected by ctDNA in 11 (73%) pts – lag-time 17 months (range 7-94). Primary EGFR mut were found in 2 pts with unknown EGFR status, allowing access to EGFR tyrosine kinase inhibitors (TKIs). Acquired T790M mut was found in 3 (20%) pts progressing on prior EGFR TKI, with the indication to receive Osimertinib. Only 1 pt with ctDNA-positive status for T790M mut received concomitant solid Bx which was negative for T790M mut. Other genomic alterations were detected in the overall population, more commonly involving TP53 (n = 12; 40%), NF1 (n = 7; 23%), MET (n = 5; 17%), BRAF (n = 4; 13%), PIK3CA (n = 3; 10%), BRCA 1/2 (n = 3; 10%), PDGFRA (n = 3; 10%), and AR (n = 3; 10%) genes. Critical review in the GRB was crucial in assessing genomic alterations of clinical significance.

Conclusions

Our study confirms the utility of ctDNA testing in advanced NSCLC pts where tumour Bx was not possible or insufficient for genotyping. Based on ctDNA, 5 pts received treatment with 1st- or 2nd-line EGFR TKIs while several pts were considered for clinical trial participation. Updated results of this ongoing study will be reported.

Legal entity responsible for the study

Sarah Cannon Research Institute UK

Funding

Guardant Health

Disclosure

R. Lanman, I. Faull: Affiliated to Guardant Health. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

135P - Genome-wide methylation analysis reveals a prognostic classifier for non-metastatic colorectal cancer (ProMCol) (ID 2040)

Presentation Number
135P
Lecture Time
13:15 - 13:15
Speakers
  • M. Gündert
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Currently, pathological staging according to the tumor-node-metastasis system remains the gold standard for the prediction of patient survival in colorectal cancer (CRC) but this classification system provides insufficient information and therefore additional prognostic markers are needed.

Methods

A genome-wide methylation analysis was done for two independent cohorts of non-metastatic CRC patients (screening cohort n = 578 and validation cohort n = 308). Initially, genome-wide differentially methylated CpG sites between 34 pairs of tumor and normal mucosa tissue samples from the same patients were identified. A variable screening for prognostic CpG sites was performed in the screening cohort using marginal testing based on the Cox model and subsequent adjustment of the p-values via independent hypothesis weighting (IHW) using the difference between tumor and normal mucosa tissue as auxiliary covariate. From the 1000 CpG sites with the smallest adjusted p-value, the 20 CpG sites with the smallest Brier Score for 3-year overall survival (in the screening cohort) were selected. Applying principal component analysis on these CpG sites, we derived a methylation-based classifier for the prognosis of non-metastatic CRC (ProMCol).

Results

The ProMCol classifier was independently validated in the validation cohort, where it showed a significant reduction in the Brier score. Regarding the three year survival, the prediction error was reduced from 0.132 (calculated only with clinical variables), to 0.124 (combination of clinical variables with ProMCol classifier). An additional replication analysis showed that the ProMCol classifier was significantly associated with overall survival (OS) of non-metastatic CRC patients in the screening (HR = 0.22, 95%CI=0.13-0.35, p=6.2E-10) and the validation cohort (HR = 0.40, 95%CI=0.22-0.74, p=0.003), adjusted for standard clinical factors. Patients with a high methylation status, represented by higher values of the ProMCol classifier, showed a better prognosis for OS than patients with a low methylation status and lower ProMCol classifier values.

Conclusions

The usage of the ProMCol classifier could improve the prognostic accuracy for non-metastatic CRC patients.

Legal entity responsible for the study

Barbara Burwinkel, German Cancer Research Center

Funding

German Research Council, German Federal Ministry of Education and Research

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

136P - Epigenetic biomarkers in breast cancer: Preliminary results from H3K27m3 assessment in endocrine-treatment resistance (ID 5456)

Presentation Number
136P
Lecture Time
13:15 - 13:15
Speakers
  • M. Fontes e Sousa
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Breast cancer (BC) is an important cause of morbidity and mortality. Most BC are hormone-receptor positive and can be treated with endocrine treatment (ET), until resistance or toxicity is developed. Specifically, H3K27m3, an epigenetic marker of gene repression, has been associated with prognosis in ET resistant BC. Herein, we aim to further understand its potential role as predictive marker of ET resistance before exposure to treatment.

Methods

A cohort of BC patients diagnosed between 1995 and 2002 at our institution were enrolled after informed consent. Expression of H3K27me3 was determined by immunohistochemistry (IHC) in formalin fixed paraffin embedded tissues. GenASIsTM software was used to assess cell-positivity using a custom profile from positive control. Pre-specified conditions: ≥ 5 frames ≥ 3000 cells analyzed/case. SPSS v24 was used for statistical purposes.

Results

A total of 102 cases were assessed for H3K27me3 immunoexpression. Median of 6 frames/case (range 5-10) and 3311 cells/case (range 3017-5292) were obtained. Of the total cases analyzed, a median of 80% of cells showed positivity (range 9-100%). Using a negative/positive 50% cut-off, 81% were considered positive (pos.). The analyzed cohort displayed a median age of 60 years (33-82 years), 89% were classified as ductal carcinoma (DC) and 38% were grade 3. Concerning IHC subtyping, 43% classified as Luminal A-like, whereas 57% Luminal B-like. Around 66% of the cases evaluated for H3K27me3 expression were treated with adjuvant Tamoxifen exclusively. BC cases with H3K27me3 50-60% positivity were associated with higher cancer-related death (CI 95% 1.10-34.38, p = 0.05), although not reaching significance. Grade 3 BC significantly associated with increased risk of death (p = 0.024). An intermediate H3K27me3 expression (60-70% pos.) was observed in non-DC BC (CI 95% 1.42-13.92, p = 0.04).

Conclusions

In this preliminary retrospective cohort, H3K27m3 positivity was not found to be a pre-treatment endocrine resistance biomarker. However, statistical trends were observed between H3K27m3 expression and increased cancer-related death risk. Thus, further studies with extended cohort of patients are warranted.

Legal entity responsible for the study

Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

137P - Bioinformatic estimate of biomarker-positive populations in genomics-driven trials using precision trial designer (PTD) (ID 3767)

Presentation Number
137P
Lecture Time
13:15 - 13:15
Speakers
  • L. Mazzarella
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Trials that prospectively accrue based on genetics are powerful methods to test hypotheses in precision oncology, but are often difficult to conduct due to the rarity of biomarker-positive cases. This is mitigated in umbrella trials that maximize accrual by testing many hypotheses. However, multiple treatment arms frequently generate conflicting treatment allocations due to simultaneous actionable mutations; the allocation algorithm should be planned based on relative mutation frequencies to accurately estimate sample size and dropout rates. Large sequencing projects like the TCGA could be exploited but proper tools are lacking.

Methods

We developed Precision Trial Designer (PTD), a bioinformatic tool that simulates genomically-defined cohorts by iteratively sampling patient data from mutation databases (e.g. TCGA) and provides essential parameters to plan biomarker-driven trials using survival or response-rate endpoints. To show its potential, we simulated a 10-arm imaginary trial on multiple cancers, based on the Molecular Analyses for Personalized medicine (MAP) consensus. We then validated our approach by comparing simulated and real data from the SHIVA01 clinical trial.

Results

In the MAP trial, PTD predicted ≥1 actionable alteration in 73% patients and 32% conflicts. To adequately power each arm, we found the optimal rule that maximizes accrual (ALK inhibitors first, AKT inhibitors last) and propose various designs. In the SHIVA01 simulation, combinatorial point mutations were correctly predicted (18.9%, 95%CI 15.7-22.1 simulated vs 15.3%, 11.8-18.7 real), whereas PTD slightly overestimated copy number alterations (36.3%, 31.6-41 simulated vs 25.6%, 21.4-29.7 real), a predictable gap due to different detection techniques. Overall, 50.8% (46.2-55.7) cases were predicted as biomarker-positive, vs 37% (32.5-41.7) real. The relative contribution of each pathway of treatment (RAS/MEK, PI3K/MTOR) was conserved (47.8%, 41.7-53.9 and 25.8%, 19.9-31.5 simulated vs 53.1%, 95%CI 48.3-57.9 and 31.6%, 27.2-36.1 real, respectively).

Conclusions

PTD predicts combinatorial mutation frequencies with acceptable approximation and overcomes pressing issues in designing precision trials.

Legal entity responsible for the study

Luca Mazzarella

Funding

This research was funded by an AIRC Investigator Grant to G Curigliano and by grant ANR-10-EQPX-03 from the Agence Nationale de le Recherche (Investissements d’avenir) and SiRIC (Site de Recherche Intégré contre le Cancer).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

138P - Detection of microsatellite instability (MSI) in colorectal cancer samples with the automated Idylla MSI Test (ID 4028)

Presentation Number
138P
Lecture Time
13:15 - 13:15
Speakers
  • G. Maertens
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Detection of microsatellite instability (MSI) is recommended for all colorectal cancer (CRC) patients. Current clinical reference methods to detect MSI stain for mismatch repair proteins or analyze frequently mutated DNA repeat regions. The Idylla MSI Test is being developed using a unique set of novel biomarkers (Zhao et al. 2014; eLife) capable of faster detection with greater specificity and selectivity compared to current methods.

Methods

To assess the suitability of the novel marker set to detect MSI status in CRC, we profiled 8 or more markers in 870 CRC samples. Several clinical sites and different ethnic groups (Afro American, Caucasian, East-Asian, Hispanic and Indian) were included to assess robustness of marker selection. Repeat length was determined on FFPE DNA by PCR followed by melting curve analysis. Two-hundred and one samples were additionally screened with a reference methodology for MSI detection (Promega MSI analysis system).

Results

Hundred fifty-three samples (17.6%) were classified as MSI-H and 693 samples (79.7%) as MSS with the novel set of biomarkers, while 24 samples (2.8%) could not be classified. Concordance analysis was performed on 201 samples. The overall percent agreement with results available for both methods (173/201) was 93.6%. 11/173 (6.4%) were scored MSI-H for Idylla and MSS for the reference method; conversely no MSI-H cases for the reference method and MSS for Idylla were detected. 24/201 (11.9%) samples failed with the reference method, even after repeat testing, while only 8/201 (4.0%) samples failed with the Idylla methodology.

Conclusions

This study on a diverse set of CRC samples successfully demonstrated the validity of the novel MSI biomarkers to discriminate between MSI-H and MSS status. The Idylla MSI Test is currently under development on the most performant markers. Compatibility with the fully integrated Idylla platform will allow providing accurate and reliable results, with actionable results generated within 150 minutes from just one tumor FFPE slice (no reference sample required).

Legal entity responsible for the study

Biocartis NV

Funding

None

Disclosure

G. Maertens, B. De Craene, E. Rondelez, K. Peeters, L. Vandenbroeck, L. Van Puymbroeck, G. Vandercruyssen, E. Sablon: Biocartis Employee.

Collapse
Poster display session Poster Display session

139P - Gene expression BIRC5, Erb-b2/Her2-neu, ESR1, PGR1, MMP11, MDR1, MRP1, MXR at the CTCs in the primary breast cancer (ID 4940)

Presentation Number
139P
Lecture Time
13:15 - 13:15
Speakers
  • Y. Shliakhtunou
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Distant metastases are the main cause of death of patients with breast cancer. The substrate for the development of metastasis are the circulating tumor cells (CTCs). Determination of the expression of tumor-specific genes gives a more complete picture of the course of the tumor process.

Methods

Using PCR technology in real-time, gene expression studied BIRC5, Erb-b2/Her2-neu, ESR1, PGR1, MMP11, MDR1, MRP1, MXR at the CTCs in the surgical and adjuvant treatment of 56 cases primary non-metastatic breast cancer.

Results

In 33 (59%) prior to surgery functionally active in the CTCs-enriched peripheral blood, samples expressing the targeted range of different genes have been found. Simultaneous expression of targeted genes detected in 15 (26.8%). The highest levels of expression of the genes identified for BIRC5 1.2013 ± 0.193965 (min – 0.0017; max – 10.7083) and-Erb-b2/Her2-neu 1.6886 ± 0.0939 (min – 0.1032; max – 17.4401). Expression of ESR1, PGR1 were determined in 23 patients (42.5%). Expression family of ABC transporters detected in 14 (25%). After the operation was observed decrease in the number of CTCs and including functionally active to 13 cases (23.2%), but the CTCs data determined high levels of gene expression BIRC5, c-Erb-b2/Her2-neu, and MXR. During adjuvant, treatment with anthracyclines observed decrease in the level of gene expression BIRC5, ESR1, PGR1 at the CTCs in 7 patients. However, in 6 samples marked increase in the level of gene expression MDR1, MRP1. In the course of a taxane therapy in combination with trastuzumab in a group of patients of the 14 people there was a decrease, and the complete disappearance of the expression of a-Erb-b2/Her2-neu in 9 people, but in 5 patients there was a significant increase in the level of expression of targeted genes-Erb-b2/Her2-neu, BIRC5 and MXR. Expression of the gene in MMR11 CTCs-positive samples was determined at a high level, but significant reduction in the course of chemotherapy has not been determined.

Conclusions

The data indicate a high diagnostic, prognostic and predictive value of determining the functional activity of the CTCs on the basis of determining the dynamics of gene expression of tumor progression at the stage of planning treatment of breast cancer.

Legal entity responsible for the study

Educational Establishment “Vitebsk State Order of Peoples’ Friendship Medical University”

Funding

Belarusian Republican Foundation for Fundamental Research

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

140P - Metabolic syndrome and inflammation in castration resistant prostate cancer (CRPC) patients (pts) treated with abiraterone (abi) and enzalutamide (enza) (ID 2879)

Presentation Number
140P
Lecture Time
13:15 - 13:15
Speakers
  • V. Conteduca
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Metabolic syndrome (MS) and inflammation (INF) alterations are hallmarks of cancer progression. The study aimed to firstly assess the relationship between MS and INF and its impact on progression-free/overall survival (PFS/OS) in CRPC pts.

Methods

We retrospectively evaluated CRPC pts treated with abi and enza in 7 Italian Institutes between March 2011 and October 2016. MS was defined by modified Adult Treatment Panel III criteria, and INF characterized by the presence of at least 1 of these criteria: NLR≥3, elevated VES or C-reactive protein (CRP) levels.

Results

Eighty-three of 551 pts evaluated (15%) met MS criteria at baseline, whereas for 40 (8.5%) this occurred during treatment. No significant difference of MS incidence and cardiometabolic toxicities were reported between abi or enza. Pts with MS (MS+) showed a greater INF profile compared to MS- (79% vs 28%, p < 0.0001). We observed NLR≥3 in 70.2% of MS+ vs 38% of MS-, p < 0.0001, and median values of VES and CRP higher than upper normal limit (UNL) in MS+ vs MS- (53 vs 15, p < 0.0001, UNL<30 and 15 vs 3.3, p < 0.0001, UNL<5mg/L, respectively). Median PFS and OS were associated with baseline MS and INF and their combination defined clinically high- and low-risk prognostic groups (MS+/INF+ vs MS-/INF-) (Table). Multivariate analysis confirmed that MS was independently associated with PFS (HR = 2.07, 95%1.03-4.18 p = 0.041) and OS (HR = 4.87, 95% CI 2.36-10.03, p < 0.0001).

140P Univariate analyses of progression-free survival (PFS) and overall survival (OS) according to Metabolic Syndrome (MS) and Inflammation (INF) status

N. events/ total atientMedian PFS (months) (95% CI)pHR (95% CI)pN. events/ total atientMedian OS (months) (95% CI)pHR (95% CI)p
Overall407/5517.4 (6.4-7.8)308/55117.6 (15.5-19.0)
Baseline MS
No337/4688.3 (7.4-9.2)1.00247/46819.0 (17.4-21.6)1.00
Yes70/833.7 (3.5-4.1)<0.00012.77 (2.12-3.61)<0.000161/836.9 (5.4-9.8)<0.00013.43 (2.56-4.58)<0.0001
Baseline INF
No179/2318.5 (7.4-9.2)1.00141/23118.8 (15.9-21.8)1.00
Yes98/1334.5 (3.7-5.9)0.0021.48 (1.15-1.90)0.00282/13311.2 (8.4-16.8)0.00031.66 (1.26-2.18)0.0003
Combination MS+INF
MS- & INF-167/2189.0 (7.6-9.2)1.00130/21820.4 (16.5-24.0)1.00
MS- & INF+60/856.0 (3.9-9.8)1.25 (0.93-1.68)46/8518.0 (11.1-22.4)1.25 (0.89-1.76)
MS+ & INF-12/133.5 (1.8-4.7)3.77 (2.07-6.84)11/136.5 (3.0-11.2)3.80 (2.04-7.09)
MS+ & INF+38/483.7 (2.9-4.0)<0.00012.70 (1.88-3.89)<0.000136/486.3 (4.7-8.6)<0.00014.04 (2.75-5.93)<0.0001

Conclusions

The presence of MS is strongly associated with INF. Pre-treatment identification of MS and INF alterations may represent an available and easy to perform tool for a better prognostication of CRPC pts treated with abi or enza. A prospective evaluation is warranted.

Clinical trial identification

None

Legal entity responsible for the study

Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) Srl – IRCCS

Funding

None

Disclosure

V. Conteduca, U. De Giorgi: Speaker honoraria or travel support from Astellas, Janssen-Cilag and Sanofi- Aventis. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

141P - Ischemic stroke and cancer correlation: A stroke unit experience (ID 2731)

Presentation Number
141P
Lecture Time
13:15 - 13:15
Speakers
  • M. Rodrigues
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Cancer (Cr) and ischemic stroke (IS) are common causes of death in high-income regions. Cr patients present a higher probability of developing thromboembolic events, particularly IS. Measurable objective parameters may be helpful stating a neoplastic etiology: High D-dimer (DD) levels; C-reactive protein (CRP); Neutrophil–lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) express the severity of inflammation relating it to etiology and prognosis. Single centre retrospective study analyses patients with IS and active solid Cr (NG) with purpose of identifying clinical, laboratorial and imagological features that differentiate this from a control group (CG) without Cr.

Methods

Patients with IS admitted in a Stroke Unit from 01/2009 to 12/2014. Active cancer identified in clinical process. Transient ischemia, haemorrhagic strokes and other diagnosis excluded. For CG an age and gender matching patient was chosen. Clinical, analytical and imagological features were compared between groups. Statistical analysis using the SPSS Statistics V22.

Results

Out of 603 consecutive patients with IS, 48 (7.9%) had active solid Cr, 16 diagnosed during diagnostic work-up for IS, 24 before and 8 in the year after. Male predominance: 30 patients (62,5%). 14: metastatic disease. Most frequent Cr diagnosed were prostate and bladder (12,5% each); colon(10%) and lung(8,3%). Cardioembolism was the main subgroup in both (26 NG; 24 CG). Imagiologic pattern was similar in both groups. NG had increased laboratorial values compared to CG: LDH 602 vs 501 U/L (p 0,04); CPR 3,2 vs 1,8 mg/dL (p 0,11) and DD 0,85 vs 0,7ug/mL (p 0,28). Average NLR and PLR: 5,4 and 161,6 NG vs 2,34 and 104,1 CG (p < 0,01 and 0,03 respectively). NG and CG average NIHSS was 6,3 vs 4,9 (p 0,18) and mRs was 2,75 vs 1,95 (p 0.019) respectively. Higher NLR and PLR were associated with worse outcomes (higher NIHSS and mRs score).

Conclusions

Although Cr is a rare cause of stroke, it should not be devalued. There are several clinical, laboratory and imagiological signs of underlying neoplastic disease that should be considered, since they may promote an earlier diagnosis and approach to this pathology. Future prospective studies should be started in order to validate some of this parameters as indicators of subjacent neoplastic disease.

Legal entity responsible for the study

Mariana Rodrigues

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

142P - longitudinal exploratory analysis of prior- surgery everolimus circulating and clinical biomarkers in locally advanced and metastatic renal cell carcinoma patients (RCC) (ID 5526)

Presentation Number
142P
Lecture Time
13:15 - 13:15
Speakers
  • R. Elaidi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

If many drugs are available in RCC, we lack predictive biomarkers of disease recurrence or progression for personalized treatment. Circulating biomarkers (CB) are attractive candidates for disease monitoring. Longitudinal assessment of CB was performed in the NEORAD clinical trial (NCT01715935).

Methods

Locally advanced (LA) and metastatic (M) patients with clear cell or papillary RCC received 6 weeks prior-nephrectomy everolimus (Ev). CB assessed were: angiogenesis factors: VEGF-A, VEGFR-1&2, bFGF, SDF1, PlGF; CEC; CTC; HSC; immune cells: CD45, CD14+VEGFR-1/Tie2, Treg, IL-6. Clinical/histological (CH): ECOG-PS, tumor burden (TB), % necrosis, % sarcomatoid. Exploratory analysis (EA): CB and CH (prior to Ev, D22, D42, 4 weeks post-nephrectomy (+ 4 weeks if Ev resumed in M pts) in PD vs non-PD pts used Bayesian Model Averaging (BMA) and regularized Cox regression (LASSO). Three modeling strategies were compared for robustness.

Results

25 patients were included: LA = 14, M = 11. In LA and M cohorts, respectively 2 and 9 pts exhibited progression during the 12m post-surgery follow-up period. Most important predictors upon EA (by order): tumor burden, VEGF-A, LA/M, VEGFR-2, % necrosis, IL-6, VEGFR-1, age, PlGF. Higher TB at baseline and % necrosis were associated with increased risk of 6m post-nephrectomy progression. TB, IL-6, VEGF-A, VEGFR-1&2 exhibited nonlinear relationships suggesting complex underlying pathophysiological mechanisms involved in response to Ev, and are currently explored. Reduced VEGFR-2 at D42 was associated with worse PFS in both cohorts. Upon BMA and LASSO, % necrosis and TB were among the most retrieved predictors prior to nephrectomy, whereas CD45 and CD34 + 45-146+ at 4 weeks post-nephrectomy were the best CB predictors. No significant change in monocyte populations and in Treg was observed.

Conclusions

VEGF related CB, TB and % necrosis were the best candidates to discriminate PD- vs non-PD in LA and M pts. Extensive analysis of data collected using robust non-linear models could contribute to improve our understanding of mechanisms involved and suggest predictive biomarkers to be validated.

Clinical trial identification

NCT01715935

Legal entity responsible for the study

ARTIC

Funding

Novartis

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

143P - A comparison of treatment recommendations by molecular tumor boards worldwide (ID 3830)

Presentation Number
143P
Lecture Time
13:15 - 13:15
Speakers
  • D. Rieke
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Precision oncology holds promise to improve patient outcome. Yet, a benefit of personalized therapy in comparison to standard therapy has not been shown in prospective trials. We asked molecular tumor boards (MTB) worldwide to make treatment recommendations for sample patients to assess standards and differences in the precision oncology approach.

Methods

4 fictional patients with various degrees of genomic information (mutation, gene expression, copy number and gene fusion data, somatic and germline events) were created. A questionnaire was designed to identify methods and structures of the respective molecular tumor board’s recommendation process. 29 molecular tumor boards from 9 countries were identified and asked to participate in the survey between August 2016 and March 2017. A qualitative interpretation of the results was performed.

Results

5 MTBs from 4 countries completed the questionnaire and provided therapy recommendations. An identical treatment recommendation by all 5 MTB was not made for any one of the patients. In only one patient an overlapping treatment recommendation was made by three MTBs. Heterogeneity was larger for patients with more complex genomic information. The availability of clinical trials did not influence the recommendation heterogeneity. The setup of MTBs showed similarities in participating specialties (e.g. medical oncology, pathology, molecular biology), duration of discussion, testing methods and number of discussed patients. Differences were seen in the interpretation process. Further differences were identified in the interpretation of germline aberrations and interpretation of variant allele frequency. Comments by MTBs helped to identify minimum reporting standards for genetic testing.

Conclusions

Several differences in treatment recommendations were observed. In cases with obvious recommendations there was higher concordance among assessments. However, differences in treatment recommendations were observed in complex cases, and such heterogeneity contributes to the complexity of precision oncology. Larger studies are necessary for rational and stepwise development of principles and practice of MTB procedures.

Legal entity responsible for the study

Damian Rieke

Funding

None

Disclosure

U. Keilholz: Speaker honoraria and advisory board roles are with AstraZeneca, Bristol-Myers Squibb, Merck, MSD, Pfizer, Novartis, Innate. Corporate-sponsored research with AstraZeneca and Merck. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

144P - Identification of germline mutation using 30-gene sequencing and clinical characteristic of Chinese with hereditary breast cancer (ID 4293)

Presentation Number
144P
Lecture Time
13:15 - 13:15
Speakers
  • A. Kwong
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

With the recent discovery of other breast cancer susceptibility genes (e.g. CDH1, ATM, CHEK2, PALB2, RAD50), molecular diagnosis of hereditary breast and ovarian cancers (HBOC) using multigene panels could help to identify other moderate/low penetrance genes in patients who tested negative for BRCA mutation. However, the clinical management of these cancer predisposition genes have not been clearly defined, therefore only BRCA1 and BRCA2 are routinely included in the genetic screening. In view of the differences in the mutation spectrum across ethnicity, it is important to identify other HBOC genes to estimate the associated breast cancer risk in Chinese.

Methods

High-risk breast cancer patients who were negative for BRCA1, BRCA2, TP53 and PTEN were selected from the Hong Kong Hereditary Breast Cancer Family Registry between 2007-2016. In the study, 745 patients were subjected to 30-gene panel by next-generation sequencing (Color Genomics). All detected pathogenic mutations were further validated by bi-directional DNA sequencing. The sequencing data was co-analyzed by our in-house developed bioinformatics pipeline.

Results

Thirty-five pathogenic variants were identified in this series (4.7%), which correspond to 11 different cancer predisposition genes. Majority of the carriers (74.29%) had early-onset of breast cancer (age <45), 42.86% had ³ 2 family members with cancer and 17.14% were triple-negative. The most common mutated genes were PALB2 (1.21%), RAD51D (0.94%) and ATM (0.67%). However, the cancer risk of RAD51D in breast cancer warrants further investigation. Moreover, over 28% of patients had a variant of unknown significance (VUS) in these genes (excluding BRCA1, BRCA2, TP53 and PTEN), which account for 183 types of VUS. Data from large-cohort studies and international consortiums will help to define the pathogenicity and clinical interpretation/management of the variants.

Conclusions

Our findings suggested that detection of PALB2 should be included in the genetic test panel in Chinese with breast cancer. Multigene panel testing is an efficient tool in the diagnosis of HBOC, this could help patients to understand the cancer risk and aid the development of effective treatments.

Legal entity responsible for the study

Ava Kwong

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

145P - Selecting patients with metastatic colorectal cancer for treatment with temozolomide using proteomic analysis of MGMT (ID 3196)

Presentation Number
145P
Lecture Time
13:15 - 13:15
Speakers
  • S. Schwartz
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Temozolomide (TMZ) is a standard treatment for melanoma and glioblastoma and it has shown limited but encouraging activity in patients with metastatic colorectal cancer (mCRC). In multiple cancer types, the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) is a resistance marker for TMZ; MGMT promoter methylation is associated with loss of MGMT expression and response to TMZ. We hypothesized that mCRC patients whose tumors expressed quantities of MGMT protein below a pre-defined cutoff would have better outcomes on TMZ than patients with MGMT expression above the cutoff. To test our hypothesis, we assessed MGMT by mass spectrometry in the tumor samples of patients with refractory mCRC and MGMT promoter methylation receiving TMZ.

Methods

Archived formalin-fixed, paraffin-embedded tissue sections were obtained from 24 patients from two phase 2 trials. A pathologist marked the tumor areas, which were microdissected and solubilized. In each tumor sample, multiple protein biomarkers including MGMT were quantified with selected reaction monitoring mass spectrometry. An MGMT cutoff of 200 amol/ug was based on the limit of quantitation from a concentration curve. The Mantel-Cox log-rank and the Fisher’s exact tests were used for survival comparisons.

Results

MGMT protein was detected in 13 of 24 (54.2%) colorectal tumor samples (range: 229.3-784.8 amol/µg). The overall response rate was 29%. Patients with MGMT protein levels below a cutoff of 200 amol/ug (n = 11) had a notably higher response rate than patients with MGMT levels above the cutoff (64% vs. 0%; p = 0.001 Fisher’s test). Also a longer progression-free survival was observed (4.3 vs. 1.6 months, HR = 0.36, 95% CI = 0.13-1.10, p = 0.054). Results for overall survival were consistent but not statistically significant (8.9 vs 6.9 months, HR = 0.55, p = 0.221).

Conclusions

Patients with mCRC whose tumors expressed low or undetectable levels of MGMT protein had better outcomes following TMZ treatment than their counterparts. Quantitative proteomic analysis of MGMT could potentially be used to select CRC patients for TMZ treatment. The results of validation studies are forthcoming.

Legal entity responsible for the study

NantOmics

Funding

NantOmics

Disclosure

S. Schwartz, F. Cecchi, Y. Tian, K. Scott, T. Hembrough: Employee at NantOmics. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

146P - Predicting response to chemotherapy in gastric cancer patients randomized to docetaxel: A reevaluation of the ITACA-S trial (ID 4275)

Presentation Number
146P
Lecture Time
13:15 - 13:15
Speakers
  • F. Cecchi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

No predictive biomarker for chemotherapy has been validated for clinical use. Clinical studies suggest tumor expression of the proteins class III β-tubulin (TUBB3) and thymidine phosphorylase (TYMP) predict resistance to taxane and response to 5-FU, respectively. Immunohistochemical definitions of protein status vary widely. We evaluated relationships between survival and expression of TUBB3 and TYMP as quantitated by mass spectrometry in the archived tumor samples of 247 patients from the Intergroup Trial of Adenocarcinoma of the Stomach (ITACA-S). Patients had been randomized to monotherapy with 5-FU/LV or to FOLFIRI plus docetaxel and cisplatin.

Methods

Gastric tumor tissues were microdissected and solubilized for proteomic quantitation of 45 protein biomarkers. The cutoff for TUBB3 (750 amol/µg of total protein) was predetermined based on the assay’s limit of detection. An experimental cutoff for TYMP (1335 amol/ug) was derived in the 5-FU/LV-treated patients using Monte Carlo 2-fold cross-validation. The Mantel-Cox log-rank test was used for survival comparisons.

Results

Among gastric cancer (GC) patients treated with docetaxel-containing chemotherapy (n = 125), those with TUBB3 protein levels below the cutoff had a longer median overall survival (mOS) than patients with TUBB3 levels above the cutoff (1563 vs 886 days, p < 0.04). TUBB3 level made no difference in survival among patients who received 5-FU/LV (p = 0.64). Of note, among patients with high TUBB3 levels, those treated with 5-FU/LV survived 3 years longer than patients treated with docetaxel (mOS = 1991 vs 886 days, p = 0.048). 5-FU/LV-treated patients (n = 122) with TYMP protein expression above the cutoff survived longer than those with lower TYMP levels (mOS: 2362 vs 1062 days, p < 0.02). A combination of TUBB3 and TYMP expression predicted the longest survival in patients treated with FOLFIRI plus docetaxel (p < 0.001).

Conclusions

Quantitative proteomic analysis identified subsets of trial patients who benefitted from specific adjuvant chemotherapy regimens. Personalized chemotherapy based on quantitated TYMP and TUBB3 is promising and warrants broader evaluation.

Clinical trial identification

ClinicalTrials.gov Identifier:

Legal entity responsible for the study

NantOmics, LLC

Funding

IRCCS and NantOmics, LLC

Disclosure

F. Cecchi, S. Schwartz, S. Sellappan, T. Hembrough, Y. Tian: Employee at NantOmics. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

147P - The nationwide cancer genome screening project in Japan SCRUM-Japan GI-SCREEN: Efficient identification of cancer genome alterations in advanced pancreatic cancer (ID 2953)

Presentation Number
147P
Lecture Time
13:15 - 13:15
Speakers
  • D. Naruge
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

We have conducted the Nationwide Cancer Genome Screening Project in Japan since April 2015 using Next Generation Sequencing in cancers of digestive system, called as the SCRUM-Japan GI-SCREEN. In this presentation, we show the result of advanced pancreatic cancer (aPC).

Methods

This study is ongoing with the participation of 20 major cancer centers. Patients who plan to or receive systemic chemotherapy were eligible. DNA and RNA were extracted from FFPE tumor samples and were analyzed by the Oncomine Cancer Research Panel (OCP) which allows to detect gene mutation, copy number variant (CNV) and fusions across 143 genes in a CLIA certified CAP accredited laboratory. The detected genomic variant data were classified according to genetic drivers of cancer including gain- and loss-of-function or single nucleotide variant based on the Oncomine Knowledgebase. In this presentation, we show the results of aPC cohort.

Results

As of 31st October 2016, a total of 179 aPC samples were analyzed. The sequence with the OCP was successfully performed in 120 (67.0%). Out of 120 patients, the proportion of location of tumor and histology type is follows; Ph = 44%, adenocarcinoma 97%. The proportion of procedure for sample collection is follows; surgical resection 35.0%, needle biopsy 38.3%, EUS-FNA 20.0%, other 5.8%, and unknown 0.8%.The frequently detected mutations (> 10%) in 120 samples of which results were available were KRAS (93%), TP53 (63%), CDKN2A (12%), and SMAD4 (11%). Other important/druggable mutations were GNAS, BRCA2, and ATM (3%, each). Most frequently detected CNVs (≥ 7copies) was MYC (3%), and no gene fusion was detected.

Conclusions

This nationwide screening system is efficient to detect rare gene alterations in aPC. Alterations in potentially druggable genes were limited in aPC, but homologous recombination repair genes were attractive target. This novel knowledge provides an intriguing background to investigate new target approaches and represents a progress toward more precision medicine.

Clinical trial identification

Clinical trial information: UMIN000016344

Legal entity responsible for the study

SCRUM-Japan GI-SCREEN

Funding

15 SCRUM-Japan collaborating pharmaceutical companies, AMED, NCC

Disclosure

D. Naruge: Research Funding in institution: Taiho, Ono Pharmaceutical, Merck Serono, Takeda, Lilly Japan, Chugai Pharma, Bayer, GlaxoSmithKline, Yakult, Novartis, J-Pharma, Janssen, Sanofi, Kyowa Hakko Kirin, Daiichi Sankyo, Astellas, Pfizer, etc. C. Morizane: Consulting Role: AstraZeneca, Yakult, Novartis, Taiho Pharmaceutical. Honoraria: Pfizer, Novartis, Yakult, Lilly, Nobelpharma, Fujifilm. Research Funding: GlaxoSmithKline, Pfizer, Nobelpharma, Eisai, Yakult, ONO PHARMACEUTICAL, Taiho Pharmaceutical. M. Ueno: Consulting Role: Eisai. Honoraria: Taiho, Yakult, Ono Pharmaceutical, AstraZeneca, Novartis, Lilly. Research Funding of your institution: Taiho Pharmaceutical, Daiichi Sankyo, Eisai, AstraZeneca, Merck Serono, Ono Pharmaceutical, MSD. H. Takahashi: Honoraria: Taiho Pharmaceutical. Research Funding of your institution: Bayer, Bristol-Myers Squibb. Travel, Accommodations, Expenses: Pfizer. Y. Kawamoto: Honoraria: Sawai Pharmaceutical Co. S. Shimizu: Honoraria: Taiho Pharmaceutical, Nihonkayaku, Novartis. T.E. Nakajima: Honoraria: Taiho, Lilly, Chugai, Kyowa Hakko Kirin, Merck Serono, Takeda, Ono. Research Funding: Taiho, Ono, AstraZeneca, Lilly, Chugai, Kyowa Hakko Kirin, Merck Serono, Takeda, MSD, Eisai, Amgen Asetlas Biopharma K.K., Eisai Pharm, etc. H. Ishii: Advisory Role: Ono Pharmaceutical. Honoraria: Yakult, Taiho, Lilly Japan, Kyowa Hakko Kirin, Nobelpharma, Mochida Pharmaceutical Co. Ltd., Hospira. Research Funding: Taiho Pharmaceutical. T. Kato: Speakers\' Bureau: Chugai Pharma, Takeda, Lilly, Yakult. T. Kudo: Research Funding: Yakult, Chugai Pharma, Ono Pharmaceutical. S. Nomura: Employment: An Immediate Family Member is employed by Asahi-Kasei Pharma. K. Shitara: Research Funding: Daiichi Sankyo, Chugai Pharma, Lilly, MSD, Taiho Pharmaceutical, Dainippon Sumitomo Pharma. A. Ohtsu: Employment: An Immediate Family Member is employed by Celgene. Research Funding: Bristol-Myers Squibb. T. Yoshino: Research funding: GlaxoSmithKline K.K., Boehringer Ingelheim GmbH. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

BREAST CANCER, EARLY STAGE (ID 5673)

Lecture Time
13:15 - 13:15
Speakers
  • E. De Azambuja
  • C. Criscitiello
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15
Poster display session Poster Display session

164P - Impact of lack of surgery on outcomes in elderly patients with non-metastatic breast cancer (BC): A population based study using the SEER 18 data base (ID 1425)

Presentation Number
164P
Lecture Time
13:15 - 13:15
Speakers
  • C. Truica
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Elderly women with non-metastatic BC do not always receive standard of care definitive surgical treatment. Both provider and patient related reasons have been cited. The impact of omitting surgery in elderly patients who otherwise would be candidates for surgery has not been addressed. We performed a population based study to evaluate the impact of lack of surgery on survival outcomes in elderly women with BC in modern era.

Methods

The Surveillance, Epidemiology and End Results database was queried from 2010 to 2013 for female patients age 60 and older with a diagnosis of invasive ductal or lobular BC with AJCC stage I, II, III. To determine the relationship between surgery at diagnosis and survival and to take into consideration the effect of comorbidities, we organized patients in the following groups: a. Surgery performed, b. Surgery recommended, but not performed; c. Surgery not recommended and not performed. The Kaplan –Meier method was used to generate survival curves and the log-rank test was performed to compare OS rates among different groups.

Results

119,404 patients were eligible with a median age between 70 to 74 years old. 71,638 (60%) patients were stage I, 37,524 (31.42%) were stage II and 10,245 (8.58%) were stage III. 85.2% were ER +, 12.4% were Her 2 + and 8.8% were triple negative (TN). Compared with the patients who received surgery, patients who did not receive surgery had a significantly worse outcome (all patients: HR = 7.39, 95% CI, 6.98–7.83, P < 0.001. Patients who were recommended to have surgery but did not receive it had significantly inferior survival than patients who underwent surgery (HR = 5.08, 95% CI, 4.48–5.76, P < 0.001) although better OS than those who were recommended against surgery (HR = 0.62, 95% CI, 0.54–0.71, P < 0.001). Similar results were found in subgroup analyses regardless of age, tumor stage, ER or HER2 status. Patients with TNBC who did not received surgery also had a significantly worse OS than those who received surgery. (HR = 4.89, 95% CI, 4.07–5.88, P < 0.001).

Conclusions

Definitive surgery should be performed in medically-fit elderly patients with non-metastatic BC due to a significant survival benefit.

Legal entity responsible for the study

Cristina Truica

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

166P - Eight-year follow up results of the OTOASOR trial: The optimal treatment of the axilla – surgery or radiotherapy after positive sentinel lymph node biopsy in early-stage breast cancer. A randomized, single centre, phase III, non-inferiority trial (ID 1626)

Presentation Number
166P
Lecture Time
13:15 - 13:15
Speakers
  • Á. Sávolt
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The National Institute of Oncology, Budapest conducted a single centre randomized clinical study. The OTOASOR (Optimal Treatment Of the Axilla – Surgery Or Radiotherapy) trial compares completion of axillary lymph node dissection (cALND) to regional nodal irradiation (RNI) in patients with sentinel lymph node metastasis (pN1sn) in stage I-II breast cancer.

Methods

Patients with primary invasive breast cancer (cN0 and cT ≤ 3cm) were randomized before surgery for cALND (standard treatment) or RNI (investigational treatment). Sentinel lymph nodes (SN) were investigated with serial sectioning at 0.5 mm levels by haematoxylin-eosin staining. Investigational treatment arm patients received 50Gy RNI instead of cALND. Adjuvant treatment and follow up were performed according to the actual guidelines. Between August 2002 and June 2009, 1,054 patients were randomized for cALND and 1052 patients for RNI. SN was evaluated in 2,073 patients and was positive in 526 patients (25.4%). 474 cases were evaluable (244 in the cALND and 230 in the RNI arm), and in the cALND group 94 of 244 patients (38.5%) who underwent completion axillary surgery has additional positive nodes. The two arms were well balanced according to the majority of main prognostic factors. Primary endpoint was axillary recurrence and secondary endpoints were overall survival (OS) and disease-free survival (DFS).

Results

Mean follow-up was 97 months (Q1-Q3 80-120). Axillary recurrence was 2.0% in cALND arm vs. 1.7% in RNI arm (P = 1.00). OS at 8 years was 77.9% vs. 84.8% (P = 0.060), and DFS was 72.1% in cALND arm and 77.4% after RNI (P = 0.51). The results show that RNI is statistically not inferior to cALND treatment.

Conclusions

The long term follow-up results of this prospective-randomized trial suggest that RNI without cALND does not increase the risk of axillary failure in selected patients with early-stage invasive breast cancer (cT ≤ 3 cm, cN0) and pN1(sn). Axillary radiotherapy should be an alternative treatment for selected patients with sentinel lymph node metastases.

Legal entity responsible for the study

The trial was approved by the National Institute of Oncology’s Ethical Committee

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

167P - Time to surgery in early breast cancer treated with neoadjuvant chemotherapy (ID 3579)

Presentation Number
167P
Lecture Time
13:15 - 13:15
Speakers
  • M. Cinausero
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

A delay between surgery and adjuvant chemotherapy (CT) has been associated with worse outcome in early breast cancer (BC), but little is known about timing-related consequences in the neoadjuvant setting. Aim of this study is to investigate the impact of the interval between the end of neoadjuvant CT and surgery (CTTS).

Methods

This retrospective study analyzed a series of 469 consecutive BC patients (pts) receiving neoadjuvant CT at the Department of Oncology of Udine (n = 222) and of the Istituto Nazionale Tumori of Milan (n = 247), between 2004 and 2015. CTTS was defined as the time between the last CT administration and surgery. Prognostic impact was investigated through Cox regression.

Results

Luminal-like subtype was the most frequent (53.69%) followed by HER2-positive (29.26%) and triple negative BC (17.05%). Median follow-up was 55.07 months (mo). Estimated overall survival (OS) at 24 and 60 mo was 96.4% and 88.2%, respectively. Estimated relapse free survival (RFS) at 24 and 60 mo was 83.6% and 65.8%. Median CTTS was 1.08 mo (25%-75% range: 0.89 - 1.2 mo). Among the total population no statistically significant differences were observed in terms of OS and RFS between CTTS > 1 vs < 1 mo (HR 1.18, 95%CI 0.72-1.98; HR 1.28, 95%CI 0.89-1.85, respectively). On multivariate analysis, grade 3 and Ki67 > =20% were associated with worse RFS (HR 2.09, 95%CI 1.31-3.33; HR 2.77, 95%CI 1.30-5.91, respectively); on the other hand, a pathological complete response was associated with better RFS (HR 0.23, 95%CI 0.09-0.56). In terms of OS, grading and Ki67 were marginally significant. Subgroup analysis for CTTS showed no statistically significant differences when stratification for the main clinico-pathological features was applied. Notably, a trend for interaction in the nodal status stratification was observed.

Conclusions

This study explored the impact on RFS and OS of the interval between the end of neaodjuvant CT and surgery. Notwithstanding the exploratory purpose, the results suggest that an interval superior to 1 month was not significantly detrimental in terms of both RFS and OS.

Legal entity responsible for the study

N/A

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

168P - Updated results of the Breast cancer task force phase II study of neoadjuvant weekly carboplatin (Cp) added to paclitaxel (P) followed by epirubicin (E) and cyclophosphamide (C) in triple negative breast cancer (TNBC) patients (pts) (ID 3605)

Presentation Number
168P
Lecture Time
13:15 - 13:15
Speakers
  • C. Fontaine
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Introduction: TNBC has the highest mortality of all BC subtypes. Neoadjuvant platinum added to the taxanes-antracycline chemotherapy has been reported to improve pathologic complete response (pCR) rate and survival in TNBC. Aim: To evaluate the efficacy and toxicity of the addition of weekly Cp to Pl and dose dense EC on the pCR rate in an open-label phase II study in stage II/III TNBC pts.

Methods

Patients and methods: Sixty three pts received dose dense P (80mg/m2/wk) concurrent with Cp (AUC=2) for 12 wks, added to two-weekly E (90mg/m2) and C (600mg/m2) for 4 cycles, and followed by surgery and radiotherapy. The primary endpoint was pCR in the breast and axilla. Additionally adverse events are registered. A correlative assessment of germ line mutations in HRD genes is ongoing. Pts are monitored for clinical response by magnetic resonance and mammography and also for relapse free survival and time to treatment failure. The study sample size has been calculated according to the optimal Simon’s two-stage design method. The target sample size was 63 patients with 80% power to detect a pCR rate of > or = 47% (α = 0.05).

Results

Accrual to the study is completed and 63 eligible pts with operable, non-inflammatory stage II/III TNBC were included. Most pts were between 40 and 60 yr old and 49 out of 63 were stage T2. Forty percent were clinically node + and 68% were G3. Seventy three percent received breast conserving surgery. Thirty eight out of 63 pts (60%) achieved a pCR rate in the breast and axilla. In 52 evaluable pts for toxicity, the main toxicity for part 1 (Cp+P) of the combination was neutropenia G3/4 in 37 pts (71%) and for part 2 (EC) febrile neutropenia G3/4 in 18 pts (34%) despite primary prophylaxis, followed by thrombocytopenia G3/4 in 11 pts (21%). Only three pts had a neuropathy G3.

Conclusions

The addition of weekly carboplatinum to neoadjuvant dose dense paclitaxel and EC is feasible and a pCR rate in the breast and axilla as high as 60% in early TNBC pts is obtained. Correlation with genomic HRD deficiency is ongoing.

Clinical trial identification

2014-003723-21; 28-02-2014

Legal entity responsible for the study

Breast Cancer Task force on behalf of the BSMO (Belgian Society of Medical Oncology)

Funding

Amgen and Teva

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

169P - Neoadjuvant eribulin following anthracycline and taxane in triple negative breast cancer (HOPE): A multicenter, two stage, phase II trial (ID 4914)

Presentation Number
169P
Lecture Time
13:15 - 13:15
Speakers
  • S. Di Cosimo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Neoadjuvant chemotherapy for triple negative breast cancer (TNBC) patients (pts) is mostly based on anthracycline and taxane (AT). The microtubule dynamics inhibitor eribulin mesylate (E) has been proved to increase survival in heavily pretreated metastatic breast cancer pts. We argue that sequential AT and E would benefit also non-metastatic TNBC pts.

Methods

Pts with primary TNBC>2 cm received doxorubicin 60 mg/m2+paclitaxel 200 mg/m2 day 1, q 21 for 4 cycles, followed by E 1.4 mg/m2 days 1,8 q 21 for 4 cycles. The primary endpoint was the pathological complete response (pCR) rate; secondary endpoints included clinical response rate by RECIST 1.1, metabolic response by EORTC criteria, safety profile and biomarker analysis. Sample size was estimated to detect a 40% pCR, with 20% for a minimal hypothesis. With a type I error of 0.05 and a statistical power of 80%, 13 patients were to be initially enrolled. Observation of ≥ 4 pCR would justify continuation of accrual up to 43 pts.

Results

Thirteen pts were enrolled. The combination was safe with mostly G1/2 toxicities. The only G ≥ 3 event was neutropenia, which was related to AT in 4 pts and to E in 2 pts. An analysis of the degree of response by imaging was feasible in 11 pts. Overall 6/11 (54%) pts achieved a partial response after AT; of note 3/5 (27%) AT-unresponsive pts responded to E, 1 had stable disease and 1 pt definitely progressed. FDG PET/CT scans were available in 12 pts after AT and in 10 pts after 2 cycles of E. Median SUVmax values were 13, 3, and 2 at baseline, after AT and after E, respectively. Of the 4 pts with complete metabolic response (2 after AT and 2 after E), 2 achieved pCR at surgery. Immunostaining of paired pre- and post-treatment tumor bioptic specimens revealed reduction of beta-catenin, cyclin D and c-myc expression in the absence of n-cadherin modulation. The trial was halted because of the unmet primary endopoint (3 pCR).

Conclusions

Despite an unmet primary endpoint, E showed clinical and biological activity in TNBC pts. The observed modulation of beta-catenin pathway core proteins, purportedly outside the domain of epithelial mesenchymal transition, claims for further investigation and for the refinement in the definition of the clinical endpoints.

Clinical trial identification

EUDRACT: RELEASE: November 2012

Legal entity responsible for the study

Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy

Funding

Eisai

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

170P - Impact of neoadjuvant therapy (NT) and pathological complete response (pCR) on breast-conserving surgery (BCS) in patients (pts) with breast cancer (BC): A meta-analysis (ID 5038)

Presentation Number
170P
Lecture Time
13:15 - 13:15
Speakers
  • C. Criscitiello
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

We performed a meta-analysis of randomized trials that evaluated the pCR and surgical decisions after NT for BC.

Methods

Studies were retrieved by searching the PubMed database and the proceedings of major conferences. The primary outcome was BCS rate. Secondary outcomes were pCR rate and association to BCS. Meta-analyses were performed using random-effects (RE) models that use inverse-variance weighting for each treatment arm based on the number of evaluable pts. Point estimates are reported with 95% confidence intervals, and p < 0.05 was considered statistically significant.

Results

36 studies were identified (N = 12,311 pts). 17/36 studies reported both pCR and BCT for at least 1 treatment arm, and comprise the studies in this analysis. The number of arms per study ranged from 1 to 6, such that 42 independent units were available to evaluate the association between pCR and BCT. The BCS rate ranged from 5% to 76% across arms with an average BCS of 57% (95% CI 52%-62%). Significant heterogeneity was observed among the trials (Cochrane Q = 787, p < 0.001, I2= 97%). In the meta-regression model, BCS rates were not significantly associated with year of first patient-in (p = 0.89), grade (p = 0.93), hormone-receptor status (p = 0.39). Clinical N-stage (p = 0.01) and HER2 status (p = 0.03) were significantly associated with BCS. pCR rate ranged from 3% to 60% across studies. The average pCR across all study arms was 24% (95% CI 19%-29%). No association was observed between pCR rate in a study arm and the resulting BCS rate in a univariate model (p = 0.34) nor after adjusting for HER2 and clinical nodal status (p = 0.82). Change in BCS with NT was further derived from the subset of 14 multi-arm trials, where 17 pairwise contrasts were made between randomized populations: 2 studies showed a significant difference in BCS. The difference in pCR rates between treatment arms ranged from -22% to 24%. However, no significant association is seen with the corresponding difference in BCS rates (p = 0.27).

Conclusions

This meta-analysis indicates that, despite increasing pCR, there is no concomitant increase in BCS. Future NT studies should reconsider using BCS as an endpoint.

Legal entity responsible for the study

Carmen Criscitiello

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

171P - Composite index of risk shows that benefit from adjuvant dose dense chemotherapy is not confined to triple negative breast cancer (ID 4548)

Presentation Number
171P
Lecture Time
13:15 - 13:15
Speakers
  • F. Puglisi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Compared with the standard interval adjuvant chemotherapy, dose-dense schedule is proved to increase disease-free survival (DFS) in node-positive early breast cancer (EBC) patients (pts). To date, GIM2 is the only trial supporting the role of dose-dense chemotherapy in pts with hormone receptor-negative (HR-) or hormone receptor-positive tumours (HR+) (Del Mastro et al. Lancet 2015). To further refine the evidence of treatment effect in the HR+ subgroup, a composite index of risk was developed including clinico-pathological features.

Methods

The randomized phase III GIM2 trial enrolled 2091 pts with node-positive EBC (primary endpoint: DFS). A continuous, composite measure of treatment benefit was determined from a Cox model incorporating potential predictive factors (age: 25-40/41-55/56-71; histological grade: 1 + 2/3; HR status: positive/negative; ki-67 levels: ≤20%/>20%). Subpopulation treatment effect pattern plot methodology was used to reveal differential treatment effects on DFS according to composite index. The study analyzed the cohort of pts with HER2- (N = 1127) disease with a special focus on HR+ disease (N = 980).

Results

On average, the magnitude of benefit with dose dense chemotherapy versus standard chemotherapy widely varied according to composite measure of specific features. In the HER2- subgroup, the highest benefit was observed in pts with G3, HR-, >10 positive nodes, age <40 yrs, ki-67 > 20% (hazard ratio for DFS 0.57, 95% CI 0.35-0.94). Notably, among pts with HR+ disease, the following clinic-pathological characteristics conferred the highest benefit: G3, ≥4 positive nodes, age ≥56 yrs, ki-67 > 20% (hazard ratio for DFS 0.66, 95% CI 0.38-1.15).

Conclusions

Composite risk evaluation and corresponding subpopulation treatment effect pattern plot methodology suggest that benefit of dose dense adjuvant chemotherapy is not confined to triple negative EBC.

Legal entity responsible for the study

Gruppo Italiano Mammella

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

172P - The prognostic impact of chemotherapy induced amenorrhea in women treated with early stage breast cancer (ID 2598)

Presentation Number
172P
Lecture Time
13:15 - 13:15
Speakers
  • C. Ordu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In this study; Amenorrhea caused by chemotherapy in early stage breast cancer patients and its effects on survival were investigated.

Methods

A total of 389 patients who received adjuvant chemotherapy from 600 premenopausal patients who were treated with early stage breast cancer during 2000-2013 and followed up were included in the study. Patients who did not undergo medical ovarian ablation (OA); Amenore as developing and non-developing, two groups were separated and compared with clinicopathologic features and survival. SPSS 17. version was used.

Results

Disease-free survival (DFS): median (m) 57 months (4-197 months), overall survival (OS): m 60 (10-168 months) and follow up time m 60 months (23-168 months). During follow-up, chemotherapy induced amenorrhea (CIA) was observed in 145 (57.5%) of 252 patients who did not have any ovarian ablation (OA. The 5-year OS rate of patients with CIA was significantly higher than the patients without CIA (p = 0.042, 95.9% vs 89.7 vs 158.88 vs 135.33 months, respectively). In the subgroup analysis, the OS in patients with CIA was significantly higher than in those without CIA in patients had HR (+) (p = 0.036, 97.5% vs 91.5 vs 162.13 vs. 136.20 months, respectively). There was no significant difference in the duration of OS between with CIA and without CIA of the patients who had not HR (+) (p = 0.736, 90.9% vs 86.8% and 126.16 and 133.76 months, respectively). The duration of OS was significantly longer in patients with CIA in the luminal A molecular subtype than in those luminal B molecular subtype, but the difference was not significant in patients without CIA (p = 0.027 vs p = 0.074 respectively).

Conclusions

The development of amenorrhea due to chemotherapy provides a significant survival advantage over those patients who do not develop amenorrhea due to chemotherapy. This advantage is more pronounced in hormone receptor positive, lymph node involvement and advanced disease. The development of amenorrhea due to chemotherapy in patients with HR negative does not affect survival. Amenorrhea development further prolongs survival compared to luminal B in the luminal A molecular subtype.

Legal entity responsible for the study

Istanbul Bilim University, Florence Nightingale Group of Hospitals

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

173P - Determinants and outcomes associated with delays in adjuvant chemotherapy among breast cancer patients (ID 4144)

Presentation Number
173P
Lecture Time
13:15 - 13:15
Speakers
  • M. Chavez Mac Gregor
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Adverse outcomes have been associated with delays in the administration of adjuvant chemotherapy among breast cancer patients. We evaluate the determinants and outcomes associated with delays in time to chemotherapy (TTC) in a large cohort of older breast cancer patients.

Methods

We used the NCI-Surveillance Epidemiology and End Results (SEER) and Texas Cancer Registry (TCR)-Medicare linked data bases to identify patients ≥66 years old diagnosed with localized or regional breast cancer between 2001-2011. All patients received chemotherapy within 9 months of surgery. Delayed TTC was defined as > 90 days. Multivariable logistic regression was used to identify predictors of treatment delay. A Cox Proportional Hazards model was fit to determine the association between treatment delay, overall survival (OS) and breast cancer specific survival (BCSS).

Results

25,096 patients were included, of them 2,676 (10.7%) had a TTC >90 days. In multivariable analysis factors associated with delays in TTC were: recent year of diagnosis (2011 vs 2001 OR = 1.31; 95%CI 1.03-1.67), older age (76-80 vs 66-70 OR = 1.51; 95%CI 1.33-1.72), black race (OR = 1.35; 95%CI 1.14-1.58), having state buy-in (as an indicator of poverty) (OR = 1.27; 95%CI 1.1-1.47), comorbidities (Charlson score 1 OR = 1.23;95%CI 1.09-1.37; score 2 OR = 1.57; 95%CI 1.37-1.81), mastectomy (OR = 1.49; 95%CI 1.33-1.67), mastectomy with immediate reconstruction (OR = 1.85; 95%CI 1.37-2.48), Oncotype DX testing (OR = 1.68; 95%CI 1.4-2.02), mastectomy >30 days after the initial surgery (OR = 16.91; 95%CI 12.07-23.68), brachytherapy (OR = 4.11; 95%CI 3.17-5.34) and whole breast radiation prior to chemotherapy (OR = 31.9; 95%CI 28.05-36.49). After adjusting for potential confounders, patients with TTC >90 days had worse OS (HR = 1.37; 95%CI 1.27-1.48) and BCSS (HR = 1.34; 95%CI 1.19-1.51).

Conclusions

A delay in adjuvant chemotherapy administration >90 days is associated with adverse outcomes among older breast cancer patients. Determinants of delays were sociodemographic in nature, related to patient’s characteristics and to treatment received. Every effort should be made to identify vulnerable patients and to administer chemotherapy in a timely manner.

Legal entity responsible for the study

The University of Texas MD Anderson Cancer Center

Funding

Susan G. Komen, CIPRIT

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

174P - Adjuvant chemotherapy in pT1ab node-negative triple negative breast carcinomas: Results of a national multi-institutional retrospective study (ID 1339)

Presentation Number
174P
Lecture Time
13:15 - 13:15
Speakers
  • A. De Nonneville
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Triple negative breast cancers (TNBC) are considered as associated with poor outcome, but prognosis of subcentimetric, node-negative disease remains controversial and evidence that adjuvant chemotherapy (CT) is effective in these small tumors remains limited.

Methods

Our objective was to investigate the impact of adjuvant CT on survival in pT1abN0M0 TNBC. Patients were retrospectively identified from a cohort of 22,475 patients who underwent primary surgery in 15 French centers between 1987 and 2013. Since rare pathological types may display very particular prognoses in these tumors, we retained only the invasive ductal carcinomas of no special type according to the last WHO classification witch is most common TNBC histologic type. End-points were disease-free survival (DFS) and metastasis-free survival (MFS). A propensity score for receiving CT was estimated using a logistic regression including age, tumor size, SBR grade, and lymphovascular invasion.

Results

Of a total of 284 patients with pT1abN0M0 ductal TNBC, 144 (51%) received post-operative CT and 140 (49%) did not. Patients receiving CT had more adverse prognostic features, such as tumor size, high grade, young age, and lympho-vascular invasion. Adjuvant CT was not associated with a significant benefit for DFS (Hazard ratio, HR = 0.77 [0.40-1.46]; p = 0.419, Log-rank test) or MFS (HR = 1.00 [0.46-2.19], p = 0.997), with 5-year DFS and MFS in the group with CT vs. without of 90% [81%-94%] vs. 84% [74%-90%], and 90% [81%-95%] vs. 90% [83%-95%], respectively. Results were consistent in all supportive analyses including multivariate Cox model and the use of the propensity score for adjustment and as a matching factor for case–control analyses.

Conclusions

This study did not identify a significant DFS or MFS advantage for adjuvant CT in subcentimetric, node-negative ductal TNBC. Although current consensus guidelines recommend consideration of adjuvant CT in all TNBC larger than 5 mm, clinicians should carefully discuss benefit/risk ratio with patients, given the yet unproven benefits.

Legal entity responsible for the study

SIRIC program (INCa-DGOS-Inserm 6038)

Funding

SIRIC program (INCa-DGOS-Inserm 6038)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

175P - OHERA: A real world study of cardiac events in > 3700 patients with her2-positive early breast cancer treated with trastuzumab: Final analysis (ID 4629)

Presentation Number
175P
Lecture Time
13:15 - 13:15
Speakers
  • E. Lidbrink
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

As of Sept 2016, > 2.2 million breast cancer (BC) patients (pts) have received trastuzumab (Herceptin®; H) in clinical trial or real world settings. Risk of cardiac failure in pts treated with H in real world practice may differ from that observed in a clinical trial setting.

Methods

OHERA (NCT01152606) is a non-interventional post-approval safety study that investigated incidence of symptomatic congestive heart failure (CHF) and cardiac death in pts with HER2-positive early BC (EBC) receiving adjuvant intravenous H (H IV) in routine clinical practice, per the EU Summary of Product Characteristics (SmPC). Pts with HER2-positive EBC (stage I–IIIb) considered for treatment with H IV per the EU SmPC were enrolled, treated and monitored per local practice. Primary endpoints were incidence of symptomatic CHF (New York Heart Association [NYHA] class II–IV) and incidence of cardiac death. Secondary endpoints included time to onset of CHF. The final analysis included pt data collected for up to 5 years or until death, loss to follow-up or consent withdrawal.

Results

Pts were enrolled Aug 2007–Nov 2010 at 199 sites across 9 countries. The safety population included 3733 pts with EBC treated with H IV. Median treatment duration was 11.8 months; median follow-up was 60.1 months. Incidence of symptomatic CHF was 2.8% (n = 106); including severe symptomatic CHF (NYHA class III–IV) in 1.0% (n = 38) pts. Median time to onset of symptomatic CHF was 5.7 months (95% CI 5.3–6.5) and 77 (72.6%) pts achieved CHF resolution. Incidence of cardiac death was 0.2% (n = 6). 251 pts had a left ventricular ejection fraction (LVEF) drop of ≥ 10% from baseline to < 50% and 169/251 (67.3%) achieved LVEF drop resolution. Incidence of CHF was higher in pts with baseline risk factors such as pre-existing cardiac conditions, age ≥65 years or baseline LVEF ≤55%. Pts who had left-side radiotherapy at baseline did not have higher CHF incidence.

Conclusions

OHERA is the largest study investigating the cardiac safety of adjuvant H IV in a real world setting to date. Final analysis results were consistent with cardiac safety results from previous adjuvant H IV clinical trials in EBC, and the baseline risk factors for CHF reported in the H IV EU SmPC.

Clinical trial identification

Protocol number: BO20652/RO 45-2317 ClinicalTrials.gov NCT01152606

Legal entity responsible for the study

F Hoffmann-La Roche Ltd

Funding

F. Hoffmann-La Roche Ltd., Basel, Switzerland

Disclosure

A. Bouhlel, M. Liste Hermoso: Employee of F Hoffmann-La Roche Ltd. S. Lauer: Contract work for Hoffmann-La Roche Ltd. E. Nüesch: Stock ownership: Roche. Employee of F. Hoffmann-La Roche Ltd. M. Shing: Previous employee of Genentech Inc. & current collaboration with Genentech Inc. V. Misra: Membership of an advisory board: Amgen, Eisai, Roche. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

176P - Clinical outcomes of delayed start of trastuzumab treatment in patients with early breast cancer: ml25232 study (ID 2384)

Presentation Number
176P
Lecture Time
13:15 - 13:15
Speakers
  • S. Beslija
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Breast cancer patients in low and middle income countries have limited access to targeted therapies such as trastuzumab. The discontinuous availability of trastuzumab created waiting lists and subsequent very delayed treatment. Since few studies have systematically analyzed possible deleterious effect of delayed trastuzumab treatment, we designed a study to investigate its consequences on overall survival and disease-free survival.

Methods

This was a multicenter cohort study of HER2-positive early breast cancer patients (n = 223) diagnosed between 01/05/2005 and 01/05/2010 in the Federation of Bosnia and Herzegovina. The study began in 01/01/2010, and enrollment was completed in 30/06/2012. Last follow up and cut off date for analysis was 31/03/2015.

Results

A total of 223 women (median 55 years; IQR: 49-61 years) were recruited. Since 131 (59%) patients waited for > 6 months after surgery to receive trastuzumab, we categorized our patient cohort into three groups: non-waiting group (n = 92; wait time < 6 months), and waiting group 1 (n = 85; wait time between 7 to 12 months) and waiting group 2 (n = 46; > 13 month wait). OS at 5 years in non-waiting group was 84%, compared to 72% in wait group 1 and 75% in wait group 2 (p > 0.05). DFS at 5 years in the non-wait group was 79%, compared to 65% in wait group 1, and 68% in wait group 2 (p > 0.05).

Conclusions

Unfortunate and unique circumstances in developing countries have created waiting lists for trastuzumab treatment—our systematic analysis of 223 women has shown that delayed start of trastuzumab treatment does not have a statistically significant effect on clinical outcomes, but shows a trend towards worse OS and DFS for women with delayed treatment. Thus, trastuzumab treatment has a persistent benefit even when administered with delayed start.

Clinical trial identification

ML25232

Legal entity responsible for the study

Roche

Funding

Roche

Disclosure

T. Ceric: Honoraria: Roche, Novartis, Pfizer. Consulting or Advisory Role: Roche, Novartis, Pfizer. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

177P - Effects of neratinib (N) on health-related quality of life (HRQoL) in early-stage HER2+ breast cancer (BC): longitudinal analyses from the phase III ExteNET trial (ID 4297)

Presentation Number
177P
Lecture Time
13:15 - 13:15
Speakers
  • S. Delaloge
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The international, randomized, placebo (P)-controlled phase III ExteNET trial (NCT00878709) showed that N for 1 y after trastuzumab-based adjuvant therapy significantly improved 2-y invasive disease-free survival in early HER2+ BC patients (pts) (HR 0.67; 95% CI 0.50–0.91; p = 0.0091) [Chan et al. Lancet Oncol 2016]. Detailed longitudinal evaluation of HRQoL was an exploratory endpoint of ExteNET.

Methods

2840 pts received N 240 mg/d or P for 1 yr. Pts completed FACT-B and EQ-5D questionnaires at baseline and months (M) 1, 3, 6, 9, and 12. Changes in scores from baseline were compared between groups using ANCOVA with no imputation for missing values. Sensitivity analyses using alternative methods were applied. Changes in HRQoL scores were considered to be clinically meaningful if greater than minimal clinically important differences (MCID) reported in the literature.

Results

2407 pts were evaluable for FACT-B (N, n = 1171; P, n = 1236), and 2427 for EQ-5D (N, n = 1186; P, n = 1241). Compliance with questionnaires exceeded 85%. N was associated with decreased HRQoL scores at M1 vs P, after which between-group differences diminished (Table). They were consistently less than MCIDs, except for physical well-being (PWB) subscale at M1. BC subscale (BCS) showed small improvements with N at M3–M9, all less than MCIDs. Different sensitivity methods did not alter the results.

177P

Adjusted mean difference in change from baseline:
ScaleMCID range N vs P
M1M3M6M9M12
FACT-B total7–8–2.9*0.1–0.6–0.5–0.8
TOI-PFB5–6–2.6*–0.3–0.7–0.4–1.0*
TOI-ESB5–601.2*0.60.50.4
PWB2–32.4*–1.1*–1.0*–0.9*–1.1*
BCS2–30.30.7*0.4*0.6*0.2
EQ-5D index0.09–0.10–0.02*–0–000
EQ health state7–10–2.7*0.1–1.3*–0.7–0.4

For baseline score;

Statistically significant at p < 0.05 without adjustment for multiple testing TOI = trial outcome index; PFB = PWB + functional WB + BCS; ESB = emotional WB + social WB + BCS.

Conclusions

N was associated with decreased HRQoL, in particular in PWB, at M1, possibly due to N-related diarrhea. Based on their small magnitude, differences observed after M1 in PWB favoring P and in BCS favoring N, may not be clinically important.

Clinical trial identification

NCT00878709

Legal entity responsible for the study

Wyeth, Pfizer and Puma Biotechnology

Funding

Puma Biotechnology

Disclosure

S. Delaloge: Grants and personal fees from Roche and GSK. Y. Ye: Employment: Puma Biotechnology Inc. M. Buyse: Employee and shareholder of IDDI. A. Chan: Personal fees for educational meetings from Pfizer, Amgen. Non-financial support from Puma Biotechnology outside the submitted work. C. Barrios: Grants from Amgen, AstraZeneca, Boehringer Ingelheim, Novartis, Pfizer, Roche, Celgene, Sanofi, Lilly, Puma. Personal fees from GSK, Novartis, Pfizer, Roche, Eisai. B. Ejlertsen: Grants to institution from NanoString, Novartis, and Roche, outside the submitted work. Travel support for educational meetings from AstraZeneca and Celgene. G. von Minckwitz: Research funding to the institution from Amgen, Roche, Novartis, Celgene, Teva, AstraZeneca, Myriad Genetics, AbbVie and Vifor Pharma. M. Gnant: Grants from Sanofi-Aventis, Novartis, Roche, GSK, Pfizer, Smith Medical. Personal fees from Novartis, AstraZeneca, Accelsiors, Eisai. S. Moran, A.H. Auerbach: Employment and stock options: Puma Biotechnology Inc. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

178P - Second interim analysis of HerSCin, a German non-interventional study of subcutaneous trastuzumab for HER2-positive early breast cancer in routine clinical practice (ID 2944)

Presentation Number
178P
Lecture Time
13:15 - 13:15
Speakers
  • K. Lüdtke-Heckenkamp
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Compared with IV trastuzumab, subcutaneous trastuzumab (HSC) showed non-inferior outcomes in the HannaH trial and was preferred by patients (pts) and healthcare professionals in the PrefHer study. The ongoing HerSCin study (NCT01959386) is evaluating HSC in routine clinical practice in Germany.

Methods

Pts with HER2-positive early breast cancer treated with (neo)adjuvant HSC (investigator’s chosen regimen) in routine oncology practice between Nov 2013 and Nov 2016 were eligible. Pts could be enrolled retrospectively up to 9 wks after starting HSC. Baseline characteristics, treatment, adverse events (AEs), clinical outcomes and quality of life (EORTC QLQ-C30/QLQ-BR23) data are collected prospectively. Primary efficacy endpoints are pCR rate (neoadjuvant setting) and 2-year disease-free survival (adjuvant setting).

Results

At the data cut-off for the second planned interim analysis (Nov 2016), 420 of 1003 pts enrolled to date from 103 German centres had completed therapy and were eligible for analysis. The median duration of follow-up was 12.2 (range 3.3–25.8) mo. Baseline characteristics are below. The mean duration of HSC was 8.8 mo (neoadjuvant: 9.2; adjuvant: 8.7). All-grade and grade ≥3 AEs were reported in 63% and 15% of pts, respectively. The most common all-grade AEs were fatigue (10%), diarrhoea (9%) and arthralgia (7%). AEs led to treatment interruption/withdrawal in 48 pts (11%). Only 1 of the 4 fatal AEs was considered treatment related (cardiac/respiratory failure). The pCR rate (including carcinoma in situ) in the neoadjuvant subgroup was 60.3% (95% CI 48.5–71.2). Efficacy results in the adjuvant subgroup are not mature.

178P

Parameter, No. of pts (%)All pts (n = 420) Neoadjuvant subgroup (n = 78) Adjuvant subgroup (n = 342)
Median age, years (range) 56 (20–90) 52 (20–77) 57 (27–90)
ECOG performance status 0 258 (61) 44 (56) 214 (63)
1 116 (28) 26 (33) 90 (26)
2 10 (2) 1 (1) 9 (3)
Missing 36 (9) 7 (9) 29 (8)
Cardiac conditions at baseline Arterial hypertension 134 (32) 24 (31) 110 (32)
Coronary heart disease 14 (3) 3 (4) 11 (3)
HER2 status by IHC 0/1+ 3 (1) 0 3 (1)
2+ 104 (25) 13 (17) 91 (27)
3+ 310 (74) 65 (83) 245 (72)
Missing 3 (1) 0 3 (1)
Histological grade 1 7 (2) 0 7 (2)
2 184 (44) 30 (38) 154 (45)
3 222 (53) 46 (59) 176 (51)
Missing/unknown 7 (2) 2 (3) 5 (1)
Subtypea Ductal 343 (82) 65 (83) 278 (81)
Lobular 24 (6) 3 (4) 21 (6)
Other 54 (13) 10 (13) 44 (13)
Positive nodal status 161 (38) 25 (32) 136 (40)
Hormone receptor status ER positive 280 (67) 42 (54) 238 (70)
PgR positive 234 (56) 39 (50) 195 (57)
ER and PgR negative 127 (30) 31 (40) 96 (28)

One patient (adjuvant setting) recorded as both ductal and lobular. ER=oestrogen receptor; IHC=immunohistochemistry; PgR=progesterone receptor.

Conclusions

The 60.3% pCR rate is consistent with prospective trials of IV trastuzumab and HSC. Tolerability is as expected based on results from randomised trials. HSC is an active, feasible and tolerable treatment for use in routine oncology practice as well as the clinical trial setting.

Clinical trial identification

NCT01959386

Legal entity responsible for the study

Roche Pharma AG

Funding

Roche Pharma AG

Disclosure

S. Kümmel: Membership on advisory board or board of directors: Roche Pharma AG.

\r\n

S. Busch-Liles: Employment: Roche Pharma AG.

\r\n

M. Schmidt: Membership on advisory board or board of directors: Novartis, Pfizer, Pierre-Fabre, Roche. Corporate-sponsored research: Pierre-Fabre.

\r\n

All other authors have declared no conflicts of interest.

Disclosure

S. Kümmel: Membership on advisory board or board of directors: Roche Pharma AG.

S. Busch-Liles: Employment: Roche Pharma AG.

M. Schmidt: Membership on advisory board or board of directors: Novartis, Pfizer, Pierre-Fabre, Roche. Corporate-sponsored research: Pierre-Fabre.

All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

179P - Timing of initiation of trastuzumab (T) and long-term outcome of patients (pts) with early-stage (ES) HER2-positive (HER2+) breast cancer (BrCa): Impact of neo-adjuvant (NAdj) versus adjuvant (Adj) strategy (ID 4162)

Presentation Number
179P
Lecture Time
13:15 - 13:15
Speakers
  • G. Gullo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The optimal schedule of anti-HER2 Tx for HER2+ ESBrCa with respect to chemotherapy and surgery remains undefined. We performed a retrospective analysis of a large, prospectively maintained single institution data base to study the impact of treatment schedule on clinical outcome.

Methods

Our database included all pts treated with T for Stage I to III HER2+BrCa who had a minimum follow up (FU) of 3 years. Time-to-first-T (TFT) was calculated from the date of the first diagnostic breast biopsy to the date of the first T. Pts with stage N3b and N3c or inoperable disease were excluded from the study.

Results

A total of 506 pts treated between October 2001 and March 2014 were included in the study. T was administered as part of AdjTx in 386 (76%) pts, and of NAdjTx in 120 (24%), 338 (67%) pts had TCH [docetaxel/CBDCA/T] or “TCH-like”, 119 (24%) pts had delayed concomitant (i.e. AC-TH)/sequential T. Median FU is 73.3 months (range 1.4-176.3). Median TFT for the overall cohort was 12 weeks (range 1.9-122.3). Median TFT was significantly shorter in the NAdj than in the Adj cohort: 4.4 vs 14 weeks [p<0.00001]. In the entire cohort, DFS and OS rates were 83% and 91%, respectively. DFS and OS rates were 90% and 96% vs 75% and 85% for pts with TFT ≤12 weeks vs TFT >12 weeks, respectively [p < 0.0001]. Pts with TFT >12 weeks had a significantly higher risk of recurrence [HR 1.96; p = 0.008] and death [HR 2.84; p = 0.006] than pts with TFT ≤12 weeks. Pts with positive lymph nodes (LN+) and TFT >12 weeks had significantly higher risk of relapse [HR 2.40, p = 0.001] and death [HR 2.10, p = 0.024] than pts with TFT ≤12 weeks. However, despite NAdj pts having significantly higher rate of LN + (75% vs 53%, p < 0.0001), DFS and OS were superimposable in the two cohorts. Pts with LN+ had superior DFS when treated with NAdjTx compared with AdjTx [p = 0.006].

Conclusions

Our mature data indicate that timing of anti-HER2 Tx significantly affects long-term outcome and T should be initiated ≤12 weeks from first diagnosis of ES HER2+BrCa. The early institution of T in the NAdj cohort abolished the negative impact of LN+, thus suggesting that this should be considered the optimal Tx strategy for ES HER2+BrCa.

Clinical trial identification

Not applicable.

Legal entity responsible for the study

Giuseppe Gullo

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

180P - Adjuvant endocrine treatment: Stop or continue? (ID 5178)

Presentation Number
180P
Lecture Time
13:15 - 13:15
Speakers
  • E. Muller
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

There is currently a trend towards extending adjuvant endocrine treatment in higher risk breast cancer patients up 10 years. However, a trade off has to be made between persisting side effects of endocrine treatment vs a small advantage in recurrence risk. It is not well known if patients still suffer side effects after 5 years of endocrine treatment, and if these may be reversible. Therefore, we studied the change in side effects of endocrine treatment and overall quality of life during and 3 months after cessation, in patients who completed at least 5 years of treatment.

Methods

We included 101 patients from 2 oncological practices who underwent curative treatment for breast cancer and whose adjuvant endocrine therapy ended between 2013 and 2016. Patients willing to cooperate filled out a questionnaire before and 3 months after stopping their endocrine therapy. Hot flashes, joint pain, muscle pain and fatigue were scored as absent, a little, severe or very severe. Overall quality of life was scored on a scale from 0 to 10.

Results

101 patients were included. Average was 61 years. Tumors were T1-T4, N0- N3, M0. Most patients received tamoxifen for 2-3 years, followed by an aromatase-inhibitor for 3-6 years. The main finding of this survey is that overall quality of life improved significantly after stopping endocrine therapy from 6.9 (range 4-10) to 7.7 (5-10) (p < 0.01 Wilcoxon paired rank test). 22 women improved ≥2 points. Patients who scored high on muscle aches and joint complaints improved the most.

Conclusions

Even patients who completed at least five years of endocrine treatment suffer side effects up to the end of treatment. After cessation these ameliorate in many, and this improves quality of life significantly. These findings are relevant when deciding on extended adjuvant endocrine treatment in individual patients. Detailed analysis will be presented.

Clinical trial identification

Under Dutch law no obligations for protocol submission for this type of survey, only institutional approval.

Legal entity responsible for the study

E.W. Muller

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

181P - Use and effectiveness of adjuvant ovarian function suppression (OFS) in premenopausal women with early breast cancer (ID 4121)

Presentation Number
181P
Lecture Time
13:15 - 13:15
Speakers
  • A. Ferreira
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

OFS either in association with tamoxifen (TAM) or an aromatase inhibitor (AI) improved disease-free survival in young women (≤35) and in those premenopausal women at higher risk of recurrence. However, its survival benefit remains largely unknown. In this study we characterize real-world use of adjuvant OFS from 2006 to 2015 and analyze its overall survival (OS) impact.

Methods

Retrospective observational cohort study of premenopausal women with Stage I-III hormone receptor-positive (HR+) breast cancer treated at one of 5 large centers in Portugal and diagnosed from 2006-2015. Study outcomes were use of OFS and OS. Pearson’s Chi2 test, logistic regression and Cox proportional hazards models were used.

Results

Of 1717 eligible patients, 304 (17.3%) were treated with adjuvant OFS, of which 271 (15.4%) in combination with TAM and 33 (1.9%) with AI. Baseline characteristics differed by subgroups: patients treated with OFS were younger, had larger, less differentiated (grade III 16% vs 24% for OFS), more frequently HER2 positive (14% vs. 19% for OFS) tumors, and underwent more frequently mastectomy (48% vs 57% for OFS), radiotherapy (25% vs 31% for OFS) and (neo)adjuvant chemotherapy (73% vs 79% for OFS). Adjuvant OFS was used at least since 2006 with an increase in its use from 2014 onward (16% vs 25% since 2014), particularly for the combination with AI (0.4% vs 8% since 2014). In a multivariate model, characteristics associated with use of OFS included younger age and year of diagnosis ≥2014 (both p < 0.001). Median time on OFS was 25 mo. (interquartile range 21-27). With a median follow-up of 38 mo. (IQR 20-66) and after controlling for age at diagnosis, staging, histologic grade, HER2 status, use of (neo)adjuvant CT, type of surgery and year of diagnosis, patients treated with OFS had a better OS when compared to those not treated with OFS (adjusted-HR 0.44, 95% CI 0.19-0.96; p = 0.040). Absolute benefit at year 5 was 2.1% (93.2 [95% CI 90.8 – 94.9) vs 95.3 [95% CI 89.7 – 97.9]).

Conclusions

In the real-world setting, a quarter of premenopausal women with early breast cancer were already treated with adjuvant OFS in 2014. After a median follow-up of 3 years, adjuvant OFS showed an OS benefit.

Clinical trial identification

Not applicable.

Legal entity responsible for the study

Hospital de Santa Maria, Centro Hospitalar de Lisboa Norte

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

182P - A phase II randomised study of Adjuvant hypo-fractionated radiotherapy with concurrent vs sequential letrozole in post-menopausal women with hormone receptor positive breast cancer: Report of pulmonary toxicity and cosmetic outcome (ID 4446)

Presentation Number
182P
Lecture Time
13:15 - 13:15
Speakers
  • R. Upadhyay
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The sequence of hormonal therapy with adjuvant radiation(RT) is debated because of anticipated morbidity. We conducted a phase II study to evaluate feasibility and efficacy of concurrent and sequential letrozole along-with hypo-fractionated RT(HFRT).

Methods

A total of 50 Post-menopausal women with hormone-receptor positive, Stage I-III Breast cancer received adjuvant HFRT 42.5Gy/16fr/3weeks and were randomly assigned to either concurrent (arm A) or sequential letrozole (arm B). Letrozole was started 3 weeks before RT in the concurrent, and 3 weeks after RT in sequential group. Pulmonary toxicity was assessed by clinical examination, chest x-ray, pulmonary function tests and HRCT chest (if indicated) at baseline, at one and six months(m) post RT. Cosmetic outcome was reported in both arms with six parameters (Table) at 6 m post RT.

Results

A total of 48 patients(pts) were followed up for 6 m (25 in Arm A and 23 in Arm B). None of the pts developed acute pulmonary toxicities. Mean (R) FeV1 and FVC values at baseline, 1 and 6 m post RT were 1.8 l (1.6-1.9) and 2.2 l(2.1-2.4), 1.79 l(1.5-1.9) and 2.1 l(1.9-2.4) and 1.85 l(1.6-2) and 2.2 l(2-2.4) respectively, and were comparable. FeV1 and FVC remained within 80 to 120% of the baseline values in 37 pts (20 Arm A vs 17 Arm B, p = 0.5). FeV1 and FVC were reduced by more than 80% at 6 m in 3 pts of Arm A and 5 pts in Arm B, (p = 0.7), while this was improved by over 120% in 5 pts (2 vs 3, p = 1). RTOG grade 2-3 radiation dermatitis was seen in 33 pts (15 vs 18, p = 0.55) while 5 pts had grade 4 toxicity (2 vs 3, p = 1). There was no treatment interruption because of toxicity.

182P

Cosmetic Outcome
Mild change
Marked change
ARM AARM BARM AARM B
Breast Shrinkage6523
Breast Hardness22 10
Breast Swelling100 0
Change in Skin appearance9833
Self-breast assessment141355
Photographic breast assessment151532

Overall, 18 pts had excellent cosmesis (7 vs 11, p = 0.4) while 32 had good cosmesis. (18 vs 14, p = 0.4).

Conclusions

HFRT along-with concurrent Letrozole is well tolerated. However, patients are being followed to assess loco-regional disease control and late toxicities.

Legal entity responsible for the study

All India Institute of Medical Sciences

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

183P - Intrinsic tumor features underlying clinical subtype discordance in early breast cancer (ID 2365)

Presentation Number
183P
Lecture Time
13:15 - 13:15
Speakers
  • V. Kotoula
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Despite efforts for laboratory and method harmonization, discordant clinical subtypes for ER/PgR/HER2 that determine treatment selection for breast cancer patients in clinical practice, still pose a challenge.

Methods

We investigated the clinical relevance of discordant clinical subtypes and their clinicopathological and genotype characteristics (60-gene panel) in a series of 1427 breast cancer patients treated within 4 adjuvant trials (2 in the pre- and 2 in the post-trastuzumab era; recruitment period 1997 – 2012). Treatment decisions were based on local laboratory typing; all tumors were re-typed centrally. Disease-free survival was assessed.

Results

We observed 340 (23.8%) discordant tumors for ER/PgR and/or HER2, ranging from 30% in the oldest to 19% in the most recent trial (p = 0.004); Cohen’s K was 0.512 for all subtypes, 0.583 for ER/PgR and 0.687 for HER2. ER/PgR discordance was associated with ER (p < 0.001) and PgR (p = 0.017) heterogeneity, basal phenotype, as well as higher grade, TILs and Ki67 labeling (all p < 0.001). HER2 discordant tumors had lower HER2 gene and CEN17 copies, and lower HER2/CEN17 ratios (all p < 0.001). Triple-positive tumors were rarely (0.5%) retyped as triple-negative (TN). ER/PgR discordant tumors had mutation patterns resembling HER2+ and TN, e.g., inversed TP53 and PIK3CA mutation prevalence (p < 0.001). Mutation clustering and phylogenetic analysis distinguished between concordant ER+/PgR+/HER2- tumors (73% of all tumors) and all other subtypes, with strong associations between ER±/PgR±/HER2- and ER+/PgR+/HER2 ± (p < 0.001). More relapses were noticed in patients with ER/PgR and HER2 negative-to-positive cases who did not receive hormonotherapy and trastuzumab (multivariate p = 0.048 and p = 0.016, respectively), but not in positive-to-negative cases.

Conclusions

Apart from technical considerations, clinical subtype discordance may reflect the genetic background of breast cancers, which appear to evolve by deviating from the ER+/PgR+ status. Development and reporting of phenotypic surrogates predictive of discordance is needed for increasing diagnostic accuracy and appropriate treatment selection.

Clinical trial identification

N/A

Legal entity responsible for the study

Hellenic Cooperative Oncology Group (HeCOG)

Funding

HeCOG

Disclosure

H. Gogas: Advisory or consultancy role: Amgen, Bristol-Myers Squibb, MSD, Novartis, Roche. C. Papandreou: Honoraria and/or Advisory Role: Astellas Pharmaceuticals, AstraZeneca, Janssen Pharmaceutical, Merck S.A., Roche (Hellas) S.A., Sanofi-Aventis, Pfizer Hellas S.A., Merck Sharp & Dohme. G. Fountzilas: Honoraria: AstraZeneca. Consulting or Advisory Role: Pfizer, Sanofi, Roche. Stock ownership (an immediate family member): Ariad. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

184P - Distribution of genomically defined recurrence risk in luminal A and B breast tumors defined by inmunohistochemistry: A retrospective study in Spanish population (ID 1347)

Presentation Number
184P
Lecture Time
13:15 - 13:15
Speakers
  • S. Perez Ramirez
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Semiquantitative inmunohistochemical (IHC) expression of progesterone receptor (PR) adds prognostic value to the current IHC-based luminal A (LA) definition, such that patients (pts) with LA (Ki67 <14% and PR > 20%) tumors can be spared from adjuvant chemotherapy (CT). Oncotype Dx® (ODX) and MammaPrint® (MP) assays have been validated as predictors of CT benefit. This study assessed the distribution of recurrence risk in LA and LB breast tumors as defined by Ki67 and PR.

Methods

A retrospective analysis was performed in 889 T1-2, N0-Nmic, M0 tumors for which ODX or MP results and local pathology data were available. Ki67 was assessed by IHC (high ≥14% and low <14%). PR was assessed by semiquantitative IHC (PR low < or = 20%, high >20%). Histological grade was defined using the Nottingham grading system.

Results

Median age 54 years (18-77). All pts had HER2 negative tumors. Median tumor size 15 mm (2-88). Three hundred (33.7%) tumors were classified as LA and 589 (66.3%) as LB. Grade 1 tumors were higher in LA (27%) than in LB (19%) pts (p < 0,001). CT was first recommended in 137 pts (45.7%) with LA vs. 361 pts (61.3%) with LB tumors. ODX was performed in 432 (48.6%) pts and MP in 457 (51.4%). Recurrence risk distribution varied significantly between groups (p < 0.001). ODX: among LA pts, 71.4%, 25.7% and 2.9% had low, intermediate and high recurrence risk respectively; among LB pts respective values were 46.2%, 44.2% and 9.6%. MP: among LA pts, 81.2% and 18.8% had low and high recurrence risk, respectively; among LB pts respective values were 54.2% and 45.8% (Table). After test results CT was recommended to 61 pts (20.3%) with LA vs. 268 pts (45.5%) with LB tumors.

184P

Recurrence Risk (%)LA (n = 300)LB (n = 589)
ODX (n = 432)
Low71.446.2
Intermediate25.744.2
High2.99.6
MP (n = 457)
Low81.254.2
High18.845.8

Conclusions

There is a wide distribution of recurrence risk results between LA and LB tumors defined by Ki67 and PR which confirms the important role of gene-expression assays in adjuvant decision making. Of note about half of pts with LB tumors had low recurrence risk indicating minimal benefit from adjuvant CT.

Legal entity responsible for the study

Ministry of Health of the Community of Madrid (Spain)

Funding

Ministry of Health of the Community of Madrid (Spain)

Disclosure

S. López-Tarruella: Advisory role: Novartis, Pfizer, Celgene. Honoraria/lecturer: Novartis, Pfizer, AstraZeneca, Celgene, Roche. I. Márquez-Rodas: Advisory role: Bristol-Myers Squibb, MSD, Novartis, Roche, Pierre Fabre, Amgen and Bioncotech. M. Martin Jimenez: Holds a patent on the predictive value of the PAM50 gene profile assay. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

185P - The first report of multicenter validation study of 95-gene classifier, a multi-gene prognostic assay of estrogen receptor positive and node negative breast cancer patients (ID 1524)

Presentation Number
185P
Lecture Time
13:15 - 13:15
Speakers
  • T. Kinoshita
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

95-Gene Classifier (95GC; Curebest™ 95GC Breast) is one of the multi-gene prognostic assays of estrogen receptor (ER) positive and node negative breast cancer patients, developed 95 gene-set without overlap with that used in another RT-PCR based product (Oncotype DX®). According to the original paper, the prognostic capability of 95GC has been shown even in the “intermediate” patients by microarray-based simulation model, “Recurrence Online”, of above RT-PCR product. But still 95GC has been validated only using the data from single institute and public database. Here we report the result of the first multi-center validation study for this multi-gene assay.

Methods

ER-positive and T1-2/N0/M0 breast cancer patients who received only hormonal therapy, without chemotherapy, in adjuvant were enrolled retrospectively. For each patient, fresh frozen tissue was applied to the assay and the classification result of 95GC, “L” or “H”, was used for the validation on 5 year recurrence free survival (5Y-RFS).

Results

We analyzed 75 eligible cases out of 150 enrolled, and found 47 patients classified as “L” showed 96.5% of 5Y-RFS (95%CI:89.5-98.9) while 28 patients of “H” showed 79.1% of 5Y-RFS (95%CI:63.8-88.5). There was a statistically significant difference between RFS of “L” and “H” groups by Log-Rank test (p = 0.0017). Other factors having significant association with 95GC were histological grade (p = 0.0012), “Recurrence Online” (p < 0.001) and “PAM50” (p < 0.001). And the patients of histological grade 2, of intermediate group by “Recurrence Online” and of Luminal B by “PAM50” could be classified into “L” and “H” by 95GC with different trends of RFS.

Conclusions

95GC was well validated by this first multi-centered retrospective study on 5Y-RFS of ER-positive, node-negative patients who received only hormonal therapy in adjuvant. The result indicates the usefulness of this novel multi-gene assay, as it can classify target patients into 2 groups, “L” and “H”, according to the prognosis of 5Y-RFS.

Legal entity responsible for the study

Takayuki Kinoshita

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

186P - Breast cancer PAM50 subtypes: Correlation between RNA-Seq and multiplexed gene expression platforms (ID 2414)

Presentation Number
186P
Lecture Time
13:15 - 13:15
Speakers
  • A. Picornell
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Gene expression signatures are a key tool for decision-making in breast cancer. In 2000 Perou et al. identified 4 intrinsic subtypes of breast cancer from gene expression data: LumA, LumB, HER2-enriched and Basal-like. These breast cancer subtypes yielded a superior prognostic impact than classical immunohistochemistry factors. From the initial intrinsic subtype, a 50-gene signature was developed for subtype assignment. PAM50 is being successfully used in multiplexed gene expression platforms such as NanoString nCounter®, which is the basis for the Prosigna® test. The latter was approved for the risk of distant relapse estimation in postmenopausal women with hormone receptor+, node+/- early stage breast cancer patients; and is a daily-used tool assessing the need of adjuvant chemotherapy.

Methods

The analyses were performed in paraffin embedded tissues (FFPE) from 96 patients recruited in a multicenter, prospective, non-randomized triple negative breast cancer trial (NCT01560663). Pre-treatment core biopsies were performed following clinical practice guidelines and conserved as FFPE for further RNA extraction. PAM50 was performed on both NanoString nCounter® and RNA-Seq technologies. Subtype assignment was based on the nearest centroid classification following this procedure for both platforms.

Results

Subtype calling agreed on 96% of the cases (NanoString nCounter®/RNA-Seq discordances: 3 Basal-like/HER2-enriched and 1 HER2-enriched/LumA). Both the Spearman correlation to each of the centroids and the risk of recurrence (ROR) were above 0.89 in both platforms. Furthermore, the agreement on proliferation score reached up to 0.97. In addition, 82% of the individual PAM50 genes showed a correlation coefficient >0.80.

Conclusions

The RNA-Seq is a fundamental research tool for whole transcriptome analysis. However, it cannot be massively used in the daily clinical practice, due to its processing time requirements and economic costs. We demonstrated that the RNA-Seq technology provides similar results to the NanoString nCounter®, with the latter providing lower cost and more simplicity in its use.

Clinical trial identification

NCT01560663

Legal entity responsible for the study

Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM)

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

187P - Summary of head-to-head comparisons of patient (pt) risk classifications by the 21-gene recurrence score (RS) assay and other genomic assays for early breast cancer (EBC) (ID 4122)

Presentation Number
187P
Lecture Time
13:15 - 13:15
Speakers
  • Z. Varga
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Many genomic assays that assess recurrence risk in EBC are prognostic, but they differ in risk group stratification, which can affect clinical utility. Prospective outcomes of > 50K pts treated based on 21-gene RS results have shown that pts with low RS EBC can safely forgo chemotherapy. Because of its extensive validation and wide clinical use, the RS assay is a common comparator in head-to-head studies with other assays.

Methods

Published/presented studies of the RS assay performed on same tumor samples with Breast Cancer Index (BCI), EndoPredict (EP) or EP+clinical features (EPclin), MammaPrint (MMP), and/or Prosigna (ROR) assays were reviewed. Study findings were summarized descriptively.

Results

14 studies were found that compared the RS assay with BCI (1), BCI, EPclin, and ROR (1), EP/EPclin (2), MMP (6), and ROR (4). Overall discordance in risk stratification ranged from 43% to 66% between assays (Table). The RS assay classifies 12% of pts as high risk, compared with EP (63%), EPclin (48%), and MMP (46%), assays with low/high risk groups, and compared with BCI (16%) and ROR (33%), assays that, like the RS assay, use three risk groups.

187P

Discordancea Between the RS Assay and Other Assays
BCI
ROR
EP/EPclin
MMP
Studyb1-level2-levelOverall1-level2-levelOverall1-level2-levelOverall1-level2-levelOverall
Sestak 201637%5%42%
Bartlett 2016c40%10%50%
Alvarado 201537%10%46%
Dowsett 201341%3%43%
Sinn 201745%20%66%
Varga 201329%/29%18%/21%47%/50%
Clough 201338%19%57%
Denduluri 201134%25%58%
Maroun 201531%22%53%
Shivers 201326%19%44%

a. Overall=any discordance in risk classification between the RS assay and other; 1-level=discordance of one risk category (low ↔ intermediate or intermediate ↔ high); 2-level=discordance of two risk categories (low ↔ high). b. Four studies lacked risk classification information appropriate for inclusion in this table. c. Study used nonstandard RS cutoffs for the RS vs. MMP comparison.

Conclusions

The five most common genomic assays in clinical use for EBC risk-stratify pts differently and thus are not interchangeable. Of these, the RS assay classifies the smallest proportion of pts as high risk.

Clinical trial identification

N/A

Legal entity responsible for the study

Zsuzsanna Varga

Funding

Genomic Health

Disclosure

Z. Varga: Consultant/advisor: Genomic Health, Roche. P. Sinn: Advisor: Genomic Health. D. McCullough, A. Lau, M.C. Stöppler, F.L. Baehner, C. Chao: Employment and stock ownership: Genomic Health. A. Seidman: Consultant/speaker: Genomic Health.

Collapse
Poster display session Poster Display session

188P - The impact of Oncotype DX breast cancer assay results on clinical practice: A UK experience (ID 1944)

Presentation Number
188P
Lecture Time
13:15 - 13:15
Speakers
  • V. Crolley
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Gene expression profiling is increasingly being used by clinicians to help determine whether or not to offer adjuvant chemotherapy. Oncotype DX is a 21 gene panel developed to predict the risk of tumour recurrence in patients with oestrogen receptor (ER) positive, HER2 negative breast cancer. NICE has recommended its use for patients at intermediate risk of recurrence, where this information would help clinicians to assess the potential benefit of chemotherapy versus endocrine therapy alone. Our aim was to see how oncologists are interpreting Oncotype DX tests in their clinical practice.

Methods

Data on patient and tumour characteristics (size, grade, ER/PR/HER2/nodal status), Oncotype DX recurrence score, treatment options offered and treatment outcomes were collected from 14 cancer centres across the UK.

Results

Of the 628 patients tested, 317 (50%) were in the low risk category (recurrence score <18), 224 (36%) were intermediate risk (score 18-30) and 81 (13%) were high risk (≥31). Chemotherapy was recommended for 52 patients in the low risk group, and was discussed/offered to a further 21. Chemotherapy was recommended for 91.8% of high risk patients. For patients with intermediate Oncotype scores, chemotherapy was recommended for 101 patients (45.9%), the option of chemotherapy was discussed/offered to 31 (14.1%) and 88 (40.0%) were not offered chemotherapy. Overall, 160 patients (25.9%) received chemotherapy. Where oncologists recommended chemotherapy to patients (n = 231), 59.7% of patients went on to receive chemotherapy. Where oncologists had offered or discussed chemotherapy as an option (n = 58), 27.6% of patients went on to receive it. The most common regimes were FEC75x6 (23.9%), ECx6 (13.8%) and ECx4 (9.4%), with 13.2% of patients receiving 3rd generation chemotherapy (FEC/T, TC or EC/taxane); other regimes included ACx4, TCx4 and weekly paclitaxel.

Conclusions

Throughout the UK, about half of patients tested had low risk Oncotype scores and the majority (74.1%) of patients tested did not receive chemotherapy. The widest variation in clinical practice was observed in interpreting intermediate risk Oncotype results, and in the chemotherapy regimens offered.

Legal entity responsible for the study

Judy King

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

189P - Enhancing decision-making about adjuvant chemotherapy in ER+, HER2- early breast cancer (EBC) following EndoPredict testing (ID 2284)

Presentation Number
189P
Lecture Time
13:15 - 13:15
Speakers
  • L. Fallowfield
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Chemotherapy side-effects can be substantial. There is increasing recognition that with surgery, radiotherapy and hormone treatment (tmt), many patients (pts) derive no benefit from chemotherapy and experience only iatrogenic harm. Gene expression profiling tests can help refine recurrence risk and likely chemotherapy benefit. EndoPredict is a multigene test which includes clinico-pathologic parameters to produce an EPclin score classifying risks of distant recurrence as low or high for ER+ve HER2–ve pts treated with adjuvant endocrine tmt alone. We compared tmt decisions pre and post EndoPredict test results, pts’ anxiety, decisional conflict and oncologists’ confidence about decisions made.

Methods

14 oncologists in 7 UK hospitals saw 149 pts judged to have equivocal indications for chemotherapy. Pts and oncologists discussed provisional tmt decisions based on usual prognostic factors. These decisions were reconsidered when EPclin results were available. Pre and post-test pts completed Spielberger’s State/Trait Anxiety inventory (STAI) and a decision conflict scale (DCS). Oncologists additionally recorded:- basic clinical details, their agreement with, and confidence about tmt decisions (endocrine (E) therapy +/- chemotherapy(C)).

Results

66.7% pts with an initial E alone decision and a high risk result upgraded to E+C. 9.4% pts with initial E+C decisions and high risk results down-graded to E alone. None of 46 pts initially favouring E alone who were low risk changed decisions. 82.8% who initially wanted E+C and had low risk scores downgraded to E alone. Endopredict results increased oncologists’ confidence (8% ‘strongly agreed’ pre-test, 50% post-test). Oncologists neither agreeing nor disagreeing with decisions fell (24% to 5%). Anxiety was stable in pts with unchanged decisions. Pts whose tmt was downgraded had significantly lower anxiety scores (p < 0.01); those whose tmt was upgraded had increased scores (p < 0.001). Likewise overall uncertainty on DCS fell post-test (p < 0.023).

Conclusions

EndoPredict scores increased oncologists’ and pts’ decision-making confidence, generally improved the matching of risk with therapy decisions.

Clinical trial identification

ISRCTN69220108

Legal entity responsible for the study

David Bloomfield

Funding

Myriad Genetics

Disclosure

L. Fallowfield, D. Bloomfield: Educational grant from Myriad Genetics. J. Mackay: Consultancy role to Myriad Genetics and travel expenses. S. Hinde: Educational grant from Myriad Genetics. A. Moss: Consultancy for Myriad Genetics. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

190P - Circulating ESR1 mutations at the end of aromatase inhibitor adjuvant treatment and after relapse in breast cancer patients (ID 5199)

Presentation Number
190P
Lecture Time
13:15 - 13:15
Speakers
  • V. Allouchery
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Detection of ESR1 circulating mutations is associated with acquired resistance to aromatase inhibitor (AI) in metastatic breast cancer. Until now, the presence of ESR1 circulating mutations at the end of the adjuvant treatment by AI in early breast cancer had never been clearly established. In this context, the aim of the present study was to evaluate the ESR1 circulating mutation frequency at the end of adjuvant treatment in patients with a subsequent local or metastatic relapse.

Methods

This monocentric retrospective study was based on available stored plasmas and included all early breast cancer patients who completed at least 2 years of AI adjuvant treatment and experienced a documented relapse at least 6 months after the end of their treatment. ESR1 circulating mutations (D538G, Y537S/N/C) were detected by droplet digital PCR in plasma samples taken both at the end of adjuvant treatment and on AI progression in patients re-exposed to AI during the metastatic phase.

Results

A total of 39 patients were included, with a median adjuvant AI exposure of 60 months (range 41-85). One patient (2.6%) had a local relapse only, while all the others (97.4%) had a metastatic relapse during follow-up. Median delay between the end of adjuvant treatment and relapse was 25 months (range 6-71). No ESR1 circulating mutation was detectable at the end of AI adjuvant therapy. In contrast, among the 25 patients (64%) who progressed on AI during the metastatic setting, 17 plasma samples were available and 7 patients (41,2%) had a detectable mutation.

Conclusions

Our results highlighted that there is no emergence of circulating ESR1 mutation at the end of an AI-based adjuvant treatment in hormone receptor positive breast cancer patients. In contrast, and as expected, we showed that re-exposure to AI in the metastatic setting induced circulating mutation detection in a significant fraction of the patients. Our present findings point out the low interest in ESR1 circulating mutation detection during the adjuvant setting, even for patients that will relapse.

Legal entity responsible for the study

Centre Henri Becquerel

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

191P - Pathological proliferation score to predict genomic risk categories in early stage breast cancer (ID 2632)

Presentation Number
191P
Lecture Time
13:15 - 13:15
Speakers
  • A. Bulbul
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Five of the 16 cancer-related genes used to calculate the Recurrence score (RS) are proliferative genes. Appropriate utilization of an expensive test is important especially in areas of limited resources. A relatively inexpensive ‘Pathological Proliferative score’ (PrS) of a tumor may help group patients in risk categories correlating with the RS.

Methods

We retrospectively studied 205 patients with Lymph node negative, hormone receptor (HR) positive, HER2 negative status (ODX candidates) between1990-2015 treated across three rural community oncology practices. Proliferation score was calculated by combining tumor grade, visual mitotic score and Ki67 immunostaining (on a scale of 1-3, lowest score of 3; highest score of 9). Log-rank test was used for survival analysis.

Results

PrS correlated with ODX risk recurrence (p < 0.001, Fischer's Exact test) [Table]. PrS predicted FFP (p = 0.014) at 10 years with PrS (3-4) 96%±2%, PrS (5-7) 91%±5% and PrS > (7-9) 75%±1%. It did not predict PFS(p = 0.77), OS(p = 0.84). Type of adjuvant treatment or none did not affect Low Prs(3-4) 10 yr PFS (p = 0.18 and OS (p = 0.33). Int/High PrS (5-9) showed benefit with adjuvant hormonal therapy compared to none at 10-year OS (p = <0.001), PFS (p = 0.002) and FFP (p = 0.003). The 10 yr OS (p = 0.75), PFS (0.76) and FFP (p = 0.88) was not influenced by addition of adjuvant chemotherapy.

191P Correlation of ODX with PrS

Proliferative Score (n = 190)
3 – 4 (n = 119) 5 – 6 (n = 37) 7 – 9 (n = 40) p-value*
Genomic Risk <0.001
Low 41 (87%) 11 (58%) 2 (11%)
Intermedium 6 (13%) 7 (37%) 1 (5%)
High 0 (0%) 1 (5%) 16 (84%)
Unknown 72 18 21
Genomic Risk <0.001
Low 41 (87%) 11 (58%) 2 (11%)
Intermedium/High 6 (13%) 8 (42%) 17 (89%)
Unknown 72 18 21

p-value based on Fisher’s Exact test

Conclusions

PrS which may represent an inexpensive screening approach to identify patients with a low ODX RS that have excellent outcomes despite the type of adjuvant treatment. ODX testing is unlikely to re-categorize them. Higher (5-9) PrS was not predictive of chemotherapy benefit, unlike high ODX. Lack of standardization of Ki67 staining, retrospective nature of the study while important should be tested in an expanded and prospective setting.

Legal entity responsible for the study

Kymera Independent Physicians

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

192P - Population sizes of patients (pts) with node negative (N0), HR+, HER2− primary breast cancer (BC), using standard and TAILORx 21-gene recurrence score (RS) cut-off values (COV) (ID 3999)

Presentation Number
192P
Lecture Time
13:15 - 13:15
Speakers
  • J. Blohmer
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Whether to use adjuvant chemotherapy (CT) is a crucial decision for pts with HR+, HER2− primary BC. The 21-gene Recurrence Score® (RS) assay is validated to predict adjuvant CT benefit and risk of recurrence, using standard RS COV of < 18, 18-30, and ≥31. Of ∼59K N0/N1 pts from two real-world registries, those with RS < 18 (>50% of total) largely treated without CT had favorable 5-y outcomes. The prospective TAILORx and PlanB clinical trials showed excellent 5-y outcomes for RS < 11 and RS ≤ 11 subgroups, respectively, treated without CT. The AJCC now recommends use of RS < 11 to down-stage to 1A. The capacity to generalize regional study outcomes globally requires first that RS subgroup sizes are geographically consistent. Here, we analyze RS subgroup sizes across geographical regions using standard and TAILORx COV.

Methods

Pts with N0, HR+, HER− primary BC and RS results from 2004 to April 2017 were included (data from Genomic Health). Subgroup sizes were determined for RS < 11, 11-17, 18-25, 26-30, and ≥31 in the US, Germany, the UK, France, and the rest of the world (RoW).

Results

Of 609,247 unique RS records analyzed, 513,035 were from the US, 29,248 from EU countries, and 66,964 from RoW. The relative population sizes of RS subgroups were highly consistent across geographical regions. Deviations in percentages for each RS range were within ±3% (Table). Across all regions, >50% of pts had RS < 18.

192P

US (N = 513035)
UK (N = 10154)
Germany (N = 14856)
France (N = 4238)
RoW (N = 66964)
All regions (N = 609247)
RS groupn%n%n%n%n%n%
<111093962117751727141881819129891912769221
11-1717807035317131529536142634234993521146135
18-2513578326284428420728118528186222816264127
26-30355127848811618393950007429147
≥315427411151615147910416106854106453911

Conclusions

Our analysis revealed highly consistent RS subgroup classifications across geographic regions, mirroring observations from registry studies, suggesting that tumor biology as characterized by RS results does not vary by geography. Our findings therefore support the generalizability of outcomes-study results using standard or custom COV across geographic regions.

Clinical trial identification

N/A

Legal entity responsible for the study

Jens-Uwe Blohmer

Funding

Genomic Health

Disclosure

M. Verrill: Speaker/advisory/research support: Amgen, Genomic Health, Novartis/GSK, Roche. Speaker/advisor: AstraZeneca, Eisai, Teva. Advisor/research support: Pfizer. Advisor: Merck. C. Fortmann, C. Chao: Employment and stock ownership: Genomic Health. J. Gligorov: Advisor: Eisai, Genomic Health, Novartis, Pfizer, Roche. Research support; Eisai, Roche. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

193P - Oncotype DX score, menopausal status and body mass index (ID 4596)

Presentation Number
193P
Lecture Time
13:15 - 13:15
Speakers
  • S. Picardo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The Oncotype DX assay uses expression of 21 genes to predict the risk of distant disease recurrence in patients with oestrogen receptor positive Her2 negative breast cancer. It has been demonstrated that increased intra-tumoural inflammation is associated with higher Oncotype score. An inflammatory milieu within the breast is associated with more aggressive histological cancer subtypes, and also with obesity, which is recognised as a risk factor for breast cancer. The association between obesity and breast cancer is higher in post-menopausal than pre-menopausal women. Therefore this study aimed to investigate whether there is a correlation between Oncotype score, menopausal status and obesity.

Methods

All patients with Oncotype assays performed between 2008 and 2016 in several centres in Ireland were identified. Data was retrospectively collected on weight, body mass index (BMI), menopausal status and Oncotype scores, including oestrogen (ER) and progesterone (PR) scores. Statistical analysis assessed correlation between Oncotype scores and these factors.

Results

Analysis was performed on 269 patients from a single centre with early stage breast cancer. Median age was 53.4 years and the majority of patients (58%) were post-menopausal at diagnosis. Patients who had a BMI less than 25 (normal weight) had significantly lower Oncotype score than those with BMI greater than 25 (overweight/obese) (p < 0.05). The Oncotype ER and PR scores were similar in normal weight and overweight patients, as were tumour size and grade. Post-menopausal patients had higher Oncotype scores than pre-menopausal patients (p < 0.001), with higher ER scores (p < 0.001) and lower PR scores (p < 0.001). On multivariate analysis, menopausal status remained significant as a predictor of Oncotype score. Complete analysis of an additional 600 patients will be performed.

Conclusions

Oncotype score is higher in overweight patients with early stage node-negative breast cancer. This difference appears to be independent of ER and PR scores and correlates with menopausal status. This retrospective study is the first to suggest that the body mass index and menopausal status of patients with early stage breast cancer may influence the Oncotype DX recurrence score and analysis of a further cohort of patients is ongoing.

Legal entity responsible for the study

Seamus O’Reilly

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

194P - Comparisons of tumor-infiltrating lymphocytes and 21-gene recurrence score in ER-positive/HER2-negative breast cancer (ID 1730)

Presentation Number
194P
Lecture Time
13:15 - 13:15
Speakers
  • S. Ahn
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Recent meta-analysis showed that tumors with high tumor-infiltrating lymphocyte (TIL) have a higher probability of pathologic complete response even in Luminal/HER2-negative breast cancer. Also, the 21-gene recurrence score (RS) predicts the clinical benefit of chemotherapy for ER-positive/HER2-negative women. We compared two markers in those cancer.

Methods

In ER-positive/HER2-negative patients treated with primary surgery, the RS (Oncotype DX® Breast Cancer Assay; Genomic Health, Inc., USA) was obtained. We evaluated TIL in H&E slides of surgical specimens by standardized methodology proposed by the international TIL-working group. In 198 women, the percentage of stromal TIL was successfully assessed. In accordance with the recent meta-analysis, the degree of TILs were categorized as high ( > =60%), intermediate (11-59%), and low ( < =10%).

Results

Ninety-seven (49.0%), 88 (44.4%), and 13 patients (6.6%) had low, intermediate, and high TILs, respectively. There is a significant but weak correlation between continuous RS and continuous TIL (Pearson’s R = 0.201, P = 0.004). The average of continuous RS was significantly highest in the high TIL tumors (17.8±10.7 in low TIL, 19.4±8.7 in intermediate TIL, and 26.2±8.2 in high TIL; P = 0.014). Whereas the average of continuous TILs was compared according to categorized RS, it was significantly higher in the intermediate RS or the high RS tumors (15.4±13.2 in low RS, 26.6±13.6 in intermediate RS, and 19.8±19.2 in high RS; P < 0.001). When we compared categorized RS and TIL, we found that the rates of the high TIL-tumors was significantly higher in the intermediate RS or the high RS (1.0% for low RS tumors, 12.5% for intermediate RS tumors, and 10.0% for high RS tumors; P = 0.007).

Conclusions

We found that tumors with high TIL tend to have a higher RS in ER-positive/HER2-negative breast cancer. We also noted that the rate of high-TIL tumors was significantly higher in the intermediate-RS tumors as well as in the high-RS tumors. Clinically, our findings suggest that TIL count might be referred in decision-making of chemotherapy in the intermediate RS-patients.

194P

Low or Intermediate TILHigh TILP-vlaue
Low RS (N = 98)97 (99%)1 (1%)0.007
Intermediate RS (N = 80)70 (88%)10 (12%)
High RS (N = 20)18 (90%)2 (10%)

Legal entity responsible for the study

N/A

Funding

This research was supported by the Basic Science Research Program through the NRF, funded by the Ministry of Science, ICT, & Future Planning (NRF-2015R1C1A1A02037104), and grant from the National R&D Program for Cancer Control, Ministry of Health & Welfare, Republic of Korea (1520120).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

195P - The 70-gene signature in node positive breast cancer: 10-year follow-up of the observational RASTER study (ID 4213)

Presentation Number
195P
Lecture Time
13:15 - 13:15
Speakers
  • S. Vliek
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In early stage breast cancer patients, with axillary lymph node metastasis, the 70-gene signature, MammaPrint® (MP) identifies patients with a High or Low Risk of distant breast cancer (BC) recurrence. For MP Low Risk (G(enomic)-low) in patients with up to 3 positive lymph nodes (N1-3), the MINDACT trial (Cardoso, NEJM 2016) showed that it might be safe to forgo adjuvant chemotherapy. Here we evaluated the prognostic value of MP at 10 years follow-up in patients with lymph node positive early stage BC.

Methods

Between 2004 and 2006 812 women with early stage BC participated in the observational RASTER trial (Bueno de Mesquita, Lancet Oncol, 2007). 181 patients were node positive and not included in the primary analysis, 176 of them gave consent for future research. On 164 tumor samples (FFPE) MP was performed retrospectively. Survival data was collected and samples were allocated to clinical high (C-high) or C-low risk as used in MINDACT. Patients with over 3 axillary lymph node metastases (N4+) were all considered C-high. 10-year distant-recurrence-free-interval (DRFI) was compared between subgroups based on the MP and clinical assessment.

Results

In 3 patients the clinical assesment could not be determined. Over 95% of patients received chemotherapy, 82.9% (136/164) of tumors were ER-positive and 18.3% (30/164) of patients had N4+. MP identified 47% (n = 77/164) as Low Risk, including 16,9% (13/77) with N4+. 10-year DRFI in patients N1-3 and G-Low or G-High was 94.9% and 80.7% respectively (HR 4.7; 95%CI 1.3-16.2). With the clinical assessment 13.7% (n = 22/161) were low risk, only one was diagnosed with distant BC recurrence. 10-years DRFI was 94.4% in C-low and 85.8% in C-high (HR 3.7 95%CI 0.5-28.5). In N4+ 10-years DRFI was 69.7%. Combining the clinical assessment with MP risk assessment in patients N1-3 the 10-years DRFI in clinical high risk patients was 95.2% for G-Low (n = 44) and 79.6% for G-High (n = 65) (HR 4.83 95%CI 1.1-21.4).

Conclusions

We again confirm the prognostic value of Mammaprintin BC patients with axillary lymph node involvement after 10 years follow up. In N1-3 patients with clinical high risk, MP can identify a subgroup with excellent prognosis after standard adjuvant systemic therapy.

Legal entity responsible for the study

S. Linn

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

196P - Higher expression of estrogen response genes in the primary tumor is associated with a greater risk for late recurrence in patients with ER+/HER2-breast cancer (ID 1364)

Presentation Number
196P
Lecture Time
13:15 - 13:15
Speakers
  • R. Bense
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In patients with ER+ breast cancer, 50% of recurrences occur > 5 years after diagnosis (i.e. late recurrence). Clinical trials report contradicting results on the effect of extended endocrine therapy > 5 years to reduce late recurrence risk. Using publically available breast cancer gene expression profiles, we aimed to gain insight into the biology that increases the risk for late recurrences.

Methods

Gene expression profiles of primary ER+/HER2- tumors of breast cancer patients were collected with disease-free survival (DFS) data, defined as time of diagnosis to local recurrence or distant metastasis. We defined (i) a group containing all patients (n = 2,231), (ii) a group that received 5 years of endocrine therapy only (n = 591), and (iii) a group that received no systemic therapy (n = 497). For each group, genes were ranked on their association with DFS as determined by multivariate Cox regression with age, tumor size, grade, lymph node status, and therapy as covariates. Gene set enrichment analysis (GSEA) was performed on these gene lists with the Hallmark collection from the Molecular Signatures Database. Within each group, associations with early recurrence were studied in all patients with censoring at 5 years if no event occurred < 5 years after diagnosis (set I). To study associations with late recurrence, a second set was defined that contained only patients with a follow-up ≥ 5 years and no event < 5 years after diagnosis (set II).

Results

Within all patients and the group that received 5 years of endocrine treatment only, higher expression of genes belonging to the Hallmark ‘estrogen response late’ was associated with longer DFS in set I and shorter DFS in set II. This Hallmark contains estrogen responsive genes identified in estradiol treated ER+ breast cancer cell lines. However, in patients who received no systemic treatment, higher expression of these genes was associated with shorter DFS in both set I and II.

Conclusions

Higher expression of estrogen response genes is associated with a greater risk for late recurrence in patients with ER+/HER2- breast cancer. Potentially, patients with ER+ tumors with high expression of these genes might benefit most from extended endocrine therapy.

Legal entity responsible for the study

R.S.N. Fehrmann

Funding

Dutch Cancer Society grants RUG 2010-4739 and RUG 2013-5960, NWO-VENI grant (916-16025) and a Mandema Stipendium.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

197P - Understanding BRCA1 and BRCA2 mutated breast cancer cases in Romania: First report on founder mutations in Romanians (ID 2670)

Presentation Number
197P
Lecture Time
13:15 - 13:15
Speakers
  • A. Eniu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

First systematic analysis of BRCA 1(B1) or BRCA2(B2) mutations in high-risk Romanian breast cancer patients (pts) aiming at defining founder mutations.

Methods

This prospective study evaluated the germline B1/B2 mutations in 250 high-risk breast cancer pts tested between 02.2015-12.2016 at IOCN. Inclusion criteria selected pts diagnosed with triple negative breast cancer under the age of 50, or having conventional family history criteria. All pts signed an informed consent. B1/B2 testing was performed using an AmpliSeq-based sequencing analysis, on the Ion Torrent Personal Genome Machine at RCFG. Pathogenic mutations were validated using Sanger technology. MLPA was performed for all pts.

Results

Of the 250 pts with breast cancer, 44 (17.6%) carried pathogenic mutations, 29 pts (11.6%) in B1 and 15 (6%)in B2, while 18 patients (7.2%) carried a Variant of Uncertain Significance (VUS). Patient features analysis confirmed the prevalence of younger age, higher grade, hormone receptor negative and Her2 negative status among mutated patients (data not shown). Out of the 16 distinct deleterious mutations identified, 7 (43.75%) occurred in B1 and 9 (56.25%) in B2. The founder mutations identified in B1 gene were: c.5329_5330insC (c.5266dupC) 11 pts (37.93%), c.3607C>T 9 pts (31.03%) and c.181T>G 4 pts (13.79%). Other B1 mutations where c.1687C>T 2 pts (6.89%), and c.4218delG (3.44%), c.212 + 1G>T (3.44%), c.68_69delAG (3.44%) in one patient respectively. For B2 gene, c.9371A>T (46.66%) was identified as founder mutation (7 pts, 46.66%). Other mutations were found each in one patient (6.66%): c.1528G>T, c.4022C>G, c.7007G>A, c.8695C>T, c.9253delA, c.8680C>T, c.8755-1G>A, c.8695C>T. Of the founder mutations identified, two (c.3607C>T and c.9371A>T) have not been previously identified as founder mutations in any Eastern European country.

Conclusions

This prospective study presents the first extensive results of germline B1/B2 mutations in Romanian high-risk breast cancer pts. Our results indicate that at least four recurrent B1/B2 mutations qualify as founder mutations; two being newly identified as carrying a founder effect. ClinicalTrials.gov Identifier: NCT02317120.

Clinical trial identification

NCT02317120

Legal entity responsible for the study

Alexandru Eniu

Funding

AstraZeneca

Disclosure

A. Eniu: Research support: AstraZeneca, Roche, Novartis, Celltrion. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

198P - The prevalence of CD146 expression in breast cancer subtypes and its relation to outcome (ID 3680)

Presentation Number
198P
Lecture Time
13:15 - 13:15
Speakers
  • I. De Kruijff
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

CD146 has several putative (patho)physiological roles in breast cancer. The most prominent is its involvement in the induction of epithelial-to-mesenchymal transition, which might have an effect on cancer phenotype and aggressiveness. Here, we investigated the prevalence of CD146 expression and its prognostic role in breast cancer subtypes.

Methods

In total, 1,025 breast cancer patients were available for this retrospective study. From all patients, formalin-fixed paraffin-embedded primary breast cancer tissue was collected and embedded in tissue microarrays, which were stained for CD146. CD146 expression was defined as > 1% of the tumor cells showing CD146 membrane staining. Clinical data were available from all patients (median follow up 118 months, range 4-120). For subtype analysis the Pearson chi-square test was used and the Cox proportional hazards model for survival analyses. Only patients who were lymph node negative and did not receive (neo)adjuvant systemic treatment were included in the survival analyses (n = 551).

Results

113 (11%) out of 1,025 tumors showed CD146 expression. Of these, 43% of the tumors had > 50% of the tumor cells showing CD146 membrane staining. From the molecular subtypes, CD146 positive tumors are often of the triple negative subtype (76 out of 119 (64%), p < 0.001) and histologically of the medullary type (11 out of 23 (48%), p < 0.001). In univariable analysis, CD146 was a prognostic factor for both poor metastasis-free survival (MFS) and overall survival (OS) (respectively HR 1.65, 95% CI 1.02-2.66, p = 0.041 and HR 1.66, 95% CI 1.03-2.69, p = 0.037). When correcting for the traditional prognostic factors (including age, tumor size and grade, ER, PR and HER2) in multivariable analysis, CD146 was not an independent prognostic factor for MFS and OS (respectively HR 1.63, 95% CI 0.93-2.87, p = 0.088 and HR 1.46, 95% CI 0.82-2.61, p = 0.197).

Conclusions

CD146 protein expression is present in 11% of the primary breast cancer tumors and is most prevalent in the triple negative and medullary subtypes. CD146 is a prognostic factor for MFS and OS in breast cancer patients, but it is not independent of the traditional prognostic factors. Its potential impact on outcome to systemic treatment such as endocrine therapy, remains to be established.

Legal entity responsible for the study

Erasmus University Medical Center

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

199P - Neutrophil-to-lymphocyte and lymphocyte-to-monocyte ratios in breast cancer (ID 4613)

Presentation Number
199P
Lecture Time
13:15 - 13:15
Speakers
  • L. Gerratana
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Immunity plays a pivotal role in cancer progression and prognosis. A high neutrophil-to-lymphocite ratio (NLR) or a low lymphocyte-to-monocyte ratio (LMR) are respectively associated with systemic inflammation and immune suppression and have been associated with a poor outcome. Aim of this study is to further investigate the interaction between the immune system and breast cancer (BC) through the NLR and LMR.

Methods

This retrospective study analyzed a consecutive cohort of 657 patients (pts) with a diagnosis of pT1 BC, without restrictions regarding lymph node status (T1BC), or metastatic BC (MBC) treated between 2004 and 2017 at the Department of Oncology of Udine (Italy). Differences in terms of NLR and LMR among the two cohorts and between clinico-pathological characteristics in the T1BC subgroup were explored through the Kruskal-Wallis test. The prognostic impact in terms of OS in the T1BC population was investigated through uni- and multivariate Cox regression.

Results

Both NLR and LMR were significantly different between the T1BC and the MBC cohorts. In particular, pts with T1BC had a higher median LMR (3.9 vs 2.9; P = 0.0001) and lower NLR (2 vs 2.7; P = 0.0001). After stratification according to molecular profile, T1BC and MBC cohorts of Luminal B-like subtype were significantly different in terms of both LMR (4.2 vs 3; P = 0.0001) and NLR (2 vs 2.5; P = 0.0001). In triple negative subtype, the difference between TB1C and MBC was observed for NLR (1.9 vs 3.2; P = 0.0272) only. On the other hand, no differences between TB1C and MBC were highlighted for the other subtypes. When focusing on the clinico-pathological characteristics of the T1BC cohort, LMR was associated with progesterone receptor (PR) expression (P = 0.0261) and marginally with the estrogen receptor (ER) expression, while NLR with tumor diameter (P = 0.0240) and marginally with grading. Furthermore, among T1BC pts, NLR had no prognostic impact in terms of OS, while LMR was associated with a better outcome also when corrected for ER, PR and HER2 status (HR 0.44, 95%CI 0.28 - 0.71, P = 0.001).

Conclusions

These results suggest a role for systemic inflammation and immune-suppression in breast cancer, especially in the triple negative and luminal B-like subtypes.

Legal entity responsible for the study

University of Udine

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

200P - Vitamin D as a prognostic factor in triple negative early breast cancer (ID 4847)

Presentation Number
200P
Lecture Time
13:15 - 13:15
Speakers
  • S. Morales
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Triple negative breast cancer remains without a target therapy. Interventions that could improve pathological complete response (pCR) rates are required. Metabolits of vitamin D could be involved in chemotherapy response.

Methods

A series of 147 patients with early or locally advanced triple negative breast cancers was retrospectively analyzed from 2007 to 2016. Patients from 2015 to 2016 period were supplemented with vitamin D and calcium (880UI/1000mg). Analysis of clinicopathological, immune variables and vitamin D pathway were correlated to pCR.

Results

Median age was 53, median tumor size 30mm, 48% had nodal involvement, and median ki67 expression was of 70%. Androgen receptor was expressed in 28% of tumors analyzed, EGFR in 89%, CK5/6 in 63%. Mean stromal T lymphocytes infiltrates (sTILS) was of 28%, mean PDL1 expression of 128, mean 53BP1 expression of 125, and mean VDRnuc expression of 132. pCR rate was of 40%, and within patients with vitamin D supplementation was 64% (16/25). Only VDRnuc expression was associated with pCR (p = 0.047) in the univariate and multivariate analysis. Patients with high expression of VDRnuc in tumor had no evidence of relapse (p = 0.024), with similar curves than those who achieve pCR (p = 0.000).

Conclusions

VDRnuc expression is a strong predictive (p = 0.047 with pCR) and prognostic (p = 0.024 with relapse) in triple negative breast cancer. Role of supplementation needs to be tested if it could improve VDRnuc levels; whereas in our series patients with supplementation had better pCR rates.

Legal entity responsible for the study

Hospital Universitari Arnau de Vilanova de Lleida Institut de Recerca Biomèdica

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

201P - CDK12: New breast and ovarian cancer predisposition gene in Tatar population? (ID 4995)

Presentation Number
201P
Lecture Time
13:15 - 13:15
Speakers
  • E. Shagimardanova
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The development of hereditary ovarian and breast cancer (OC/BC) is often caused by genetic defects in the DNA repair system. However, the diagnostics in most medical centers of Russia includes PCR-based identification only the eight common mutations in BRCA1/2 for the Slavic population. Previously we established that patients of the Tatar population with OC/BC did not possess most of Slavic mutation and a significant part of the predisposition is due to other mutations in the genes of the homologous recombination (HR) system. The aim of this work is the analysis of the germline mutations in the HR genes.

Methods

The DNA from 175 blood samples from patients of the Volga District with hereditary OC/BC were analyzed by targeted NGS (Roche NimbleGen, Illumina MiSeq), the comparison groups included blood samples from patients of Slavic origin.

Results

62% of the detected pathogenic mutations were presented in the BRCA1/2 genes. The remaining mutations were found in other genes of the reparation system (HGMD Professional 2017.1 database). An unexpected finding was the detection of a germline splicing mutation c.1047-2A>G in CDK12 gene (Chr17(GRCh37):37627130A>G, NM_016507.3) in patients of Tatar origin (Table).

201P CDK12 c.1047-2A>G frequency distribution

Subjects origin BC or OC Healthy controls
Tatar from Volga District 6/94 (6.4%) 0/32 (0%)
Non-Tatar from Volga District 1/81 (1.2%)
Slavic from Moscow 0/49 (0%) 0/284 (0%)
ExAC NFE 29/64446 (0.045%)
1000G 2/5006 (0.039%)

Mutation c.1047-2A>G is more common in patients with OC/BC in comparison with healthy controls (7/224 vs 0/316, p = 0.002, OR = 21.49, CI 95% = 1.22–377.25).

Conclusions

Gene CDK12 is one of the most frequently altered genes in serous ovarian cancers, but significance of CDK12 germline mutations in hereditary cancers remains to be defined. Its role in carcinogenesis of OC was established recently and CDK12 was not included in most NGS panels of HR genes. Our study demonstrates that CDK12 may be novel candidate gene for OC/BC genetic predisposition. Notably, frequency of CDK12 c.1047-2A>G (6.4%) mutation is comparable with frequency of founder-mutation BRCA1 5382insC (7.4%), that indicates its possible founder role in Tatar population.

Legal entity responsible for the study

Tatarstan Cancer Center

Funding

Kazan (Volga Region) Federal University, Tatarstan Cancer Center

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

202P - Prognostic value of master transcriptional regulators (MTRs) in early stage breast cancer (ID 4844)

Presentation Number
202P
Lecture Time
13:15 - 13:15
Speakers
  • S. Barron
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Multigene prognostic signatures (MGPS) enable identification of candidate patients (pts) for treatment de-escalation in early stage BC. However, currently available MGPS do not completely address clinical needs by adequately incorporating lymph node (LN)-positive pts and clinicopathological information (CPI). Here, we present OncoMasTR, a MGPS for determining the risk of distant recurrence (DR) in ER-positive, HER2-negative BC pts with up to 3 involved LNs. OncoMasTR, discovered via a novel network analysis methodology that determines upstream MTRs has been mechanistically verified and offers improved prognostic value compared to existing MGPS. OncoMasTR has been further trained to include LN-positive pts and CPI.

Background

Multigene prognostic signatures (MGPS) enable identification of candidate patients (pts) for treatment de-escalation in early stage BC. However, currently available MGPS do not completely address clinical needs by adequately incorporating lymph node (LN)-positive pts and clinicopathological information (CPI). Here, we present OncoMasTR, a MGPS for determining the risk of distant recurrence (DR) in ER-positive, HER2-negative BC pts with up to 3 involved LNs. OncoMasTR, discovered via a novel network analysis methodology that determines upstream MTRs has been mechanistically verified and offers improved prognostic value compared to existing MGPS. OncoMasTR has been further trained to include LN-positive pts and CPI.

Methods

Two independent sample sets: 225 pts from Malmö University Hospital and 106 pts from Skåne University Hospital were used for training, cross-validation and refinement of OncoMasTR. RNA extracted from 225 archived tissues was analysed by RT-qPCR and expression levels of the MTRs were determined by normalising against the expression levels of reference genes. The strongest prognostic combinations of MTRs were identified using statistical models of all possible combinations of MTRs. Clinical performance of the models with the best cross-validated performance in the training data were further evaluated in the 106 independent samples.

Methods

Two independent sample sets: 225 pts from Malmö University Hospital and 106 pts from Skåne University Hospital were used for training, cross-validation and refinement of OncoMasTR. RNA extracted from 225 archived tissues was analysed by RT-qPCR and expression levels of the MTRs were determined by normalising against the expression levels of reference genes. The strongest prognostic combinations of MTRs were identified using statistical models of all possible combinations of MTRs. Clinical performance of the models with the best cross-validated performance in the training data were further evaluated in the 106 independent samples.

Expected Results

OncoMasTR classifies up to 70% of LN0 pts and 56.2% of LN0-3 pts as low risk, with only 5% and 4.9% recurrence rate within the respective groups. When incorporating selected CPI, its performance further improved to identifying X% of pts as low risk, with Y% of recurrence in the group. Results showed that the OncoMasTR Molecular score (mS) alone adds statistically significant information to the CPI, and the Combined score (cS) also adds statistically significant information to the mS.

Results

OncoMasTR classifies up to 72% of LN0 pts and 60% of LN0-3 pts as low risk, with only 4.9% and 5.5% recurrence rate within the respective groups. When incorporating selected CPI, its prognostic performance further improved to a concordance index of above 0.8. Results showed that the OncoMasTR Molecular score (mS) alone adds statistically significant information to the CPI, and the Combined score (cS) also adds statistically significant information to the mS.

Expected Conclusions

OncoMasTR offers significant prognostic information to the standard CPI and addresses the unmet clinical need of LN-positive pts. The binary output giving no ambiguous intermediate group helps eliminating uncertainty in the formation of final treatment decision. OncoMasTR is ready for large scale clinical validation and, subsequently, clinical translation.

Conclusions

OncoMasTR offers significant prognostic information to the standard CPI and addresses the unmet clinical need of LN-positive pts. The binary output of OncoMasTR, giving no ambiguous intermediate group helps eliminate uncertainty in the formation of the final treatment decision. OncoMasTR is ready for large-scale clinical validation and, subsequently, clinical translation.

Legal entity responsible for the study

OncoMark Ltd

Funding

OncoMark Ltd.

Disclosure

S. Barron, B. Moran, C-J.A. Wang, T. Loughman, B. Fender, P. Dynoodt, C. Lopez-Ruiz: Employee of OncoMark Ltd. W. Gallagher: Employee of OncoMark Ltd, has stock or ownership of OncoMark Ltd, is a co-inventor of the patent licensed to OncoMark Ltd and received travel, accommodation and expense from OncoMark Ltd. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

203P - Survival outcomes of all patients treated with breast carcinosarcoma at a UK specialist cancer centre over a 10 year period (2004–2014) (ID 5379)

Presentation Number
203P
Lecture Time
13:15 - 13:15
Speakers
  • M. Alameddine
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Carcinosarcoma of the breast is a rare and aggressive type of breast cancer presenting as a high grade tumour with lower rates of both lymph node metastasis and oestrogen and progesterone receptor (ER/PR) expression when compared to the more common types of breast cancer, carrying a less favourable prognosis. We present the clinical and pathological findings and outcomes of a series of patients diagnosed and treated for breast carcinosarcoma at a UK specialist cancer centre.

Methods

We conducted a retrospective review of data for all patients diagnosed with breast carcinosarcoma between October 2004 and October 2014 at the Clatterbridge Cancer Centre NHS Foundation Trust.

Results

Nine patients were diagnosed in the 10-year period, with a median age at diagnosis of 73 years (range 37-76 years). Seven patients (77.8%) were postmenopausal. Six patients (66.7%) presented with a palpable mass. T1, T2, and T3 were found in 1, 6 and 2 patients respectively. N0, N1, and N2 were found in 6, 2 and 1 patients respectively. All patients had G3 disease with a median diameter of 3cm (range 1.9-9.0 cm). Oestrogen receptor (ER) and progesterone receptor (PR) were both negative in 8 patients (88.9%). Whilst one patient had wide local excision, all the rest had mastectomy, of whom 4 had axillary nodal clearance and 4 had sentinel nodal biopsy. Five patients received adjuvant radiotherapy. Adjuvant chemotherapy was delivered to 5 patients (2 patients received neither treatments) and adjuvant hormone therapy was delivered to 2 patients (one of whom had a concurrent contralateral ER/PR positive tumour). Patients were followed up for a median period of 15 months (range 1-60 months). Median DFS is estimated to be 25 months and median OS is estimated to be 49 months (95% CI: 14-84 months). Two patients (22.2%) developed metastases with a DFS time of 13 and 14 months respectively, and both died within 5 months.

Conclusions

Within a 10 year period during which our specialist cancer centre were referred 16,500 new breast cancer patients, only 9 patients had carcinosarcoma. Prognosis following recurrence is poor within our limited cohort, in agreement with the published literature. In order for more meaningful analysis of survival outcomes for such a rare form of breast cancer, a multicentre collaborative approach is required.

Clinical trial identification

not applicable

Legal entity responsible for the study

Clatterbridge Cancer Centre

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

204P - ESR1, Ph-mTOR, CDK4/6 and PD-L1 expression as prognostic (and potentially druggable) drivers for pure invasive lobular breast carcinoma (ILC): Preliminary results of prognostic outliers according to a clinical-pathological model (ID 3611)

Presentation Number
204P
Lecture Time
13:15 - 13:15
Speakers
  • L. Carbognin
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The biological drivers of prognosis for pure ILC are not entirely clear. The aim of this analysis was to investigate the molecular and immune-related portrait of prognostic outliers to identify different patterns of expression associated with prognosis and potentially druggable.

Methods

Clinical-pathological multi-center data of resected early-stage pure ILC patients (pts) were correlated to disease-free and overall-survival (DFS/OS). A continuous score was derived according to multivariate Hazard Ratios, in order to derive a 3-class model (Poor/Intermediate/Good Prognosis). IHC (for Ph-mTOR, CDK6, PD-L1), FISH (for ESR1, Ph-mTOR, CDK4, PD-L1) and H&E evaluation (for stromal Tumor Infiltrating Lymphocytes, sTILs) were performed upon pts at Poor and Good Prognosis. Odds Ratio (OR) with 95% CIs for the risk of association with prognostic class of biomarkers was determined.

Results

Data from 457 pts were gathered (median age 57 years, median follow: 75 months). The 3-class cross-validated model significantly differentiated DFS and OS (p < 0.0001, prognostic accuracy: 0.65 and 0.71, respectively). Based on DFS, 154 and 20 pts with Good and Poor prognosis, respectively, were identified. The preliminary and exploratory analysis of the first 34 pts (Good/Poor 14/20) is reported (OR < 1: higher chance to be associated with Good prognosis; OR > 1 higher chance to be associated with Poor prognosis).

204P

Biomarker [Method]OR95% CIs
High sTILs [H&E]0.220.01-5.8
Ph-mTOR deletion [FISH]0.330.07-1.49
ESR1 gain [FISH]0.530.09-3.18
PD-L1 positive [IHC]0.670.08-5.4
CDK4 deletion [FISH]1.220.1-15.1
PD-L1 gain [FISH]1.250.19-8.2
Score 3+ CDK6 [IHC]2.290.21-24.68
Score 3+ Ph-mTOR [IHC]2.480.61-10.05
ESR1 deletion [FISH]2.750.27-23.04
CDK4 gain [FISH]3.180.15-66.36

Conclusions

Despite unpowered, these preliminary data suggest that Poor and Good prognosis are potentially associated to differential expression of a cluster of biomarkers: ESR1 deletion, CDK4 gain, CDK6 and ph-mTOR over-expression versus high sTILs, PD-L1 positive, ESR1 gain and ph-mTOR deletion, respectively.

Clinical trial identification

Not applicable.

Legal entity responsible for the study

University of Verona

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

205P - The pregnancy and fertility (PREFER) study: A prospective cohort study on fertility-preserving (FP) strategies in young early breast cancer (EBC) patients (pts) (ID 2331)

Presentation Number
205P
Lecture Time
13:15 - 13:15
Speakers
  • C. Dellepiane
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Premature ovarian failure and subsequent infertility are possible long-term side effects of chemotherapy (CT) in young EBC pts. Limited data are available on the number of pts who consider FP strategies and on the reasons for refusal of these procedures. To address the significant challenges related to fertility issues, the PREFER study was developed as a national comprehensive program aiming to optimize care and improve knowledge around this topic.

Methods

This is a prospective cohort study ongoing across several Italian centers affiliated to the GIM (Gruppo Italiano Mammella) study group. Oncologists offer the available FP strategies to young EBC pts undergoing (neo)adjuvant CT: oocyte cryopreservation (OC), ovarian tissue cryopreservation (OTC) and LHRH analogue (LHRHa) during CT. Eligible pts are premenopausal, ≤ 45 years, no previously exposed to CT and/or radiotherapy. Primary objective is to obtain data about preferences and choices of young EBC pts on the FP strategies. Secondary objectives are to evaluate the success and safety of FP strategies, hormonal changes during CT and survival outcomes. The present analysis reports preliminary results of the study including pts enrolled at the coordinating center from November 2012 to May 2017.

Results

A total of 131 EBC pts were enrolled; median age was 38.9 years (24.8-45.34). Nine pts (6.87%) refused all FP options. Reasons for refusal were no interest in fertility preservation (5 pts), previous pregnancy (3 pts), no interest in having children (1 pts). LHRHa was accepted by 120 pts (91.6%) and 27 pts (20.6%) accepted gynecologic counseling. Among these pts, 10 (7.6%) accepted OC or OTC. Main reason for refusal of cryopreservation procedures was fear of delaying cancer treatment (3 pts). No complications were observed among women who underwent OC or OTC. Median number of mature oocytes yielded and cryopreserved was 8.5 (4-13). A patient had a spontaneous pregnancy following adjuvant treatment.

Conclusions

Despite the great importance of fertility issues in young EBC pts, a minority of them (7.6%) require to access cryopreservation procedures. This is crucial information from a public health perspective and for resource allocation.

Clinical trial identification

NCT02895165

Legal entity responsible for the study

IRCCS AOU San Martino IST, Genoa Italy

Funding

AIRC - Associazione Italiana per la Ricerca sul Cancro (IG 2013 No14272).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

206P - Incidence of permanent alopecia following adjuvant chemotherapy in women with early stage breast cancer (ID 5191)

Presentation Number
206P
Lecture Time
13:15 - 13:15
Speakers
  • J. Crown
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Alopecia is one of the most distressing toxicities of adjuvant chemotherapy for patients with breast cancer. Historically, oncologists have reassured patients (pts) that chemotherapy-induced alopecia is temporary, and followed by full hair recovery. More recently there have been troubling reports of permanent alopecia following adjuvant taxanes (Tax). We studied the incidence of long-term hair loss in patients treated on adjuvant trials in our institution. Patients who were enrolled on clinical trials involving Tax (D-Docetaxel, P-Paclitaxel) and/or Anthracyclines (A) were included.

Methods

We conducted a telephone interview survey of pts who had completed adjuvant or neo adjuvant A and/or T chemotherapy on clinical trials more than one year before. Ongoing alopecia was graded as 0 (full hair recovery), 1 (mild hair loss) or 2(severe/total). The study was approved by the hospital audit committee.

Results

We studied 295 pts who has been treated on 12 studies. Drug exposure: D-260 pts (D nonA-185, D+A-75); A-nonTax-12 pts; A+P 23 pts. The overall incidence of alopecia was 15% (11% grade 1 and 4% grade 2). For all D the incidence was 15% (12% Grade 1 and 3% Grade 2). For D+A-24% (19% Grade 1 and 5% Grade 2). For D non A the incidence was 13% (8% grade 1 and 5% grade 2). For A non T 8% (Grade 2-8%). For PA-13% (4% grade 1 and 9% grade 2). For patients receiving D non-A regimens, there were two levels of D exposure, 300mg/m2 (90 pts) or 450 mg/m 2 (95 pts). The incidence of alopecia was significantly D dose dependent: D300- 7% (all grade 1) and D 450-19% (14% grade 1, 5% grade 2) (p=.02 chi2) . Among the higher dose D group, the companion drug choices carboplatin for HER2 positive, (55 pts) or cyclophosphamide (40 pts) were associated with similar incidences of permanent alopecia (22% v 16%).

Conclusions

Permanent alopecia is a common complication of adjuvant chemotherapy. The risk appears to be highest in regimens which contain A and D, but it is also seen in D non-A, AP and in A non-Tax. For patients receiving D non A, the risk is dose-dependent. Our set contains few P pts, and no pts undergoing low dose weekly P. Oncologists should warn all patients undergoing adjuvant therapy of the risk of permanent alopecia.

Legal entity responsible for the study

John Crown

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

207P - Significant changes in dietary intake and physical activity after breast cancer diagnosis in a Chinese breast cancer cohort study (ID 2000)

Presentation Number
207P
Lecture Time
13:15 - 13:15
Speakers
  • Y. Lei
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The diagnosis of cancer can motivate survivors to modify lifestyles, typically involving diet and physical activity. Few data have reported pre- and post-diagnostic lifestyle habits in Chinese population.

Methods

In an on-going prospective cohort study which involved 1462 Chinese women with early-stage breast cancer, we evaluated dietary intake and lifestyle factors pre- and post- breast cancer diagnosis. Validated food frequency questionnaires were used to evaluate dietary intake. Leisure time physical activity was measured by a modified Chinese Baecke questionnaire. This report compared changes of dietary intake and physical activity between 12 months before and 18 months after diagnosis of breast cancer.

Results

Intake of whole grains, refined grains, fruits, vegetables, eggs, and nuts increased significantly post-diagnosis (Range, 54%-72% increase; P < 0.001, each; Table). Conversely, after diagnosis consumption of red meat, processed meat, poultry, dairy products, soy, sugar drinks, and coffee significantly decreased (Range, 32%-63% decrease; P < 0.001, each). The level of physical activity (MET-hour per week) post-diagnosis was significantly increased (median, 0.75 vs. 5.25; P < 0.001), with 58% of patients became more physically active. However, the proportion of women that met the WCRF/AICR Cancer Prevention recommendations about physical activity (10 MET-hour per week) was still low, only 21% women met at pre-diagnosis, increasing to 34% at post-diagnosis.

207P

Food groupPre-diagnosis [Median (IQR), g/1000kcal/day] Pre-diagnosis [Median (IQR), g/1000kcal/day]
Refined grains269.9 (213.3-348.3)319.6 (257.8-391.2)
Whole grains3.6 (0-11.9)7.4 (0.7-30.0)
Fruits95.1 (54.3-137.8)140.4 (92.0-193.6)
Vegetables194.1 (136.8-263.2)271.5 (197.3-359.8)
Eggs8.3 (4.1-14.8)10.1 (5.2-16.9)
Red meat48.3 (29.1-72.7)38.2 (20.3-62.0)
Processed meat1.7 (0.3-4.8)0.4 (0-2.4)
Poultry20.4 (9.6-38.2)1.1 (0-7.1)
Dairy products15.4 (5.0-44.5)5.7 (0.6-15.9)
Soy28.1 (12.8-55.1)23.3 (8.3-49.0)

IQR, inter quartile range.

Conclusions

In this cohort study, Chinese breast cancer patients reported significant changes in dietary intake and increased physical activity level and a higher proportion met the WCRF/AICR Cancer Prevention recommendations after cancer diagnosis. These findings provided crucial information on lifestyle behaviors in Chinese breast cancer survivors, and provided information to healthcare professionals on survivors’ health and quality of life.

Legal entity responsible for the study

The Chinese University of Hong Kong

Funding

World Cancer Research Fund International (Grant Number WCRF 2010/249 and WCRF 2014/1197).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

208P - Prevention of cardiotoxicity in breast cancer patients: A randomized prospective study (ID 4348)

Presentation Number
208P
Lecture Time
13:15 - 13:15
Speakers
  • A. Elkhateeb
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Adjuvant anthracyclin chemotherapy (ANTC) and trastusumab are documented to prolong survival in breast cancer patients. However, these drugs are well known to induce Left Ventricular Systolic Dysfunction (LVSD). Multiple studies showed that acetyl choline-esterase inhibitor (ACEIs) and beta Blockers (BBs) can prevent LVSD.

Methods

One hundred and twenty six patients with M0 breast cancer patients to be treated with ANTC ± trastuzumab were randomized to an intervention group (group 1) (n = 63 patients) which received cardioprotective drugs; (ACEI;enalapril) and (BB;carvedilol) or to a control group which did not receive cardioprotective drugs (group 2) (n = 63 patients). To evaluate systolic and diastolic functions conventional echocardiography (Simpson method and M- mode) and cardiac magnetic resonance imaging (CMR) were performed at baseline, after 3 cycles and 6 cycles of ANCT, and after 1 year of follow-up. Cardioprotective drugs received: Both enalapril and carvedilol were started at least 24 hours before the first chemotherapy cycle.

Results

In the intervention group 58 patients had 3 cycles ANTC, 6 patients received 6 cycles ANTC, and 12 patients received trastuzumab. Whereas in the control group 47 patients had 3 cycles ANTC, 16 patients were given 6 cycles ANTC and 18 patients received trastusumab. After 3 ANTC cycles, LVEF did not change in group 1 (64.35% at baseline vs. 63.59%, p 0.2) but decreased by M- mode in the control group (64.84% at baseline vs. 63.42%, p 0.03) associated with statistically significant deterioration of diastolic function grades. At 1 year follow-up, while no change was observed in LVEF in group 1, there was decrease in LVEF by CMR in group 2 (65.78% at baseline, 61.48% at 1 year, p 0.048). No cases were detected with heart failure or with final EF < 45% in either group. Compared to controls, the intervention group had a statistically significant lower incidence of decrease EF ≥ 10% after finishing ANTC by CMR(1.9% vs. 12.5%, p 0.04).

Conclusions

Combined prophylaxis with ACEI (enalapril) and BB (carvedilol) may prevent LVSD in patients with non-metastatic breast cancer treated with anthracyclines containing chemotherapy ± trastuzumab. The clinical relevance of this strategy should be confirmed in larger randomized studies.

Legal entity responsible for the study

Academic Group

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

209P - Quality-of-life results from a randomized, phase-II-study of the therapeutic cancer vaccine L-BLP25 (Stimuvax®) in the preoperative treatment of women with primary breast cancer (ABCSG-34) (ID 3270)

Presentation Number
209P
Lecture Time
13:15 - 13:15
Speakers
  • V. Bjelic-Radisic
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In ABCSG-34, patients with HER2-negative breast cancer were randomized to preoperative standard of care therapy (SoC) with or without L-BLP25 (Stimuvax). This report describes the quality-of-life (QoL) results of the trial.

Methods

400 patients were randomized to receive SoC with or without BLP25. Postmenopausal women with low-risk disease (ER +++/ ++, Ki67 <14%, G1/2) received 6 months of letrozole; premenopausal and postmenopausal patients with triple-negative, ER-/+/ ++, Ki67 ≥14%, or G3 tumors received 4 cycles of epirubicin/cyclophosphamide plus 4 cycles of docetaxel with or without L-BLP25. Primary end point results RCB and pCR rates were presented previously (SABCS 2016; Abstract Nr 850339). QoL was assessed with the EORTC QLQ-C30 and EORTC QLQ-BR 23 at baseline, before surgery, and up to 4 weeks thereafter. The objective was to evaluate differences of QoL in women treated with or without L-BLP25, as well as between the two regimens.

Results

385 patients from 17 centers were included in the QoL analysis. There were no differences in QoL between patients receiving SoC only and those receiving additional L-BLP25. Impact on QoL was determined by the SoC therapy and by the timepoint of the assessment. Before surgery and 4 weeks thereafter patients receiving chemotherapy ± TL-BPL25 showed more impairment in the QoL scales role and social functioning, financial problems and body image than the patients receiving endocrine therapy ± TL-BPL25. Fatigue and hair loss were significantly more common in the chemotherapy than in the endocrine arm. At the time of surgery and thereafter, patients in both SoC arms had significantly negatively impacted QoL (physical, role, emotional, cognitive, social, sexual functioning, body image domains) as well as more fatigue, pain, dyspnoea, breast and arm symptoms. There were no differences in global health status between the arms at the different time points.

Conclusions

Addition of L-BLP25 (Stimuvax) to SoC in HER2-negative EBC patients does not impair QoL.

Legal entity responsible for the study

ABCSG

Funding

This study was supported by Merck KGaA, Darmstadt, Germany Role of the Funder/Sponsor: ABCSG was the regulatory sponsor of this trial. Merck provided financial funding and the study drug (IMP). The study was designed and conducted by ABCSG. Merck was not involved in collection, management, analysis, and interpretation of the data. ABCSG prepared and approved the manuscript. All co-authors have decided to submit the manuscript for publication. This study was supported by Merck KGaA, Darmstadt, Germany Role of the Funder/Sponsor: ABCSG was the regulatory sponsor of this trial. Merck provided financial funding and the study drug (IMP). The study was designed and conducted by ABCSG. Merck was not involved in collection, management, analysis, and interpretation of the data. ABCSG prepared and approved the manuscript. All co-authors have decided to submit the manuscript for publication. This study was supported by Merck KGaA, Darmstadt, Germany Role of the Funder/Sponsor: ABCSG was the regulatory sponsor of this trial. Merck provided financial funding and the study drug (IMP). The study was designed and conducted by ABCSG. Merck was not involved in collection, management, analysis, and interpretation of the data. ABCSG prepared and approved the manuscript. All co-authors have decided to submit the manuscript for publication. ABCSG was the regulatory Sponsor of this Trial. Merck KGaA provided financial funding and teh study drug (IMP). The study was designed and conducted by ABCSG.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

210P - Dedicated breast PET for predicting residual disease after breast cancer neoadjuvant chemotherapy (ID 1472)

Presentation Number
210P
Lecture Time
13:15 - 13:15
Speakers
  • S. Sasada
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Diagnostic methods to evaluate the response to breast cancer neoadjuvant chemotherapy (NAC) have been established. Dedicated breast PET (DbPET) is a high-resolution molecular breast imaging method, and we investigated the ability of DbPET to predict the presence of residual primary tumors after NAC, compared with whole-body PET (WBPET).

Methods

Forty-five patients (47 tumors) underwent WBPET and ring-type DbPET after NAC, and tumors were completely resected between January 2016 and March 2017. The pathological response was classified as complete remission (ypT0), residual intraductal disease (ypTis), or residual invasive disease (ypT≥1). Standardized uptake value (SUV) and tumor-to-normal tissue ratio (TNR) were assessed.

Results

Twelve patients achieved ypT0 and 5 developed ypTis. DbPET detected all cases of ypTis, and WBPET detected only one case of ypTis. The sensitivity, specificity, and accuracy of WBPET for ypT≥is were 54.3%, 83.3%, and 61.7%, respectively, and those of DbPET were 77.1%, 83.3%, and 78.7%, respectively. In the ypT0/ypTis/ypT≥1 groups, the median WBPET-SUV, DbPET-SUV, and DbPET-TNR were 1.0/0.9/1.1, 1.7/1.8/2.2, and 1.0/1.6/1.7 (P = .134, .077, and .008), respectively (Table). Area under the curves of WBPET-SUV, DbPET-SUV, and DbPET-TNR for predicting ypT≥is were 0.610, 0.648, and 0.807, respectively.

210P Comparison of predicting indexes for predicting pathological response

Pathological response WBPET-SUV
DbPET-SUV
DbPET-TNR
Median (IQR)p Median (IQR)p Median (IQR)p
ypT≥11.1 (0.9-1.7).1342.2 (1.5-3.9).0771.7 (1.1-2.9).008
ypTis0.9 (0.9-1.2)1.8 (1.4-1.9)1.6 (1.4-1.8)
ypT01.0 (0.9-1.0)1.7 (1.5-2.1)1.0 (0.9-1.1)

Conclusions

DbPET was superior to detect residual primary tumors, especially noninvasive carcinoma, after NAC than WBPET. TNR was expected as the better parameter of pathological evaluation than SUV.

Legal entity responsible for the study

N/A

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

212TiP - IMpassion031: A phase III study comparing neoadjuvant atezolizumab (atezo) vs placebo in combination with anthracycline/nab-paclitaxel (nab-pac)–based chemotherapy in early triple-negative breast cancer (eTNBC) (ID 2291)

Presentation Number
212TiP
Lecture Time
13:15 - 13:15
Speakers
  • E. Mittendorf
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Atezo is an anti–programmed death-ligand 1 (PD-L1) monoclonal antibody that prevents PD-L1 from binding to PD-1 and B7.1 receptors, thereby restoring tumor-specific immunity. TNBC is characterized by PD-L1 expression on tumor-infiltrating immune cells (IC), a high mutation rate and high levels of tumor-infiltrating lymphocytes (TILs), suggesting a therapeutic opportunity for atezo. Atezo alone and in combination with nab-pac is well tolerated, with promising activity in metastatic TNBC, supporting its investigation in early-stage disease. IMpassion031, a global Phase III, double-blind, randomized, multicenter, placebo-controlled study, is being conducted to evaluate the efficacy and safety of neoadjuvant treatment with nab-pac → doxorubicin + cyclophosphamide and either atezo or placebo in invasive stage II/III eTNBC. The choice and sequence of chemotherapy is selected to maximize the opportunity to establish a robust immune response.

Trial design

Patients (pts) with previously untreated, central laboratory–confirmed invasive TNBC with primary tumor size > 2 cm and ECOG PS 0-1 are eligible. Exclusion criteria include history of invasive BC, stage IV disease, and prior immunotherapy or autoimmune disease. Approximately 204 pts will be randomized 1:1 to receive atezo (840 mg q2w) or placebo with nab-pac (125 mg/m2 qw) for 12 weeks, followed by atezo (840 mg q2w) or placebo with doxorubicin (60 mg/m2 q2w) + cyclophosphamide (600 mg/m2 q2w) for 4 cycles before surgery. Pts will be unblinded post-surgery and pts in the atezo arm will continue to receive atezo (1200 mg q3w × 11 doses). Stratification factors include stage II vs III at diagnosis and PD-L1 expression (IC0 vs IC1/2/3). The primary endpoint is pathological CR (pCR); key secondary endpoints include pCR according to PD-L1 status, pt-reported outcomes, event-free survival and overall survival. Tumor samples will be taken at baseline, on treatment (optional), at surgery and post-recurrence and will be assessed for biomarkers associated with responses and immune escape.

Clinical trial identification

NCT number available on poster

Legal entity responsible for the study

F. Hoffmann-La Roche Ltd.

Funding

F. Hoffmann-La Roche Ltd.

Disclosure

E.A. Mittendorf: Financial support to the institution from the following to conduct clinical trials that I serve as the Principal Investigator for: AstraZeneca, EMD Serono, Galena BioPharma, Genentech. Does not personally receive any funding. C.H. Barrios: Research: Pfizer, Novartis, Amgen, AstraZeneca, Boehringer Ingelheim, Roche, Lilly, Sanofi, GSK, Taiho, Mylan, Merrimack, Merck, Astellas, Bristol-Myers Squibb. Consulting: Boehringer Ingelheim, GSK, Novartis, Pfizer, Roche-Genentech, Eisai. N. Harbeck: My COI is available for ESMO on their internal website. D. Miles: Honoraria for Advisory Boards from Roche-Genentech. S. Saji: Honoraria from AstraZeneca, Chugai Pharmaceutical, Eisai, Novartis Pharma, and research funds from AstraZeneca and Chugai Pharmaceutical. H. Zhang: Consultant for Genentech/Roche from 2015. A-N. Duc: Roche employee and stock. S. Rafii: Employed by Roche. C. Lai: Currently employed by Genentech/Roche and hold company stock. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

213TiP - PIONEER- Pre-operative wIndOw study of letrozole plus PR agonist megestrol acetate versus letrozole aloNE in post-menopausal patients with ER-positive breast cancer (ID 2601)

Presentation Number
213TiP
Lecture Time
13:15 - 13:15
Speakers
  • S. Kumar
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Recent preclinical findings have been published which provide new insights into the functional cross-talk between the estrogen receptor (ER) and the progesterone receptor (PR) in breast cancer (Mohammed et al., Nature, 2015). Addition of a PR agonist to anti-estrogens directly modifies ERa chromatin binding and the transcriptional response in breast cancer cells, and is anti-proliferative in vitro and in vivo. Megestrol Acetate (MA), an off-patent semi-synthetic derivative of progesterone, has been licensed for many years as a treatment for ER+ metastatic breast cancer. There is also good evidence for the effectiveness of MA as a supportive therapy to ameliorate endocrine therapy-related hot flushes.

Trial design

PIONEER is a three-arm, open label, multi-centre randomised phase II pre-surgical window trial evaluating effects of 15 days of preoperative therapy with Letrozole (LET), or LET plus MA 40mg, or LET plus MA 160mg in postmenopausal women with newly diagnosed, ER+ HER2- invasive primary breast cancer. Patients are being recruited in Cambridge, with 5-6 other UK sites due to open in q3/4 2017.

213TiP 3-arm randomisation

Arm ALET 2.5mg daily
Arm BLET 2.5mg daily + MA 40mg daily
Arm CLET 2.5mg daily + MA 160mg daily

The primary endpoint is % change in proliferation between baseline and day 15 tumour biopsies, measured by Ki67 immunohistochemical (IHC) assessment. Secondary endpoints include: expression of Aurora Kinase A, Caspase 3 and Androgen receptor/PR/EMT markers by IHC; and safety endpoints. Exploratory endpoints include: transcription factor mapping (ChIP-seq) on paired fresh-frozen tumour samples. Patients are randomised in a 1: 1.5: 1.5 ratio for arms A: B: C. Based on results from previous clinical trials, a mean 66% reduction in Ki67 is anticipated for LET alone (arm A), and a 77.5% reduction for combination arms B and C, based on preclinical data. A recruitment total of 189 patients is required. Pioneer will help determine if there is value in conducting a follow-on adjuvant study to investigate the longer term benefit of combining an aromatase inhibitor with MA, and if so, at what dose (40mg vs. 160mg).

Clinical trial identification

EudraCT Number: 2016-003752-79 MHRA/REC number: v2.0 5th June 2017

Legal entity responsible for the study

Cambridge University Hospitals NHS Foundation Trust and the University of Cambridge

Funding

Anticancer Fund

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

214TiP - VENTANA (SOLTI-1501): Antiproliferative effect of the addition of oral metronomic vinorelbine to endocrine therapy in luminal/HER2-negative early breast cancer: A window of opportunity trial (ID 3583)

Presentation Number
214TiP
Lecture Time
13:15 - 13:15
Speakers
  • B. Adamo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The cornerstone of luminal breast tumors treatment both in early and advanced settings is endocrine therapy (ET). Although extended adjuvant ET has demonstrated benefit, a significant percentage of patients relapse. In previous studies, the combination of ET with CDK4/6 inhibitors has shown unprecedented efficacy suggesting that inhibition of the cell cycle in combination with ET is a strategy to keep exploring. The efficacy and safety of metronomic VNB have been confirmed in preclinical and clinical studies and it is now considered a multi-mechanisms of action therapy that could offer advantages when combined with other drugs. VENTANA study is a “window-of-opportunity” trial designed to explore whether, similarly to CDK4/6 inhibitors, oral metronomic VNB in combination with endocrine therapy induces a superior antiproliferative effect than ET alone. We hypothesize that the synergistic biological effect of the combined treatment could be an alternative to CDK4/6 inhibitors in the treatment of luminal breast cancer patients.

Trial design

Pts are randomized (1:1:1) to receive LET 2.5mg daily, oral VNB 50mg 3 days a week, or the combination. After 3 weeks of treatment, pts undergo surgery. Pre- and post-treatment (surgical) samples will be analyzed for gene expression. The primary objective is to test if oral metronomic VNB and LET induce a superior anti-proliferative effect than either drug alone in pts with early BC defined as Luminal by PAM50. This will be evaluated by the expression of 11 proliferative genes contained in the PAM50 subtype predictor (BIRC5, CCNB1, CDC20, CDCA1, CEP55, KNTC2, MKI67, PTTG1, RRM2, TYMS and UBE2C) as surrogate signature biomarker of its anticancer activity. In addition, 560 BC-related gene signatures will also be analyzed. Enrollment started in July 2016 in 10 sites across Spain. To date, 47 patients have been included. We expect to report full study results by Spring 2018.

Clinical trial identification

NCT02802748

Legal entity responsible for the study

SOLTI Breast Cancer Research Group

Funding

Pierre Fabre

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

215TiP - PALLAS: PALbociclib CoLlaborative adjuvant study: A randomized phase 3 trial of palbociclib with standard adjuvant endocrine therapy versus standard adjuvant endocrine therapy alone for HR+/HER2- early breast cancer (ID 3769)

Presentation Number
215TiP
Lecture Time
13:15 - 13:15
Speakers
  • E. Mayer
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Cell cycle inhibition is a proven target for novel cancer therapeutics. Palbociclib (P) is an orally active inhibitor of CDK4/6, and arrests the cell cycle at the G1-S transition. P in combination with endocrine therapy (ET) has demonstrated efficacy in phase II and III randomized trials for patients with newly diagnosed and recurrent hormone receptor positive/HER2 negative (HR+/HER2-) metastatic breast cancer (MBC), and is approved in these settings. Given confirmed benefits of P and ET for MBC, the PALLAS study was designed to determine if the addition of P to adjuvant ET improves outcomes over ET alone in HR+/HER2- early breast cancer.

Trial design

PALLAS is an international open-label phase III trial randomizing (1:1) patients (pts) to 2 years of P (125 mg daily, 21 days on 7 days off in a 28-day cycle) combined with at least 5 years of provider choice ET (AI, tamoxifen, +/- LHRH agonist), versus ET alone. The primary objective of the study is to compare invasive disease-free survival (iDFS) for the combination of P and ET, versus ET alone. Secondary objectives include comparison of iDFS excluding cancer of non-breast origin, DRFS, LRRFS, OS, as well as safety. The principal objective of the translational investigations is to determine the predictive or prognostic utility of defined genomic subgroups with respect to iDFS and OS. Additional objectives include evaluation of cfDNA and tissue biomarkers predictive of benefit or resistance, pharmacogenomics, adherence, and patient-reported QOL. Eligible pts are pre- or post-menopausal women or men with stage II-III, HR+/HER2- breast cancer. Patients may have already initiated ET, but must be randomized within 12 months of diagnosis and 6 months of initiation of adjuvant ET. Trial sample size is 4600 pts and stage IIA pts will be capped at a total accrual of 1000 pts. Interim analyses for safety, futility/efficacy and sample size re-estimation are planned. PALLAS opened in 9/2015 and accrual is ongoing. Contact information: emayer@partners.org.

Clinical trial identification

US: IND Nr.(FDA): 126003 clinicaltrials.gov NCT02513394 FDA approval 27 May 2015 Non-US (clinicaltrialsregister.eu dates): EudraCT Number: 2014-005181-30 Sponsor Protocol Number: AFT-05/ABCSG-42/BIG_14-03 Start Date: 2015-07-09

Legal entity responsible for the study

Alliance Foundation Trials (AFT) LLC for US, ABCSG GmbH for participating countries outside of the US

Funding

Pfizer

Disclosure

E.L. Mayer, H.J. Burstein, E. Winer: Research funding to the institution from Pfizer. A.M. Demichele: Funding to the institution for clinical trials from Pfizer, Novartis, Genentech and Calithera. Advisory board activities for Pfizer, Novartis and Calithera. M. Koehler, C. Huang Bartlett, X. Huang: Employee and shareholder of Pfizer. M. Gnant: Grants and/or personal fees from Sanofi-Aventis, Novartis, Roche, GlaxoSmithKline, AstraZeneca, Nanostring Technologies, Accelsiors, Pfizer, Smith Medical, outside the submitted work. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

216TiP - OlympiA: A randomized phase III trial of olaparib as adjuvant therapy in patients with high-risk HER2-negative breast cancer (BC) and a germline BRCA1/2 mutation (gBRCAm) (ID 5297)

Presentation Number
216TiP
Lecture Time
13:15 - 13:15
Speakers
  • A. Tutt
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In a Phase II study (NCT00494234), treatment with olaparib, a potent, orally available PARP inhibitor, exerted antitumor activity in patients (pts) with advanced BC harboring a gBRCAm. In a randomized phase III trial, olaparib significantly improved PFS compared to chemotherapy (CT) for patients with HER2-negative gBRCAm advanced BC (OlympiAD, NCT02000622, ASCO 2017). OlympiA (NCT02032823) is a phase III trial of olaparib as adjuvant therapy for pts with high risk gBRCAm HER2-negative BC who have completed local treatment and (neo)-adjuvant CT.

Trial design

OlympiA is a double-blind trial in which high risk HER2-negative pts are randomized (1:1) to receive treatment with olaparib (300 mg tablets bid [2 x 150 mg]) or placebo for 12 months. Eligible pts must have completed local treatment and at least 6 cycles of (neo)-adjuvant containing anthracyclines and/or taxanes. Pts with triple negative BC (TNBC) must have ≥pT2 or ≥pN1 in the adjuvant and non-pCR in the neoadjuvant setting. Pts with hormone receptor (HR) positive BC must have ≥4 positive lymph nodes in the adjuvant and non-pCR and CPS&EG score ≥3 in the neoadjuvant setting. Pts must also harbor a deleterious gBRCAm. Stratification factors include hormone receptor status, prior neoadjuvant versus adjuvant CT, and whether pts have received platinum therapy for current BC. The primary objective is invasive disease-free survival (IDFS). Efficacy assessments will be made by mammograms/breast MRI scans annually for 10 years, beginning 6 months from randomization, and by medical history/physical examination from randomization every 3 months for 2 years, then every 6 months for a further 3 years and annually thereafter. Secondary objectives include overall survival, distant DFS, incidence of new non-BCs, HRQoL, safety and tolerability. The primary IDFS analysis will be performed after 330 IDFS events using a stratified log-rank test. Patient enrolment began in April 2014 and is currently ongoing. The target number for randomization is 1500 patients across ∼500 sites and ∼25 countries worldwide. Support: U10CA12027,-69651,-37377,-69974,-180868,-180822, -189867; AstraZeneca.

Clinical trial identification

NCT00494234

Legal entity responsible for the study

AstraZeneca

Funding

AstraZeneca and NRG Oncology

Disclosure

A. Tutt: Grants/research support: AstraZeneca. C. Goessl: Employee, stock ownership: AstraZeneca. G. Viale: Advisory Board Member: AstraZeneca. D. Zardavas, A. Arahmani, D. Fumagalli: Research grants to BIG from AstraZeneca. P. Herbolsheimer, W. Wu: AstraZeneca employee. J. Constantino: Acts as the data center for the USA part of the OlympiA trial. While mainly funded by the US National Cancer Institute, the University receives funds from AstraZeneca for some aspects of OlympiA data center activity. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

BREAST CANCER, LOCALLY ADVANCED (ID 5674)

Lecture Time
13:15 - 13:15
Speakers
  • F. André
  • L. Yates
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15
Poster display session Poster Display session

218P - Prognostic estimates of Ki-67 percentage drop after neoadjuvant chemotherapy (NAC) in luminal B (lumB) and triple negative breast cancer (TNBC) (ID 2900)

Presentation Number
218P
Lecture Time
13:15 - 13:15
Speakers
  • J. Pascual
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Pathologic complete response (pCR) and residual cancer burden (RCB) after NAC are validated prognostic markers in BC. We assessed the impact of adding Ki-67% drop (baseline biopsy - surgery) to distant metastasis relapse-free survival (dRFS) models containing CP factors plus post-treatment stage (MDACC CPS score), estrogen receptor status and tumor grade (MDACC CPS+EG score).

Methods

Records from 341 patients (pts) with lumB/HER2 neg (baseline Ki67 >20%) or TNBC who received NAC from 2008 to 2015 at our hospital were reviewed. Uni- and multivariate Cox models were constructed and concordance-index (c-index) calculated.

Results

Pts: median age 47 years (24-83), 60% lumB and 40% TNBC, 62% stage 2, 38% stage 3. pCR: 12% lumB, 32% TNBC (p < 0.01). Median Ki-67% drop: 24% lumB, 5% TNBC (p < 0.01), without differences by NAC regimen. dRFS at 5 year-median follow-up was 75% in lumB (CI95% 67-83) vs 62% in TNBC (CI95% 53-74, p < 0.01); 90% in RCB 0/1 (CI95% 81-97) vs 74% in RCB 2/3 (CI95% 65-83, p < 0.01). As compared to pts with RCB 0/1, those with RCB 2/3 plus Ki-67% drop > = 20% (best cut-off in univariate model) had similar dRFS at 5 years (90%, CI95% 81-100, p = 0.48), irrespective of molecular group. Enrichment for Ki-67 > =20% drop in lumB (60%) vs TNBC (30%, p < 0.01) was observed. Both CPS and CPS+EG scores were validated as independent prognostic factors in univariate dRFS models (c-index of 0.70 and 0.78, respectively). The addition of Ki-67% drop (< 20% vs > = 20%) to CPS and CPS+EG scores in multivariable models significantly improved their performance (c-index of 0.74 and 0.81, respectively). Ki-67 > = 20% drop associates with 70-80% reduction in distant relapse risk (HR 0.27 and 0.16 in CPS and CPS+EG models, respectively, p < 0.05).

Conclusions

Our data support the addition of Ki-67% drop after NAC in lumB and TNBC to existing dRFS online outcome calculators. In the context of RCB 2/3, Ki-67 > = 20% drop is mainly seen in lumB/HER neg tumors. Importantly, Ki-67 < 20% drop identifies a high-risk population that may be eligible to clinical trials with novel therapeutic interventions in the adjuvant setting.

Legal entity responsible for the study

Vall d'Hebron University Hospital

Funding

Vall d'Hebron Institute of Oncology

Disclosure

M. Oliveira: Consulting or Advisory Role in Roche-Genentech and Puma Biotechnology. Research Funding in AstraZeneca and Genentech/Roche. R. Dienstmann: Consulting or Advisory Role in Roche-Genentech and in Astellas Pharma. Research Funding in Merck. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

219P - Immune function and response to neoadjuvant chemotherapy in hormone receptor positive, HER2-negative breast cancer (ID 4101)

Presentation Number
219P
Lecture Time
13:15 - 13:15
Speakers
  • A. Matikas
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Gene expression (GE) signatures and Tumor Infiltrating Lymphocyte (TILs) enumeration have shown promise as predictors of response to neoadjuvant chemotherapy in Hormone Receptor negative (HR-) and HER2+, but not in HR+/HER2- breast cancer (BC). This study aimed to explore their predictive value in HR+/HER2- BC, based on previous work from our group on the association of immune function and chemosensitivity in advanced HR+ BC.

Methods

The PROMIX phase 2 trial enrolled patients with locally advanced HER2- BC to receive six cycles of epirubicin and docetaxel, plus bevacizumab during cycles 3-6. Patients underwent tumor biopsies at baseline and after cycle 2 for GE profiling using DNA microarrays and TIL enumeration according to standard guidelines. Since pathologic complete remission (pCR) is relatively rare in HR+ BC, we also associated an immune gene module score (IMS) and TIL counts with the non-dichotomous variable of decrease in tumor size.

Results

Of the 150 enrolled patients, n = 113 were HR+. For n = 71, both TIL and GE data were available at baseline, while for n = 78 and n = 49 patients longitudinal TIL and GE data at baseline and cycle 2 were available, respectively. At baseline, on both univariate (OR = 2.29, P = 0.037) and multivariate analysis (OR = 2.35, P = 0.044) IMS was associated with pCR, while its association with tumor shrinkage was only apparent on univariate (P = 0.047) and not multivariate analysis (P = 0.061). TIL infiltration >50% (n = 9) was associated with neither pCR (OR = 1.812, P = 0.61) nor tumor shrinkage (P = 0.99). However, decreases in TIL counts in cycle 2 compared with baseline were associated with lesser decreases in tumor size (P = 0.043 for univariate and P = 0.044 for multivariate analysis).

Conclusions

Baseline immune function as assessed by GE analysis, but not TIL enumeration, and a preserved abundance of TILs after chemotherapy were predictive for chemosensitivity at the neoadjuvant setting in patients with HR+, HER2- BC.

Clinical trial identification

NCT00957125

Legal entity responsible for the study

Karolinska University Hospital

Funding

Swedish Cancer Society, BioCARE, Governmental Funding of Clinical Research within the National Health 1Service and unrestricted grants from Roche Sweden AB

Disclosure

J. Bergh: Personal fees: UpToDate. Institutional grants: AstraZeneca, Amgen, Bayer, Merck, Pfizer, Roche, Sanofi-Aventis. T. Foukakis: Personal fees: Novartis, Pfizer, Roche, UpToDate. Research institutional: Pfizer, Roche. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

220P - Safe resection margins in breast-conserving surgery (ID 2892)

Presentation Number
220P
Lecture Time
13:15 - 13:15
Speakers
  • Y. Kostiuchenko
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Our first research concerning resection margins in breast-conserving surgery (BCS) for breast cancer patients (BCP) was performed in 2003-2006. In the group of BCP where after positive margins we performed mastectomy (0%) and in the group where the negative margins were achieved immediately (1.27%) the incidence of local recurrences was lower comparing to the group that had re-resections (4.76%) or the group without microscopic control of margins (8.16%). We tried to understand the causes of local recurrences after BCS, and develop recommendations on achieving safe resection margins.

Methods

To clarify the possibility of cancer spreading to the side of regional lymph nodes we were injecting a solution of aqueous methylene in four points around a tumor in 30 minutes before surgery and subsequently we were studying cuts made at the distance up to 5 cm from the tumor margin. In 12% of cases we identified tumor elements in cuts located at a distance of more than 3 cm. In this study we included 996 BCP of I-III stages who had BCS at the National Cancer Institute in 2008-2015. The main group consisted of 379 BCP who had an additional removal of tissues towards the axillary area as a monoblock during the BCS (with additional histological control as mentioned above). 617 BCP in the control group had standard BCS. The average age of BCP was 50.6±1,1 in the main group, and 49,8±0,9 in the control group (p > 0.05). Median follow-up was 142 weeks.

Results

Achieving negative margins at the area of possible metastasis significantly reduces local recurrence rates, especially in a stage II (5.34%) and III (3.35%) BCP. The incidence of distant metastases (diagnosed in 93 (9.34%) cases) was significantly lower in the main group – 21 (5.54%) comparing to the control group – 72 (11.67%), both in general and in each stage (p < 0.05). Time of distant metastases occurrence was slightly lower in the control group – 84,7±8,9 weeks, comparing to the main group – 101,3±9,5 weeks (p > 0.05).

Conclusions

BCS with the additional removal of tissues towards the axillary area and lymph nodes dissection as a monoblock is reasonable and significantly reduces local and distant metastases rates. However, more long-term research in this area is urgently needed, especially in terms of benefit in survival.

Clinical trial identification

The study is approved by the Commission on issues of ethics of the National Cancer Institute (Protocol No. 7 of 08.04.2010) and the Commission on issues of ethics of the Bogomolets National Medical University (Protocol No. 71 of 10.04.2013).

Legal entity responsible for the study

Bogomolets National Medical University, National Cancer Institute

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

221P - A clinically applicable model to predict risk of relapse in patients treated for locally advanced breast cancer: Potential utilization in future clinical trials (ID 3155)

Presentation Number
221P
Lecture Time
13:15 - 13:15
Speakers
  • O. Aseyev
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Despite advances in cancer treatment, over 25% of patients (pts) with locally advanced breast cancer (LABC) relapse during first 5 years after treatment. The primary objective was to construct a prediction tool for risk of relapse (RR) in pts with LABC after neoadjuvant therapy (NAT).

Methods

This was single center, retrospective study of 546 pts with LABC who received NAT at the Ottawa Hospital Cancer Center between 2005 and 2015. Median follow-up was 49 months. The following data collected: demographics, tumor size, nodal and receptor status, grade, HER-2, stage, treatment and clinical outcomes. Primary endpoints were local and/or distant recurrence rate and time to relapse during the first 5 years. A prediction tool was devised based on the Cox regression model.

Results

Over 60 variables were included in primary analysis. Cox regression proportional hazards model analysis resulted in only 5 factors with significant influence on risk of relapse during first 5 years of follow up. Risk factors and their risk prediction value are: 1) residual disease (yes- 4; no-0), (HR = 4.25; p-value<0.001), 2) lymph nodes status (positive-3; negative-0), (HR = 2.27; p-value=0.006), 3) Inflammatory histology (yes-2; no-0), (HR = 1.90; p-value=0.003) 4) estrogen receptors status (positive-2; negative-0), (HR = 2.07; p-value=0.001), 5) Adjuvant radiotherapy (yes-0; no-1), (HR = 1.76; p-value=0.036). When these factors are combined the following Relapse Prediction (RP) Score can be constructed. According to this simple RP score, patients can be classified into to three groups (RP score – 0-5; 6-7; 8-12). RR was 7 times higher in patients with RP Score 8-12 vs patients with score 0-5 (p-value<0.001). Internal validation of proposed model was performed. ROC analysis of the proposed model revealed a sensitivity of 75%.

Conclusions

Patients with LABC represent a heterogeneous group with diverse risk of disease recurrence that can be predicted. Patients with high risk may require additional treatment and/or more active follow-up strategies and this simple model may be used to design unique studies in LABC based on RP score. We intend to further validate this model on a larger multi center/provincial population.

Legal entity responsible for the study

Olexiy Aseyev

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

222P - Role of radiotherapy and its impact on survival of male breast cancer: Experience from a tertiary cancer center (ID 4460)

Presentation Number
222P
Lecture Time
13:15 - 13:15
Speakers
  • R. Upadhyay
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Male breast Cancer (MBC) is a rare disease accounting for about 1% of all malignancies in men and 1% of all breast cancers. These patients(pts) are managed like female breast cancers. There is limited literature available defining the role of radiotherapy(RT) in management of MBC. We conducted a retrospective analysis to study the impact of adjuvant RT on outcome of MBC pts treated at our centre.

Methods

Review of MBC pts presenting to our centre from 2005 to 2015 was done. All underwent pre-treatment evaluation in a combined tumor clinic comprising radiation, surgical and medical oncologist. Most pts were treated with surgery followed by adjuvant therapy and kept on regular follow up. Overall survival (OS) was defined as time from pathologic diagnosis to last follow up or death. Disease free survival (DFS) was defined as time from diagnosis to first relapse.

Results

96 pts of MBC were identified. Median age was 58 years (range 28-83). Clinical stage I, II, III and IV were 8, 27, 39 and 22 respectively. Of 66 pts with known receptor status, 83% were ER positive, 82% PR positive, 20% Her-2/neu positive and 7.5% triple negative. 69 pts underwent modified radical mastectomy or wide local excision. 54% were pathologically node positive. Adjuvant RT was delivered to 34% pts at 1.8-2 Gy per fraction to a median dose of 50 Gy (range 45-60 Gy). Radiation field comprised of chest wall with (75%) or without regional nodes (25%). Median follow up was 24 months (range 9-132). 16 pts had relapse out of which 4 had local and 13 had distant (most common site bone) metastases after a median duration of 19 mnths. 2 yr estimated DFS for the entire cohort was 79.2% and 2 yr OS was 85.7%. The 2 yr DFS in pts undergoing surgery was 86.4% vs 29.2% in those who did not(p = 0.001). Pts who received adjuvant RT had better 2 yr DFS (92.4% vs 52.9%, p = 0.002). Adjuvant chemotherapy did not significantly affect the 2 yr DFS (93.1% vs 73%, p = 0.123).

Conclusions

MBC mostly present in advance stages at our centre and harbor HR positive disease with low HER-2 overexpression. Adjuvant RT provided a statistically significant improvement in outcome. Longer follow up of these cohort of pts is required for accurate evaluation of role of RT in MBC.

Legal entity responsible for the study

All India Institute of Medical Sciences

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

223P - Early characteristics of breast neoplasms on contrast-enhanced ultrasonography and its clinical value (ID 1574)

Presentation Number
223P
Lecture Time
13:15 - 13:15
Speakers
  • S. Qin
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

To investigate the characteristics of breast neoplasms on contrast-enhanced ultrasonography and its clinical value.

Methods

225 female patients with breast masses unable to be diagnosed by conventional ultrasonography were examined with contrast-enhanced ultrasonography (CEUS). Their ages range were 12 to 85 years, the mean age was (45.8±17.6) years. 225 cases were investigated. For cases of multiple lesions, performed which was the highest classification level of BI-RADS; for the lesions of the same level, performed the largest lesion. The characteristics of these masses on CEUS were analyzed and compared with the results of pathology examination. The contrast agent(UCA) was SonoVue (Bracco, Italy).

Results

91 cases were malignant and 134 cases were benign. The 91 malignacies displayed: irregular shapes were 80.2%(73/91), tortuous, massive or penetrating vessels were 86.8% (79/91), heterogeneous distribution of contrast enhancement were 83.5% (76/91), perfusion defect of contrast signals were 89.0% (81/91), local retention of contrast signals were 93.4% (85/91), rapidly entering and exporting from the lesions were 65.9% (60/91). Significant differences of above CEUS characteristics were found between the benign and malignant breast lesions (P < 0.05), The two most important features were perfusion defects and local retention of the contrast signals, with the sensitivity and specificity attained to 89.0% and 91.8%, and 93.4% and 92.5%, respectively. Poorly defined boundaries of the 91 malignacies were 64.8%(59/91), and the specificity was 47.8%. The malignant cases had enlarged maximum diameter on CEUS compared to pre-contrast (P < 0.05).

Conclusions

The typical features of breast cancers on CEUS were irregular shapes, tortuous, massive or penetrating vessels, heterogeneous distribution of contrast enhancement, with perfusion defect or local retention of contrast signals, rapidly entering and exporting from the lesions, enlarged maximum diameter of the lesions on CEUS compared to pre-contrast. It is valuable for CEUS in the diagnosis and differential diagnosis of breast neoplasms clinically.

Clinical trial identification

From 2011 to 2015

Legal entity responsible for the study

The Center of Interventional Ultrasound Diagnosis in Nanjing Region

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

224P - Synchronous and metachronous breast cancer in Ukraine (ID 2817)

Presentation Number
224P
Lecture Time
13:15 - 13:15
Speakers
  • I. Motuzyuk
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

This study aims to evaluate the current state of multiple primary malignant neoplasms (MPMN) in Ukraine and develop decision criteria for type of surgical treatment for these patients.

Methods

The study included 2,032 patients who received special treatment at the Department of Breast Tumours at the National Cancer Institute in 2008-2015. Among them there were 195 MPMN patients where 107 (54.9%) presented synchronous cancer (SC) and 88 (45.1%) presented metachronous cancer (MC). The average age of patients was 46.6, and the number of postmenopausal women was 63.1%. Among SC patients there were 60 (56.1%) with only breast localizations and 47 (43.9%) with combination of breast and other localizations (gynaecological etc.), and among MC there were 41 (46.6%) with only breast localizations and 47 (53.4%) with combination of breast and other localizations. All the patients were evaluated in terms of aggressiveness of the disease, survival rates, as well as risk factors and treatment options.

Results

The clinical course of the disease (CCD) in MPMN patients was worse in SC patients comparing to MC patients (p=0.00162). A more aggressive CCD was observed in patients exposed to radiation from the Chernobyl accident (p = 0.000798). There was no influence on CCD of such factors as primary localization, type of special treatment, age and type of settlement. However, the impact of type of surgery was statistically proven, i.e. CCD in patients who underwent mastectomy was worse comparing to patients who underwent breast-conserving surgery (p=0.00048). Plastic and reconstructive surgery in SC patients was statistically proven as reasonable increasing overall survival by 29% (p=0.015). There was an influence of local recurrences on the overall survival in SC patients reducing it by 71% (p=0.033), however, there was no influence in MC patients.

Conclusions

The MPMN patients have to get an improved attentive management and treatment. Medical and surgical oncologists should concern all the risk factors that have influence on CCD in these patients and provide the best option of management. The research in this area of oncology is open and this is crucial to continue researches for better outcome of these patients.

Clinical trial identification

The study is approved by the Commission on issues of ethics of the National Cancer Institute (Protocol No. 7 of 08.04.2010) and the Commission on issues of ethics of the Bogomolets National Medical University (Protocol No. 71 of 10.04.2013).

Legal entity responsible for the study

Bogomolets National Medical University, National Cancer Institute

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

225P - Clinical outcomes of single versus double hormone receptor positive breast cancer patients treated with neoadjuvant chemotherapy (ID 3939)

Presentation Number
225P
Lecture Time
13:15 - 13:15
Speakers
  • J. Raphael
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

This study aimed to evaluate and compare tumor response rates and survival outcomes between single and double hormone receptor (HR) positive (+) [Estrogen Receptor (ER+)/Progesterone Receptor (PR) negative (-) or ER-/PR+ versus ER+/PR+] breast cancer (BC) patients with any HER2 status treated with neoadjuvant chemotherapy at a single institution.

Methods

A retrospective review was conducted using the Sunnybrook “Biomatrix” database to identify eligible patients. A multivariable logistic regression analysis (MLR) was performed to assess the association between HR status (single or double HR+) and pathologic complete response (pCR) rates at surgery. A Kaplan-Meier method was used to estimate Disease Free Survival (DFS) and a log-rank test was used to compare DFS between 3 subgroups of patients: single or double HR+ and HR negative patients.

Results

Three hundred and four BC patients were identified and included in the analysis with a median follow up of 43.3 months (Q1-Q3: 28.7-61.1) and a mean age of 49.7 years (Standard deviation 10.9). Forty seven percent (47/101), 31% (11/36) and 14% (24/167) of patients with HR negative, single HR+ and double HR+ disease achieved a pCR respectively (X2 test <0.0001). In a MLR analysis, HR status and HER2 status were associated with pCR rates. Compared to HR negative patients, patients with double HR+ disease were less likely to achieve pCR (Odd ratio (OR):0.14, 95%CI 0.06-0.31, p<0.0001) while single HR+ patients did not differ (OR:0.51, 95%CI 0.19-1.4). The association between HR+ status (single versus double HR+) and pCR rates compared to HR negative patients remained the same in subgroup analyses of HER2+ and HER2 negative patients separately. No difference in survival (DFS) was seen between the 3 subgroups of patients: HR negative, single and double HR+ patients.

Conclusions

BC patients with single HR+ disease behave differently than double HR+ patients in terms of likelihood of achieving pCR after neoadjuvant chemotherapy and do not differ from HR negative patients. This difference does not translate into a difference in DFS. Prospective studies are needed to validate these findings before considering different treatment strategies for these 2 subgroups of HR+ BC patients.

Legal entity responsible for the study

Jacques Raphael

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

226P - The correlation between toll-like receptor genes polymorphisms, tumor microenvironment characteristics and the effectiveness of preoperative chemotherapy for locally advanced breast cancer (ID 940)

Presentation Number
226P
Lecture Time
13:15 - 13:15
Speakers
  • N. Verovkina
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Toll-like receptor (TLR) activation may be an important event in tumor cell immune evasion. TLR2 and TLR4 gene polymorphisms correlate with increased susceptibility to cancer development, response to conventional chemotherapy in various organs.

Methods

Treatment results of 62 patients with breast cancer stages T1-3N0-3M0 treated with neoadjuvant chemotherapy were evaluated. The level of the post-treatment CD4+, CD8+, FOP3+ tumor-infiltrating immune cells and Ki-67 positive cells were studied. The polymorphisms of TLR4 (C399T) and TLR2 (G753A) genes were investigated using a PCR restriction fragment length polymorphism method. Statistica10.0 software was used to perform analysis of variance.

Results

ER+ and PR± expressing tumors were identified in 69.3% of patients, ER and PR negative tumors - in 30.6%. Pathological complete response (pCR) was identified in 14.6%. Genotype CC of TLR4 (C399T) gene was detected in 87%, whereas genotype CT - in 9.6% and genotype TT - in 3.4% of patients. Genotype GG of TLR2 (G753A) gene was detected in 88.7%, genotype GA - in 11.3% of patients. A correlation was found between polymorphisms of TLR2 (G753A) and axillary lymph nodes involvement. In carriers of GA genotype of TLR2 gene (G753A) we found more frequent axillary LN metastases (χ2=5.75; p = 0.01). A direct correlation was identified between level of Ki-67 and the level of regulatory FOXP3 cells in carriers of GA genotype of TLR2 gene(r = 0.96; p = 0.008). There appears to be a relationship between TLR4 gene and levels of CD4 + (p=0.01) and CD8 + (p=0.02) as well as an association between TLR4 (C399T) gene and residual cancer burden (RCB) (p = 0.04). In carriers of TT genotype of TLR4 gene the level of CD4+ cells was significantly lower (p = 0.03). In carriers of CC genotype of TLR4 (C399T) gene we found a direct correlation between the level of CD8+ cells and Ki- 67 in the residual tumor (r = 0.38, p = 0.01). Higher level of CD4+ is associated with lower RCB in carriers of CC genotype of TLR4 (C399T) gene (r = 0.3, p < 0.05).

Conclusions

Preliminary results of the study indicate that further elucidation of the role of the TLRs in breast cancer development is promising.

Legal entity responsible for the study

National Cancer Institute

Funding

National cancer institute

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

227P - Triple negative breast cancer: 10-year survival update of the applied treatment strategy in Kuwait (ID 2209)

Presentation Number
227P
Lecture Time
13:15 - 13:15
Speakers
  • M. Fayaz
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Triple negative breast cancer (TNBC) is recognized as a distinct clinical and biological entity of poor outcome for almost two decades, yet its treatment strategy still needs to be better specified. The study aim is to update the 10-year survival data of our TNBC patients and to find its association with different applied treatment modalities.

Methods

We updated 10-year survival data of 359 women diagnosed with TNBC between 1999 and 2009 in Kuwait Cancer Control Center (KCCC). The overall survival (OS), disease free survival (DFS), distant metastasis free survival (DMFS) and loco-regional free survival (LRFS) were estimated using Kaplan Meier method. Survival was correlated with different prognostic factors and treatment modalities. Statistical significance was calculated using the log-rank test and defined as p < 0.05. Cox regression was used for Multivariate analysis.

Results

TNBC represented 12% of breast cancer in Kuwait with a median age of 48 years. The stage distribution was as follow: stage I, II, III, IV in 15%, 43%, 35% and 7% of patients respectively. Regarding surgery, 33% had Conservative surgery; 67% had mastectomy and 82% had axillary clearance. Chemotherapy was neoadjuvant in 25%, adjuvant in 56% and palliative in 5% of patients. Two-thirds of patients (67%) received adjuvant radiotherapy. After a median follow-up of 108 months, the 10-year OS, DFS, DMFS and LRFS were 66%, 59%, 72% and 77% respectively. The 10-year OS was 92%, 80%, 49% and 0% for Stage I,II,III and IV respectively (p = 0.0000). OS was significantly worse with the presence of lymphovascular invasion (LVI; p = 0.003). OS was not significantly affected by age, grade or treatment modality. In multivariate analysis, the clinical stage and LVI were still significant (P = 0.0000 and 0.04 respectively).

Conclusions

In absence of biological biomarkers, the clinical stage and LVI seems to be the only significant prognostic factors for survival of TNBC patients in our study population. Timing of chemotherapy as well as the extent of surgery do not seem to affect the TNBC patients' outcome.

Legal entity responsible for the study

KCCC

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

228P - A decade of HER2-targeted therapy in older patients with invasive breast cancer at Institut Curie (ID 3488)

Presentation Number
228P
Lecture Time
13:15 - 13:15
Speakers
  • R. Geiss
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Around 40% and 20% of breast cancers (BC) occur in women aged ≥ 65 and ≥ 75 years respectively. Although HER2-targeted therapy has profoundly improved the management of HER2+ BC, literature is short of data for frail and elderly patients reflecting the poor representation of older patients in registration trials.

Methods

We conducted a retrospective analysis of any stage HER2+ BC patients aged ≥ 65 years treated with a HER2-targeted therapy at Institut Curie between 2000 and 2012 to assess treatment feasibility. Baseline data were extracted from the institution database and patients’ files were reviewed for treatment compliance and safety profile.

Results

From 2000 to 2012, 261 and 76 patients received anti-HER2 treatment in adjuvant and metastatic setting respectively. In adjuvant setting (age distribution 65-69/70-74/≥75: 109/85/67; median follow-up 65 months), the median duration of trastuzumab treatment was 12 months with an 80% completion rate (defined as > 9 months of treatment) decreasing significantly ≥ 75 years (70%). Grade ≥ 3 cardiac toxicity occurred in 9.6% of patients, was reversible in 72% of cases, and multivariate analysis identified the following risk factors for cardiac events: history of thromboembolic disease, valvulopathy and performance status (PS) ≥ 2 [OR 6.3 (95% CI: 1.4-27.7), 25.6 (4-162.8) and 18.2 (1.1-304.3) respectively], but not age. In metastatic setting (age distribution 65-74/≥ 75: 41/35; median follow-up 27 months), median duration of HER2-targeted therapy was 22.8 months (0-109.2), with no impact of age. Trastuzumab and lapatinib (alone or in combination) were mostly prescribed, pertuzumab and T-DM1 representing < 15% of cases depending on treatment line. Multivariate analysis identified the following factors for mortality: PS ≥ 2, history of thromboembolic disease [OR 3 (95% CI: 1.1-8.1) and 3.3 (0.7-16.3) respectively], but not age.

Conclusions

HER2-targeted therapy seems feasible in ≥ 65 yrs patients in both adjuvant and metastatic setting. Cardiac toxicity occurs in 10-15% but is reversible in most cases. Chronological age does not seem to affect duration of anti-HER2 treatment nor cardiac toxicity.

Legal entity responsible for the study

Geiss Romain

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

229P - Dedicated breast PET to predict pathological complete response after neoadjuvant chemotherapy for breast cancer (ID 4312)

Presentation Number
229P
Lecture Time
13:15 - 13:15
Speakers
  • M. Fujiwara
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Reports indicate that whole-body (WB) 18F-fluorodeoxyglucose (FDG) PET can predict a pathological complete response (pCR) after neoadjuvant chemotherapy (NAC). New dedicated breast PET (DBPET) can generate high-resolution images and thus might be able to predict pCR after NAC. The present study aimed to determine whether or not DbPET can predict the effects of NAC for breast cancer more effectively than WBPET.

Methods

The clinical responses of 35 consecutive patients with breast cancer (T1–4, N0–1, M0) who underwent NAC between January 2016 and January 2017 were assessed using WBPET and DBPET. We assessed maximum standardized uptake values (SUVmax), before (pre-SUVmax) and after (post-SUVmax) NAC and rates of change in SUVmax (ΔSUVmax) before and after NAC. Relationships between these parameters and pathological responses (pCR) were assessed using each modality. We created receiver operating characteristics curves (ROC), calculated areas under them (AUC) for both WBPET and DBPET images and predicted pCR.

Results

Twelve of 35 patients achieved pCR. The median pre-SUVmax, post-SUVmax and ΔSUVmax among the 35 patients determined using WBPET and DBPET were 6.8 and 18.0, 1.7 and 3.1, and 78.2 and 77.6, respectively. Uptake of 18F-FDG was indistinguishable from background in WBPET, but confirmed in DBPET after NAC in three of 23 patients with non-pCR disease. The median pre-SUVmax of WBPET and DBPET in the pCR group was higher than non-pCR group (7.88 and 21.73 vs 6.22 and 16.28, p = 0.30 and p = 0.15, respectively). In contrast, post-SUVmax of the pCR group was lower than non-pCR group (0.97 and 2.06 vs 1.54 and 3.86, p = 0.062 and p = 0.032, respectively). ΔSUVmax of pCR group was higher than non-pCR group (82.23 and 88.56 vs 76.34 and 72.55, p = 0.27 and p = 0.04, respectively). Additionally, the AUC of DBPET (pre-SUVmax: 0.543, post-SUVmax: 0.725, ΔSUVmax: 0.752) was higher than WBPET (pre-SUVmax: 0.477, post-SUVmax: 0.694, ΔSUVmax: 0.549) in either time points.

Conclusions

The diagnostic accuracy of DBPET were equal to, or better than those of WBPET. DBPET might serve as a new diagnostic modality when planning therapeutic strategies for patients with breast cancer after neoadjuvant chemotherapy.

Legal entity responsible for the study

Hiroshima University

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

230P - Efficacy and safety profile of the adriamycin/cyclophosphamide (AC) followed by docetaxel/cisplatin (DC) in locally advanced breast cancer (ID 4618)

Presentation Number
230P
Lecture Time
13:15 - 13:15
Speakers
  • T. Abdelhamid
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Neoadjuvant chemotherapy (NCT) using anthracycline - taxane based chemotherapy achieved better outcome and increase the rate of breast conservative surgery (BCS). This study evaluates the efficacy and safety profile of 4 cycles AC followed by 4 cycles of DC as NCT and detect pCR with its subsequent effect on BCS, OS, DFS and its correlation with molecular subtypes and other markers as ERCC1and Ki-67.

Methods

A prospective phase II study at the Medical Oncology Department of the National Cancer Institute (NCI), Cairo University, where 104 female patients with LABC were recruited during the period from March 2010 to July 2013 to receive eight cycles of NCT, 4 cycles of (AC) followed by 4 cycles of (DC), responding patients were referred to surgery then to radiotherapy for local control. All patients were assessable for toxicity. ER, PR, Her2neu, Ki-67 and ERCC1 were done.

Results

Median age: 51yrs, postmenopausal: 48.1%, median tumour size at presentation: 8 cm, stage IIIB: 82.7%, ER/PR positive: 78.8% and 25% were Her2neu positive. The pCR after DC according to Miller and Payne system was 20.2%, with mean tumour size:4.15± S.D 2.703. Age and menopausal status showed statistical significant correlation with pathological response. The correlation between DFS in positive and negative ERCC1 was statistically significant; (p-value ≤ 0.01). The overall survival (OS)of the patients was 85.4% at 36 months while the disease free survival (DFS) was 85.3% at 24 months. Anaemia G2 & more was encountered in 27 patients (26%); neutropenia G2 & more was reported in 49 patients (47.1%), while Peripheral neuritis was observed in 103 patients (99%).

Conclusions

Adding Cisplatin to Docetaxel for 4 cycles after 4 cycles of Adriamycin – Cyclophosphamide improved the pCR, OS and rate of BCS. ERCC1 is a predictor marker of DFS.

Legal entity responsible for the study

IRB Committee of the national cancer institute of Cairo University

Funding

National Cancer Institute - Cairo University

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

232P - Pregnancy associated breast cancer spotlights (ID 5405)

Presentation Number
232P
Lecture Time
13:15 - 13:15
Speakers
  • M. Shalaby
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Pregnancy associated breast cancer (PABC) is a rare entity of breast cancer. It is defined as breast cancer occurring during pregnancy or within 12 months from end of pregnancy. Though rare, it represents a very challenging situation to both the physician and patient. This retrospective study is an attempt to focus on PABC cases presenting at our institute regarding their personal, disease characteristics and type of treatment received.

Methods

This is a retrospective study conducted at Kasr-Al-Ainy oncology center “NEMROCK”. The files of female patients diagnosed with breast cancer under the age of 40 in the period from January 2005 to December 2014 were retrospectively reviewed. A comparison has been conducted between PABC cases and non-pregnant cases concerning personal and disease characteristics, modality of treatment and outcomes.

Results

This study included a total of 175 patients 40 years old or younger, among them 40 were PABC cases. Twenty five percent of PABC presented with distant metastasis at first presentation while only 11.6% of the non-pregnant patients presented with metastasis (P value 0.039). Time from onset of symptoms till breast cancer diagnosis was more than 6 months in 55.6% of PABC cases in comparison to 36.3% in non-pregnant cases (P value 0.043). Concerning treatment; 32.4% of PABC cases received neoadjuvant chemotherapy while 20.7% of non-pregnant patients received it (P value 0.027). There was no statistical difference in personal and disease characteristics between pregnant and non-pregnant patients including family history, pathological subtypes, stage, grade and biological subtypes. There was no statistically significant difference in disease free survival between PABC cases and non-pregnant ones (P value 0.497).

Conclusions

PABC is associated with a late diagnosis. Although PABC patients present at later stages than non-pregnant ones and the use of neoadjuvant treatment is higher in pregnant cases, the outcome of patients with PABC is comparable to that of non-PABC of matched age.

Legal entity responsible for the study

Kasr alainy department of oncology

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

233TiP - KEYNOTE-522: Phase III study of pembrolizumab (pembro) + chemotherapy (chemo) vs placebo + chemo as neoadjuvant followed by pembro vs placebo as adjuvant therapy for triple-negative breast cancer (TNBC) (ID 1878)

Presentation Number
233TiP
Lecture Time
13:15 - 13:15
Speakers
  • P. Schmid
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Endogenous anticancer immunity may be enhanced with immune checkpoint inhibition by increased tumor-specific antigen release after chemo. Combining the anti–PD-1 inhibitor pembro with chemo may be an effective treatment strategy for TNBC in neoadjuvant and adjuvant settings. KEYNOTE-522 (NCT03036488) is a phase III study of neoadjuvant pembro + chemo followed by adjuvant pembro vs neoadjuvant placebo + chemo followed by adjuvant placebo in pts with TNBC.

Trial design

Eligible pts are aged ≥18 y with previously untreated, centrally confirmed, nonmetastatic TNBC, defined as combined primary tumor (T) and regional lymph node (N) staging per AJCC (investigator-assessed: T1c N1-2, T2-4 N0-2). Pts with bilateral or multifocal primary tumors or inflammatory breast cancer are allowed. ECOG PS 0-1 and adequate organ function are required. Pts with a history of invasive malignancy ≤5 y or prior therapy before study start are excluded. Approximately 855 pts will be randomly assigned to 1 of 2 arms and stratified by tumor nodal status (positive vs negative), size (T1/T2 vs T3/T4), and carboplatin choice (Q3W vs QW). In arm 1, pts will receive 4 cycles of pembro 200 mg Q3W + paclitaxel (80 mg/m2 QW on d 1, 8, 15) + carboplatin (AUC 5 Q3W on d 1 or AUC 1.5 QW on d 1, 8, 15) and then 4 cycles of pembro + doxorubicin (60 mg/m2 on d 1) or epirubicin (90 mg/m2 on d 1) + cyclophosphamide (600 mg/m2 Q3W on d 1) as neoadjuvant therapy. Definitive surgery will be 3-6 wk after the last cycle. In arm 2, placebo will replace pembro. Post-surgery, pts will receive 9 cycles of pembro or placebo as adjuvant therapy. One neoadjuvant/adjuvant cycle = 21 d; treatment is up to 17 cycles (pembro/placebo only) or until disease progression/unacceptable toxicity. Dual primary endpoints are pCR rate (ypT0/Tis ypN0) and EFS. Secondary endpoints include pCR rate (ypT0 ypN0) in all pts and in those with PD-L1+ tumors (ie, PD-L1 staining in ≥ 1% tumor cells or stroma), pCR rate (ypT0/Tis ypN0) in pts with PD-L1+ tumors, EFS in pts with PD-L1+ tumors, pCR rate (ypT0/Tis) in all pts and in those with PD-L1+ tumors, OS, and safety. Interim analyses are planned.

Clinical trial identification

EUDRACT number: 2016-004740-11 ClinicalTrials.gov, NCT03036488, January 27, 2017

Legal entity responsible for the study

Merck & Co., Inc., Kenilworth, NJ, USA

Funding

Merck & Co., Inc., Kenilworth, NJ, USA

Disclosure

P. Schmid: Honoraria: Pfizer, Boehringer, Bayer, Puma, Eisai, Celgene. Other: spouse: Genetech/Roche. J. Cortes Castan: Advisory board member: Roche, Celgene, Aztrazeneca, Cellestia Biotech, Biothera. Honoraria: Roche, Novartis, Eisai, Celgene, Pfizer. J. Bergh: Research funding; grants: Amgen, AstraZeneca, Bayer, Merck, Pfizer, Roche and Sanofi-Aventis to Karolinska Institutet/University Hospital. No personal payments. Honoraria: from UpToDate to Asklepios Medicine HB on a chapter on breast cancer diagnostics. L. Pusztai: Advisory board: Merck, Novartis, AstraZeneca. Research funding: Merck, Novartis, Roche, AstraZeneca, Seattle Genetics. Honoraria: Merck. C. Denkert: Stock ownership: Sividon Diagnostics, Cologne. Advisory board: MSD. Honoraria: Amgen, Celgene, Teva, AstraZeneca, Myriad. S. Verma: Advisory board: Amgen, Eli Lilly, AstraZeneca, Novartis, Pfizer, Roche. H.L. McArthur: Advisory board: Celgene, Merck, OBI, Spectrum Pharmaceuticals, Syndax Pharmaceuticals, Roche, Peregrine, Calithera. J. Zhao: Employment: Merck Research Lab. G. Aktan: Employment, stocks, travel: Merck & Co., Inc. T. Dang: Employment and stock: Merck Sharp & Dohme. R. Dent: Advisory board: AstraZeneca, Pfizer, Roche, Merck. Research funding: AstraZeneca. Honoaria: Eisai, Roche, Pfizer. Travel expenses: Roche, Merck, Pfizer.

Collapse
Poster display session Poster Display session

234TiP - KEYNOTE-355: Randomized, double-blind, phase III study of pembrolizumab (pembro) + chemotherapy (chemo) vs placebo (PBO) + chemo for previously untreated, locally recurrent, inoperable or metastatic triple-negative breast cancer (mTNBC) (ID 2303)

Presentation Number
234TiP
Lecture Time
13:15 - 13:15
Speakers
  • J. Cortes Castan
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In the phase Ib KEYNOTE-012 study, the anti–PD-1 antibody pembro demonstrated promising antitumor activity and acceptable safety as monotherapy in pretreated patients (pts) with PD-L1+ mTNBC. Adding pembro to chemo may enhance antitumor activity. KEYNOTE-355 (NCT02819518) is a global phase III study of pembro+chemo vs PBO+chemo in pts with previously untreated, locally recurrent, inoperable TNBC/mTNBC.

Background

In the phase Ib KEYNOTE-012 study, the anti–PD-1 antibody pembro demonstrated promising antitumor activity and acceptable safety as monotherapy in pretreated patients (pts) with PD-L1+ mTNBC. Adding pembro to chemo may enhance antitumor activity. KEYNOTE-355 (NCT02819518) is a global phase III study of pembro+chemo vs PBO+chemo in pts with previously untreated, locally recurrent, inoperable TNBC/mTNBC.

Trial design

Eligible pts are ≥18 y and have centrally confirmed, locally recurrent, inoperable TNBC or mTNBC not previously treated with chemo (prior chemo in [neo]adjuvant setting allowed); measurable disease per RECIST v1.1; ECOG PS 0-1; ≥6 mo between definitive surgery or last dose of adjuvant chemo, whichever was last; and first disease recurrence (≥12 mo if prior treatment with same-class agent). Part 1 is an open-label, unblinded safety run-in of ∼30 pts distributed over 3 arms (pembro+nab-paclitaxel, pembro+paclitaxel, pembro+gemcitabine/carboplatin). Part 2 is a double-blind, PBO-controlled study of ∼828 pts to be randomized 2:1 to pembro 200 mg every 3 weeks + chemo (nab-paclitaxel 100 mg/m2 on d 1, 8, and 15 every 28 d; paclitaxel 90 mg/m2 on d 1, 8, and 15 every 28 d; or gemcitabine 1000 mg/m2 + carboplatin AUC 2 on d 1 and 8 every 21 d) or PBO+chemo. Crossover not allowed. Stratification factors are study chemo (taxane vs gemcitabine/carboplatin), tumor PD-L1 expression (+/-), and prior therapy with same-class agent in the (neo)adjuvant setting (yes/no). Treatment will occur for ≤35 administrations (pembro/PBO only) or until confirmed disease progression, unacceptable toxicity, withdrawal of consent, or decision to discontinue. Primary end points are safety in part 1 and PFS (by RECIST v1.1, central radiology review) and OS in part 2; secondary end points include ORR (by RECIST v1.1, central radiology review), duration of response. AEs will be graded per NCI CTCAE v4.0. Response will be assessed at wks 8, 16, 24, then at 9-wk intervals up to 1 y, and at 12-wk intervals thereafter. Interim safety analysis will occur after pts complete 1 treatment cycle in part 1.

Methods

Eligible pts are ≥18 y and have centrally confirmed locally recurrent inoperable TNBC or mTNBC not previously treated with chemo (prior chemo in [neo]adjuvant setting allowed), measurable disease per RECIST v1.1; ECOG PS 0-1; and ≥6 mo between definitive surgery or last dose of adjuvant chemo, whichever was last, and first disease recurrence (≥12 mo if prior treatment with same-class agent). Part 1 is an open-label, unblinded safety run-in of ~30 pts distributed over 3 arms (pembro+nab-paclitaxel, pembro+paclitaxel, pembro+gemcitabine/carboplatin). Part 2 is a double-blind, PBO-controlled study of ~828 pts to be randomized 2:1 to pembro 200 mg every 3 weeks + chemo (nab-paclitaxel 100 mg/m2 on d 1, 8, and 15 every 28 d; paclitaxel 90 mg/m2 on d 1, 8, and 15 every 28 d; or gemcitabine 1000 mg/m2 + carboplatin AUC 2 on d 1 and 8 every 21 d) or PBO+chemo. Crossover not allowed. Stratification factors are study chemo (taxane vs gemcitabine/carboplatin), tumor PD-L1 expression (+/-), and prior therapy with same-class agent in the (neo)adjuvant setting (yes/no). Treatment will occur for ≤35 administrations (pembro/PBO only) or until confirmed disease progression, unacceptable toxicity, withdrawal of consent, or decision to discontinue. Primary end points are safety in part 1 and PFS (by RECIST v1.1, central radiology review) and OS in part 2; secondary end points include ORR (by RECIST v1.1, central radiology review), duration of response. AEs will be graded per NCI CTCAE v4.0. Response will be assessed at wks 8, 16, 24, then at 9-wk intervals up to 1 y, and at 12-wk intervals thereafter. Interim safety analysis will occur after pts complete 1 treatment cycle in part 1.

Clinical trial identification

EUDRACT number: 2016-001432-35 ClinicalTrials.gov, NCT02819518

Legal entity responsible for the study

Merck & Co., Inc., Kenilworth, NJ, USA

Funding

Merck & Co., Inc., Kenilworth, NJ, USA

Disclosure

J. Cortes Castan: Advisory board member for Roche, Celgene, Aztrazeneca, Cellestia Biotech, Biothera. Honoraria from Roche, Novartis, Eisai, Celgene, Pfizer. Z. Guo, V. Karantza, G. Aktan: Employment and stock ownership: Merck.

Collapse
Poster display session Poster Display session

235TiP - A Phase Ib trial of xentuzumab and abemaciclib in patients with locally advanced or metastatic solid tumours, hormone receptor-positive (HR+), HER2-negative (HER2-) breast cancer (BC; +/-endocrine therapy), or non-small-cell lung cancer (NSCLC) (ID 4672)

Presentation Number
235TiP
Lecture Time
13:15 - 13:15
Speakers
  • A. Prat
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The insulin-like growth factor (IGF) and the cyclin D-cyclin-dependent kinase (CDK) 4/6-retinoblastoma pathways have been implicated in the pathogenesis and resistance mechanisms of a variety of cancers, including HR+ HER2- BC and NSCLC. IGF-ligand dependent signalling via the IGF receptor results in upregulation of cyclin D1, and thus, progression through the cell cycle, providing rationale for the simultaneous inhibition of IGF and CDK4/6. This trial assesses the maximum-tolerated dose (MTD), safety and preliminary efficacy of the IGF ligand-neutralising antibody, xentuzumab, in combination with abemaciclib, a selective inhibitor of both CDK4 and 6, +/- hormone therapy, in pts with solid tumours.

Trial design

Study BI 1280.18 (NCT03099174) is a Phase Ib multicentre, non-randomised, open-label, dose escalation trial with four dose-finding cohorts (Cohorts A–D) followed by two expansion cohorts (Cohorts E, F). Eligible pts include adults ≥18 yrs (≥20 for Japan), with measurable or evaluable disease, adequate organ function, ECOG PS ≤ 1, and unresectable advanced or metastatic solid tumours after failure on standard therapy (Cohort A), postmenopausal locally advanced or metastatic HR+, HER2- BC (Cohorts B–D, F), or stage IV NSCLC after 1–2 lines of therapy and failure after platinum-based chemotherapy (Cohort E); CDK4/6 inhibitor-naïve pts (Cohorts A–E) and pts who have received prior CDK4/6 inhibitors (palbociclib or ribociclib) plus aromatase inhibitors (Cohort F) are included. Pts will receive either xentuzumab plus abemaciclib alone (Cohorts A, E) or in combination (at MTD defined for the doublet therapy) with letrozole (Cohort B), anastrozole (Cohort C), or fulvestrant (Cohorts D, F). Primary endpoints are the MTD and/or recommended phase-2-dose (RP2D; Cohorts A–D), and objective response (Cohorts E, F). Further endpoints include antitumour activity (Cohorts E, F), and pharmacokinetic outcomes (all Cohorts). This study will be conducted in the US, Europe and Japan. Pt screening is planned to start May 17; target enrolment: N≈88.

Clinical trial identification

NCT03099174; 1280.18

Legal entity responsible for the study

Boehringer Ingelheim

Funding

Boehringer Ingelheim

Disclosure

P. Lo Russo: Advisory boards: Agios: Data Safety Monitoring Board (2016-2017), Alexion: Advisory Board Member (2016-2017), Ariad: Advisory Board Member (2016-2017), GenMab: Advisory Board Member (2016-2017), Glenmark: Advisory Board Member (2016-2017), Halozyme: Data Safety Monitoring Board (2016-2017), Menarini: Advisory Board Member (2016-2017), Novartis: Advisory Board Member (2016-2017), Roche-Genentech: imCORE Alliance (2016-2019), Genentech: Advisory Board (2016-2017), CytomX: Advisory Board Member (2016-2017), Omniox: Advisory Board Member (2016-2017), Ignyta: Advisory Board Member (2016-2017). M.P. Sablin: Advisory board: Boehringer Ingelheim. E.L. Johnston: Employment: Eli Lilly and Company. Stock ownership or options: Eli Lilly and Company. T. Bogenrieder, J. Serra, K. Stucke-Straub: Employee of BI. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

BREAST CANCER, METASTATIC (ID 5675)

Lecture Time
13:15 - 13:15
Speakers
  • F. André
  • L. Yates
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15
Poster display session Poster Display session

247P - A single-arm, phase ii study assessing the efficacy of pembrolizumab (pembro) plus radiotherapy (RT) in metastatic triple negative breast cancer (mTNBC) (ID 1633)

Presentation Number
247P
Lecture Time
13:15 - 13:15
Speakers
  • H. McArthur
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Overall response rates of 13-19% have been reported with checkpoint inhibitor monotherapy in chemotherapy-resistant, PD-L1-positive mTNBC. RT is frequently used to enhance local control in mTNBC and has been reported to induce distant (abscopal) tumor responses when combined with immunotherapy. In this study we evaluate the safety and efficacy of RT combined with the anti-PD-1 inhibitor, pembro, in a single-arm, two-stage, phase II study in mTNBC (NCT02730130).

Methods

Eligible women had biopsy-proven mTNBC, ECOG performance status 0-2, and ≥2 measurable sites of metastatic disease with at least 1 site requiring RT. A total RT dose of 3000 cGy was delivered in 5 daily fractions. IV pembro was given at 200mg within 3 days of first RT fraction then every 3 weeks until disease progression. The primary endpoint was overall response rate at week 13 in the non-irradiated lesions by RECIST v1.1. Secondary endpoints included safety and overall survival. Tumor biopsies were obtained at baseline and at week 7. PD-L1 expression was not required for study entry.

Results

As of May 1, 2017, the study has completed enrollment (N = 17) with 4 women on treatment pending 13-week evaluation. Median age was 52y (range 37-73y). Median number of prior therapies received for metastatic disease was 3 (range 0 to 8). Of the 7 women not evaluable at 13 weeks: 5 died secondary to disease-related complications (at weeks 2, 6, 7, 8, and 9) and 2 came off study due to disease progression prior to week 13. Of the 6 women evaluable at week 13, 2 (33%) had a partial response (PR), 1 (17%) had stable disease (SD) and 3 (50%) had disease progression. The 2 PRs represented 76% and 75% decreases in tumor burden by RECIST v1.1 durable for 21 and 31 weeks, respectively. SD response was durable for 30 weeks. Common toxicities were mild and included fatigue, myalgia and nausea.

Conclusions

The combination of pembro and RT was well-tolerated. This is a poor prognosis population with 5/13 (38%) evaluable patients dying within 12 weeks of study entry. However, durable responses were observed outside of the RT field in 2/6 (33%) patients who were unselected for PD-L1 expression and evaluable at 13 weeks. Safety and toxicity data for all study patients will be presented.

Clinical trial identification

NCT02730130

Legal entity responsible for the study

Memorial Sloan Kettering Cancer Center

Funding

Merck

Disclosure

H.L. McArthur: Advisory boards for Celgene, Merck, OBI Pharma, Spectrum, Syndax, Roche, Peregrine, Calithera, Eli Lilly, and TapImmune. Research supported by Bristol-Myers Squibb; Eli Lilly; MedImmune, LLC/AstraZeneca; and Merck. C.A. Barker: In the past year has received research funding from Elekta, Merck, and Amgen; honoraria from Driver Group, a biotechnology company; and served as a Pfizer advisory board consultant. A. Gucalp: Research funding from Pfizer and Innocrin related to other work in triple negative breast cancer. A. Ho: Research funding by Merck. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

248P - Impact of prior treatment on palbociclib plus letrozole (P+L) efficacy and safety in patients (pts) with estrogen receptor-positive/human epidermal growth factor receptor 2-negative (ER+/HER2–) first-line advanced breast cancer (ABC): A PALOMA-2 subgroup analysis (ID 3998)

Presentation Number
248P
Lecture Time
13:15 - 13:15
Speakers
  • R. Finn
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

PALOMA-2 confirmed the efficacy and safety of P+L vs placebo (PBO)+L in pts with treatment-naive ER+/HER2– ABC (Finn NEJM, 2016). Here, we report efficacy and safety among P+L-treated pts in 2 subgroups of either exposed or not exposed to prior neo(adjuvant) endocrine therapy (ET) and/or chemotherapy (CT).

Methods

Postmenopausal women (N = 666) were randomized 2:1 to receive P (125 mg QD; 3 wk on, 1 wk off) + L (2.5 mg QD; continuously) or PBO+L. Prior neo(adjuvant) ET/CT with curative intent was allowed; prior ET or CT for recurrent advanced or metastatic disease was not. The primary endpoint was investigator-assessed progression-free survival (PFS); other key endpoints included response rates and safety. Exposure to P among subgroups is also presented.

Results

At baseline, 249/444 pts (56%) in the P+L arm had received prior ET and 213 (48%) had received prior CT (Table). Pts previously treated were slightly younger vs those not previously treated. Exposure to P+L was similar across subgroups. In P+L-treated pts, median PFS was 22.2/25.7 mo with prior ET/no prior ET and 22.4/25.7 mo for prior CT/no prior CT. The significant clinical benefit of P+L vs PBO+L remained regardless of prior ET/CT exposure. Across all subgroups, the incidence of all-causality adverse events (AEs) was similar; neutropenia was the most frequent AE. AEs leading to permanent discontinuation occurred in < 10% of pts.

248P

Prior ET n = 249No Prior ET n = 195Prior CT n = 213No Prior CT n = 231
Pt characteristics
Median age, y60645865
White/Asian race, %75/1781/1177/1678/14
Non-Hispanic or Latino, %87878687
Median duration since diagnosis, y8.80.28.80.2
Exposure to P
Median number of cycles (range)18 (1-37)21 (1-37)19 (1-37)21 (1-37)
Median treatment duration, mo (range)18.66 (0.03-33.84)20.76 (0.07-34.07)18.89 (0.03-34.07)20.37 (0.07-33.18)
Median average daily P dose, mg (range)125 (77-125)125 (78-125)125 (77-125)125 (78-125)
Efficacy endpoints for P+L vs PBO+L
Median PFS, mo Hazard ratio (95% CI)22.2 vs 11.3 0.53 (0.40-0.70)25.7 vs 19.6 0.63 (0.44-0.90)22.4 vs 13.7 0.53 (0.40-0.72)25.7 vs 17.0 0.61 (0.44-0.84)
Objective response rate, % Odds ratio (95% CI)33.7 vs 27.0 1.38 (0.84-2.29)52.8 vs 44.8 1.38 (0.82-2.33)36.2 vs 30.3 1.30 (0.78-2.22)47.6 vs 38.9 1.43 (0.88-2.32)
Rate of clinical benefit, % Odds ratio (95% CI)81.5 vs 66.7 2.21 (1.31-3.70)89.2 vs 75.0 2.76 (1.37-5.56)81.7 vs 70.6 1.85 (1.04-3.29)87.9 vs 69.9 3.12 (1.71-5.71)

NE=not estimable.

Conclusions

Regardless of prior (neo)adjuvant treatment, P+L significantly prolonged PFS vs PBO+L and demonstrated consistent efficacy in pre-treated and non–pre-treated subgroups of postmenopausal pts with ER+/HER2– ABC. Tolerability was similar across subgroups and consistent with the known safety profile of P.

Clinical trial identification

NCT01740427

Legal entity responsible for the study

Pfizer

Funding

Pfizer

Disclosure

R.S. Finn: Honoraria: Bayer, Pfizer, Bristol-Myers Squibb, Novartis, Eisai; Consulting or advisory role: Pfizer, Bayer, Novartis, Bristol-Myers Squibb, Merck; Research funding Pfizer. K.A. Gelmon: Consulting or advisory role: Pfizer, Novartis, AstraZeneca, NanoString Technologies, Merck. J. Ettl: Honoraria: Pfizer, Novartis, Roche; Consulting or advisory role: Pfizer, Novartis Pharma KK; Speakers’ bureau: Pfizer, Novartis, Roche, Celgene. J. Asselah: Consulting or advisory role: Pfizer, Novartis. A. Castrellon: Research funding: Novartis; Consulting or advisory role: Biotheranostics. A. Ruiz Simón: Honoraria: Pfizer, Novartis, Roche. A.A. Joy: Consulting or advisory role: Pfizer, Novartis, Roche, Eli Lilly, AstraZeneca, Bristol-Myers Squibb, Boehringer Ingleheim. D. Lu, E.R. Gauthier, A. Mori: Pfizer employee and shareholder. H.S. Rugo: Speakers’ bureau and honoraria: Genomic health; Research funding (Plexxikon Macrogenics, OBI Pharma, Eisai, Pfizer, Novartis, Eli Lilly, GlaxoSmithKline, Genentech, Celsion, Merck, Clovis Oncology. V. Diéras: Consulting and advisory role: Genentech, Lilly, Pfizer, AbbVie, Novartis Pharma KK, Roche-Peru; Speakers’ bureau: Pfizer, Novartis Pharma KK, Roche-Peru.

Collapse
Poster display session Poster Display session

249P - Analysis of overall survival by tumor response in MONARCH 1, a phase 2 study of abemaciclib, a CDK4 and CDK6 inhibitor, in women with HR+/HER2- metastatic breast cancer (MBC) after chemotherapy for advanced disease (ID 2069)

Presentation Number
249P
Lecture Time
13:15 - 13:15
Speakers
  • J. Cortés
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Abemaciclib is a selective inhibitor of CDK4 and CDK6 dosed orally on a continuous schedule. Abemaciclib monotherapy (MONARCH 1) has demonstrated antitumor activity in heavily pretreated patients with HR+/HER2- MBC in terms of objective response rate (ORR= CR + PR) (19.7%; 95% CI: 13.3, 27.5), disease control rate (DCR= CR + PR + SD) (67.4%), and clinical benefit rate (CBR= CR + PR + SD ≥ 6 months [m]) (42.4%). At 18 m minimum follow-up, the median progression-free survival (PFS) was 5.95 m (95% CI: 4.21, 7.50) and median overall survival (OS) was 22.3 m (95% CI: 17.7, NR). Treatment was well tolerated. The results from exploring associations between ORR and OS are reported.

Methods

The primary objective of MONARCH 1 (NCT02102490) was to evaluate ORR per RECIST 1.1. Secondary objectives included duration of response (DoR), PFS, OS, DCR, CBR and safety. To explore whether responders have a better survival than nonresponders, a Cox proportional hazards model with responder status as a time-dependent covariate was used. A multivariable analysis using Cox regression model including baseline factors (ECOG PS, age, PgR status, liver mets, pleural/peritoneal mets, number of chemotherapies, prior capecitabine, number of metastatic sites), and response status was conducted to adjust for any differences in baseline prognostic factors in the two groups. As a supportive analysis, a landmark analysis at 6 m was performed.

Results

26/132 patients (19.7%) achieved a best response of PR (responders). In the analysis where response was included as a time dependent covariate, patients who responded had a better OS (HR 0.31; 95% CI: 0.12, 0.77; p=.01) compared with nonresponders. After adjusting for potential baseline prognostic factors, results were consistent (HR 0.31; 95% CI: 0.12, 0.79; p=.01). Landmark analysis at 6 m supported results from previous analyses (HR 0.34; 95% CI: 0.12, 0.95; p=.04).

Conclusions

An association between ORR and OS was observed. Responders have a better OS compared to nonresponders.

Clinical trial identification

NCT02102490

Legal entity responsible for the study

Eli Lilly and Company

Funding

Eli Lilly and Company

Disclosure

J. Cortés: Honoraria: Roche, Novartis, Eisai, Celgene, Pfizer. Consulting/advisor: Roche, Celgene, AztraZeneca, Cellestia Biotech, Biothera. H.S. Rugo: Grants/Research Support: GSK, Genentech/Roche, Novartis, Pfizer, Merck, Eisai, Plexxikon, Macrogenics, Lilly, OBI (all funding to UC Regents only). Speaker\'s Bureau: Genomic health. S.M. Tolaney: Grants/Research Support: Genentech, Pfizer, AstraZeneca, Exelixis, Eli Lilly, Novartis, Merck. V. Dieras: Honoraria: -Consultant: Roche-Genentech, Novartis, Lilly, Pfizer; -Speaker Roche, Novartis, GSK, EISAI, Pfizer; Travel expenses: Roche, Novartis, Pfizer, GSK, EISAI, AstraZeneca. D. Patt: Employee of Texas Oncology, and McKesson Specialty Health. H. Wildiers: Advisor for Eli Lilly and Company. S. Nanda, A. Koustenis: Employee of Eli Lilly and Company, and may hold company stocks. M.N. Dickler: Advisor: Eli Lilly. Consultant: TapImmune, Novartis, Genentech/Roche, AstraZeneca, Pfizer, Syndax, Puma biotechnology, and G1 Therapeutics. Research support: Lilly, Genentech/Roche, and Norvatis. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

250P - A global phase III clinical study comparing NK105 and paclitaxel in metastatic or recurrent breast cancer patients (ID 981)

Presentation Number
250P
Lecture Time
13:15 - 13:15
Speakers
  • T. Saeki
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Paclitaxel (PTX) is a standard chemotherapy drug for metastatic or recurrent breast cancer (m/r BC). However, it presents problems such as hypersensitivity and peripheral sensory neuropathy (PSN). NK105 is a novel nanoparticle drug delivery formulation that encapsulates PTX in polymeric micelles. In a murine model, passive targeting was shown and NK105 accumulated in tumors. We expected NK105 to have similar efficacy and a better safety profile, regarding hypersensitivity and PSN, compared with PTX, considering a past phase II study in gastric cancer patients (pts). This study aimed to verify the non-inferiority of NK105 to PTX in terms of progression-free survival (PFS) in m/r BC pts.

Methods

Eligible pts were randomly assigned at a 1:1 ratio to either the NK105 (N) or PTX (P) arm. NK105 (65 mg/m2) and PTX (80 mg/m2) were administered via intravenous infusion weekly for 3 weeks followed by a 1-week rest period until disease progression. Tumor responses were assessed every 6 weeks by RECIST Ver. 1.1. The primary endpoint was PFS, while the secondary endpoints were overall response rate (ORR), overall survival (OS), and safety. PSN was evaluated by CTCAE Ver. 4.03 and FACT/GOG-NTX Ver. 4 (FACT).

Results

From September 2012 to July 2014, 436 pts were randomized and 422 pts were included in the efficacy analysis. The median PFS (95% CI) for the N and P arms was 256 (212–302) and 260 days (211–350), respectively. The adjusted hazard ratio (95% CI) was 1.255 (0.989–1.592), exceeding the set non-inferiority margin. The ORR and median OS for the N and P arms were 31.6% vs. 39.0%, and 950 vs. 1103 days, respectively. Adverse drug reactions occurred in 96.7% pts in the N arm and 98.1% in the P arm. PSN incidences in the N and P arms were 52.8% and 70.0%, respectively and incidence of Grade 3 or more was lower in the N arm than in the P arm pts. Cumulative PSN incidences between the N and P arms were significantly different (P < 0.01) and were favored in the N arm.

Conclusions

The efficacy of 65 mg/m2 of NK105 could not be verified in terms of non-inferiority of PFS relative to PTX in this study. NK105 safety profile was generally similar to that of PTX, but the NK105 PSN profile was better than that of PTX. NK105 efficacy should be re-evaluated in future studies.

Legal entity responsible for the study

Nippon Kayaku Co., Ltd.

Funding

Nippon Kayaku Co., Ltd.

Disclosure

Y. Tokuda: Obtained financial support for research activity from Nippon Kayaku Co., Ltd. in 2014, 2015, and 2016. Y. Nambu: Managing Director and Head of Pharmaceuticals Group of Nippon Kayaku Co., Ltd. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

251P - Efficacy and safety of olaparib combined with eribulin in patients with advanced or metastatic triple negative breast cancer (TNBC) previously treated with anthracyclines and taxanes: The final analysis of a Japanese phase I/II trial (ID 2638)

Presentation Number
251P
Lecture Time
13:15 - 13:15
Speakers
  • K. Aogi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Prognosis of metastatic TNBC is poorer compared with those of other subtypes, because of the lack of effective treatment target. TNBC contains molecularly heterogeneous phenotypes, some of which are influenced by germline (g) BRCA1/2 mutation. Eribulin is one of standard treatments for metastatic TNBC and olaparib (Lymparza™), a PARP (poly ADP ribose polymerase) inhibitor, has shown remarkable efficacy in BRCA mutated breast cancer. Therefore, we evaluated the efficacy and safety of the combination of these drugs in a phase I/II trial (UMIN00009498) with an expectation of synergistic effect.

Methods

In phase I, we have determined the recommended dose as 300mg bid of olaparib twice daily and 1.4 mg/m2 of eribulin intravenously on day 1 and 8 in 21-day cycle. The primary efficacy endpoint in phase II was tumor response rate (RR) in the central review. The planned size was 24, with one-sided alpha of 0.1, power of 0.8, expected RR of 0.3 and threshold of 0.1.

Results

Twenty-four patients were enrolled from June 2014 to December 2014 in phase II. The median age was 46 years old (range: 27 to 73). The median number of prior chemotherapy regimens was 3 (range: 2 to 6). Sixteen patients (66.7%) had visceral metastasis and 8 had non visceral metastasis. Dose intensity of eribulin was 0.69 (mg/m2·week) and that of olaparib was 2086.2 (mg/week). RR in central review was 29.2% (80%CI: 17.0-44.2), including 7 with PR, 7 with SD, 7 with PD and 3 with NE; thus, the null hypothesis was rejected. In institutional decision, RR was 37.5% (9/24; 95%CI, 18.8-59.4), including1 patient with CR. Median progression-free survival was 4.2 months (95%CI: 3.0 to 7.4). Median overall survival was 14.5 months (95%CI: 4.8 to 22.0). Safety was analyzed separately for phase I and II. Significant severe adverse events ( > =G3) were leukocytopenia (87.5%, 83.3%), neutropenia (87.5%, 83.3%), febrile neutropenia (20.8%, 33.3%), anemia (16.7%, 41.7%) and thrombosis (0%, 8.3%), respectively.

Conclusions

The combination of olaparib and eribulin was well tolerated and showed a promising efficacy and safety for metastatic TNBC.

Clinical trial identification

release date: 31/March/2017 (in Japanese)

Legal entity responsible for the study

National Cancer Hospital () National Cancer Center National Cancer Center

Funding

Exploratory Oncology Research & Clinical Trial Center in National Cancer Center

Disclosure

K. Aogi: Personal fees as honoraria from Eisai, AstraZeneca, Taiho Pharmaceutical, Daiichi Sankyo, Mochida Pharmaceutical, Ono Pharmaceutical, and Eli Lilly Japan. Research funds from Chugai Pharmaceutical, Eisai and Sanofi. K. Yonemori: Personal fee as honorarium from Eisai. M. Takahashi: Personal fees as honoraria from Chugai Pharmaceutical, Eisai, and AstraZeneca. N. Masuda: Personal fees as honoraria from Chugai Pharmaceutical and AstraZeneca, and his institution received research funds from Chugai Pharmaceutical and Eisai. Y. Naito: Honoraria from Eisai, Chugai Pharmaceutical, Taiho Phamaceutical, Novartis Pharma, Eli Lilly Japan, Meiji Seika Pharma, and Bayer Yakuhin. He received research funds from Merck Serono, AstraZeneca, Eli Lilly Japan, and Nippon Kayaku. Y. Fujiwara: Honoraria from AstraZeneca, Eisai, Daiichi Sankyo, Taiho Pharmaceutical, Chugai Pharmaceutical, and Eli Lilly Japan. He received grants from Taiho, Takeda, Chugai, Eli Lilly Japan, and Nippon Kayaku. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

252P - Phase II study of gemcitabine, trastuzumab, and pertuzumab for HER2-positive metastatic breast cancer after prior pertuzumab-based therapy (ID 1747)

Presentation Number
252P
Lecture Time
13:15 - 13:15
Speakers
  • N. Iyengar
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The combination of taxanes with trastuzumab (H) and pertuzumab (P) for first line treatment of HER2-positive metastatic breast cancer (MBC) is associated with improved progression-free (PFS) and overall survival (OS). Treatment per physician’s choice with anti-HER2 therapy after second line therapy is associated with a median PFS of 3 months. While continued use of H in therapeutic combinations after progression on H-based therapy is standard, the efficacy of continuing HP-based treatment after progression on P-based therapy is unknown.

Methods

This is a single arm phase II trial of gemcitabine (G) with HP. Eligible patients (pts) had HER2-positive (IHC 3+ or FISH ≥ 2.0) MBC with prior HP-based treatment and ≤ 3 prior chemotherapies. Pts received G (1200 mg/m2) on days 1 and 8 of a q 3 week (w) cycle, and H (8 mg/kg load 6 mg/kg) and P (840 mg load 420 mg) q3w. The primary endpoint is PFS at 3 months. Secondary endpoints include OS, safety and tolerability. An exploratory endpoint is to compare PFS by RECIST criteria versus 18-F FDG-PET response criteria. The study therapy will be considered successful if at least 27/45 (60%) patients are progression free at 3 months.

Results

As of 27 April 2017, 45 of 45 pts are enrolled; 39 are evaluable at 3 months and 6 have not had 3-month evaluation. At 3 months, 30/39 (77%) are progression free (1 CR, 8 PR, 21 SD); 9 pts progressed. There are no cardiac or febrile neutropenic events to date. 4 pts required G dose reduction (3 grade 3 neutropenia and 1 grade 3 vomiting) and the study was amended to lower initial G dose to 1000 mg/m2.

Conclusions

The preliminary 3 month-PFS is 77% in evaluable pts (95% CI 62% to 87%). The updated 3 month-PFS results will be presented. Continuation of P beyond progression is associated with apparent clinical benefit. A randomized trial is justified to confirm this clinically important observation.

Clinical trial identification

NCT02252887

Legal entity responsible for the study

Memorial Sloan Kettering Cancer Center

Funding

Genentech/Roche

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

253P - Abemaciclib plus fulvestrant in patients (pts) with HR+/HER2- endocrine therapy naïve (EN) advanced breast cancer - an exploratory analysis of MONARCH 2 (ID 4124)

Presentation Number
253P
Lecture Time
13:15 - 13:15
Speakers
  • P. Kaufman
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

MONARCH 2 demonstrated that the addition of abemaciclib, dosed on a continuous schedule at 150 mg twice daily, to fulvestrant (F) significantly improved progression-free survival (PFS) and objective response rate (ORR) compared to placebo (P) plus F (PFS hazard ratio [HR], 0.553, P<.0000001; ORR in measurable disease 48.1% vs 21.3%, P<.001) in women with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer (ABC) who had progressed on endocrine therapy (ET). Here we present efficacy and safety findings from an exploratory cohort of EN pts from MONARCH 2 not included in the intent-to-treat (ITT) population.

Methods

Pts were randomized 2:1 to receive abemaciclib or P + F (500 mg, per label). Pre/perimenopausal women received a gonadotropin-releasing hormone agonist. Pts were not allowed to have had chemotherapy in the advanced setting. Pts were stratified by metastatic site (visceral, bone only, other). The primary endpoint for this analysis was investigator-assessed PFS which was described using the Kaplan-Meier method and a Cox model.

Results

Forty-four EN pts were randomized to abemaciclib + F (N = 28) or P + F (N = 16). 46% of pts presented with visceral disease, 77% with measurable disease, and 82% were postmenopausal. At the time of the analysis, 18 pts were still on treatment (13 [46.4%] in the abemaciclib + F arm and 5 [31.3%] in the P + F arm). 18 PFS events were observed (9 [32.1%] in the abemaciclib + F arm and 9 [56.3%] in the P + F arm). The median PFS had not been reached in the abemaciclib + F arm and was 23.1 months in the P + F arm (stratified HR, 0.454; 95% CI: 0.179, 1.154). The ORR in pts with measurable disease was 60.0% (5% complete response [CR]) in the abemaciclib + F arm and 57.1% (0% CR) in the P + F arm. The most common adverse events were diarrhea, nausea, fatigue, and neutropenia. Diarrhea generally occurred in the early cycles and was managed with dose adjustment and conventional anti-diarrheal medication.

Conclusions

In this exploratory cohort of EN pts, the addition of abemaciclib to fulvestrant demonstrated a comparable increase in PFS and consistent safety results to those observed in the ITT population in MONARCH 2.

Clinical trial identification

NCT02107703

Legal entity responsible for the study

Eli Lilly and Company

Funding

Eli Lilly and Company

Disclosure

P.A. Kaufman: Consulting/Advisory: Galena Biopharma, Amgen; Research Funding: Eli Lilly and Company. P. Nikolinakos: Advisory board: AstraZeneca. J.T. Beck: My institution receives research grants as part of this study. J.L. Gonzalez-Trujillo: Contract with Lilly, Bristol-Myers Squibb, Roche, Pierre Fabre, Astra for make a corporate-sponsored research. Advisory board for Bristol-Myers Squibb, MSL, Novartis. Speaker for Bristol-Myers Squibb, Novartis. Y. Lin, N. Bourayou: Employment and Stock Ownership: Eli Lilly and Company. S. Barriga: Employment Eli Lilly and Company. D. Headley: Employment and Stock Ownership: Eli Lilly and Company; Stock Ownership: AstraZeneca. A. Llombart Cussac: Honoraria: Roche, Novartis, Pfizer; Consulting/Advisory: Roche, AstraZeneca, Eli Lilly and Company; Research funding: Pfizer, Roche; Travel funds: Roche, Celgene. G.W. Sledge Jr.: Leadership, Stock: Syndax; Honoraria: Symphogen; Consulting/Advisory: Symphogen, Coherus Biosciences, Radius Health, Peregrine Pharmaceuticals, Taiho Pharmaceutical; Research Funding: Roche (Inst); Travel funds: Nektar, Radius Health, Taiho Pharmaceutical. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

254P - Matching-adjusted indirect treatment comparison of ribociclib and palbociclib as first-line treatments for HR+, HER2– ABC (ID 3005)

Presentation Number
254P
Lecture Time
13:15 - 13:15
Speakers
  • A. Forsythe
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Ribociclib (RIB) and palbociclib (PAL) combinations with letrozole (LET) vs LET alone have been evaluated in separate Phase 3 randomized controlled trials in patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2–) advanced breast cancer (ABC); however, no head-to-head comparative studies have been conducted. Classic indirect treatment comparison (ITC) can lead to biased results due to differences in patient populations and trial designs. These differences can be corrected by using the matching-adjusted indirect comparisons (MAIC) technique. Here, we compare RIB and PAL in patients with HR+, HER2– ABC using MAIC.

Methods

Individual patient data were available for the RIB trial (MONALEESA-2). As only published summary data were available for the PAL trial (PALOMA-2), RIB data were adjusted to closely match the PAL data. Data for RIB-treated patients were assigned weights so that weighted mean baseline characteristics matched those reported for PAL. Overall survival data have not been reported for PALOMA-2, thus only progression-free survival (PFS) data were compared. Adjusted hazard ratios (HRs) for PFS were calculated using weighted Cox regression models and used to calculate indirect HRs with 95% confidence intervals (CIs). Classic frequentist ITC was performed before and after adjustment. ITC of Grade 3/4 adverse events (AEs) was also performed.

Results

The unadjusted PFS HR (95% CI) for RIB+LET vs LET was 0.556 (0.429; 0.721) and for PAL+LET vs LET was 0.580 (0.460; 0.720). MAIC adjustment for age, race, region, Eastern Cooperative Oncology Group status, disease stage at diagnoses, sites of metastases, and chemotherapy setting at baseline provided a RIB+LET vs LET HR estimate of 0.501 (0.365; 0.688). The HR for unadjusted ITC of RIB vs PAL was 0.959 (0.681; 1.350) and by MAIC was 0.864 (0.586; 1.274). ITC of Grade 3/4 AEs yielded a risk ratio (RR) of 0.81 (0.61; 1.08) for RIB vs PAL.

Conclusions

Using MAIC methodology due to a lack of head-to-head trials, the resulting HRs for PFS were comparable. Similarly, using ITC, AE profiles were also comparable although the RR for AEs slightly favored RIB.

Legal entity responsible for the study

Novartis Pharmaceuticals Corporation

Funding

Novartis Pharmaceuticals Corporation

Disclosure

A. Forsythe: Consultant for Novartis. D. Chandiwana, M. Monaco: Novartis employee and stocks/shares. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

255P - Phase 1 dose-escalation study of single-agent ZW25, a HER2-targeted bispecific antibody, in patients (pts) with HER2-expressing cancers (ID 3207)

Presentation Number
255P
Lecture Time
13:15 - 13:15
Speakers
  • M. Beeram
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

HER2 is expressed on many cancers (ca) at varying levels. ZW25, a novel bispecific antibody, targets the same HER2 domains as trastuzumab (T) and pertuzumab (P). In preclinical studies ZW25 shows increased binding, internalization and anti-tumor activity relative to T in models of low to high HER2.

Methods

3 + 3 dose escalation study in pts with HER2 IHC 1-3+ or FISH+ ca, who have progressed after standard treatment (tx) including HER2 targeted agents, and have measurable or non-measureable disease per RECIST 1.1. ZW25 was given weekly (QW; 5, 10 or 15 mg/kg) or biweekly (Q2W; 20 mg/kg) in 4 week cycles. Assessments included HER2 status (local and central read), adverse events (AEs), tumor response, PET scan, immunogenicity and PK.

Results

16 pts (8 breast; 6 gastric/esophageal ≥; 1 colorectal [CRC]; 1 adnexal) have received ZW25 QW at 5 (n = 3), 10 (n = 6) and 15 mg/kg (n = 7); 20 mg/kg Q2W is enrolling. All pts had prior T; breast ca pts also had T-DM1 (n = 8), P (n = 6) or lapatinib (L; n = 6). 12 pts had IHC 3+/FISH+ ca (local and central read); 1 CRC ca was HER2 heterogeneous (IHC 3+/FISH+ and IHC 0/FISH -), and 3 GE were IHC 1 + (central read). No dose limiting toxicities were observed. The most common AEs were Diarrhea (44%, all grade 1), Infusion reaction (IR) (44%, grade 1 and 2), and Fatigue (31%). PK was non-linear and increased with dose of ZW25. 8/13 pts tested had anti-drug antibodies (ADA) at baseline. No increases in titers and no new detectable ADA were seen. In 7 breast ca pts (1 too early), best overall response (BoR) was 2 PR (both 10 mg with prior T, P, T-DM1, L); 2 SD (>8 cycles in 5 mg pt with prior T, P, T-DM1; >2 cycles in 15 mg pt, with T, P, T-DM1, L); and 3 PD. BoR in 5 GE pts (1 too early): 1 SD > 4 cycles (10 mg, prior T, IHC 3+) and 4 PD. BoR in adnexal (10 mg) and CRC (15 mg) pts was PD.

Conclusions

ZW25 was well tolerated with promising single-agent activity including SD > 6 mo and PR in pts with HER2 expressing ca who have progressed after standard tx, including multiple lines of prior HER2 targeted agents. These early signs of activity support the therapeutic potential of ZW25.

Clinical trial identification

NCT02892123

Legal entity responsible for the study

Zymeworks Inc

Funding

Zymeworks Inc

Disclosure

M. Beeram, D. Rasco: Employee of START. D. Hausman: Stockholder and employee Zymeworks Biopharmaceuticals Inc. R. Korn: Employee and leadership role at Imaging Endpoints. G. Rowse: Employee of Zymeworks Inc. J. Thimmarayappa: Employee of Zymeworks Biopharmaceuticals Inc. A. Tolcher: Employee of START, leadership role at Symphogen, member of Zymeworks Clinical Advisory Board. F. Meric-Bernstam: Honoraria: Genentech, Roche Diagnostics. Consulting or Advisory Role: Celgene, Genentech, Inflection Biosciences, Novartis, Roche. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

256P - Efficacy of palbociclib plus fulvestrant in advanced Hormone Receptor-positive (HR+) metastatic breast cancer (MBC) pretreated with everolimus: Real-life data from the french temporary authorization for use (TAU) at the Institut de Cancérologie de l’Ouest (ID 3674)

Presentation Number
256P
Lecture Time
13:15 - 13:15
Speakers
  • P. Du Rusquec
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The CDK4-6 inhibitor palbociclib, combined with hormonal therapy is a new standard of treatment for HR+ Metastatic Breast Cancer. Before the European Medicines Agency approval, a Temporary Authorization for Use (TAU) has been set up in France restricted to patients pretreated with everolimus. We present the efficacy data of this combination in this population.

Methods

Between November 2015 and November 2016, all the patients treated with palbociclib + fulvestrant according to the TAU in our institution were prospectively included. Datas from their medical records and adverse events (AE) were collected.

Results

60 patients received at least one dose of palbociclib in combination with fulvestrant with a median age of 61 years. 50 patients (83.3%) had visceral metastasis and 10 (16.7%) had bone only disease. Patients had received an average of 5.3 lines of treatment before palbociclib initiation, including hormonal therapy (mean = 3.0) and chemotherapy (mean = 2.3). Of note, 28 patients (46.7%) had received fulvestrant previously and all had been pretreated with everolimus. With a median follow-up of 8.1 months, median progression free survival (PFS) was 6.1 months (95% CI, 4.2 to 7.4) and median overall survival was not reached. PFS was the same according to the presence of visceral metastasis or no (HR 1.46 (95% CI, 0.57 to 3.74), p = 0.42). Interestingly, patients treated previously with fulvestrant and subsequently re-challenged with fulvestrant and palbociclib had a PFS of 6.4 months, which was similar to patients who didn’t receive fulvestrant previously (HR = 1.00 (95% CI 0.55 to 1.83), p = 1.00). The most common AE were neutropenia (n = 53), thrombocytopenia (n = 25) and anemia (n = 20). At the time of this analysis (April 2017), 36 patients received a further line of treatment after progression.

Conclusions

In this heavily pretreated population, the association of fulvestrant plus palbociclib provides an interesting median PFS of 6.1 months. Patients previously treated with fulvestrant seem to derive the same magnitude of benefit compared to fulvestrant naive patients.

Legal entity responsible for the study

Institut de Cancérologie de l'Ouest

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

257P - A phase 1 study of BYL719, an α-isoform selective PI3K inhibitor, in Japanese patients with advanced solid malignancies (ID 1405)

Presentation Number
257P
Lecture Time
13:15 - 13:15
Speakers
  • J. Tomomatsu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

BYL719 is an oral inhibitor that selectively targets the α-isoform of class l PI3K. In a first-in-human ph1 study in mostly Western patients (pts) with PIK3CA alteration, the maximum tolerated dose for once-daily (qd) BYL719 was declared as 400 mg and preliminary antitumor activity was observed. Previous preliminary findings showed tolerability, safety, and pharmacokinetic (PK) results in dose escalation of the first-in-Japanese ph1 study. Here, we report results of the food effect on the PK profile of BYL719 at steady state, additional safety, and preliminary efficacy in Japanese pts.

Methods

Pts were aged ≥18 years with histologically confirmed, advanced solid tumors. Pts received BYL719 qd in 28-day cycles until disease progression, unacceptable toxicity, or investigator/pts decision. The objectives in the expansion part were to assess food effect on PK profile, preliminary antitumor activity, and safety. Pts with PIK3CA alteration were selected in the expansion part and were randomized to receive BYL719 at the recommended dose 350 mg qd in fasted or fed condition on cycle 1 day 22 and cycle 2 day 1 in a crossover fashion. Pts received BYL719 ∼1 hr following a light breakfast and continued to be fasted for 1 hr after each dose.

Results

Thirty-three pts were enrolled and all pts discontinued treatment. The median duration of exposure was 71.0 days (range, 6-462). The common BYL719-related all-grade (Gr) AEs (>30%) were hyperglycemia, rash maculopapular (48.5%, each), diarrhea (45.5%), and decreased appetite (33.3%). BYL719-related Gr 3 or 4 AEs (≥20%) were rash maculopapular (24.2%) and hyperglycemia (21.2%). Eight pts were enrolled in the expansion part; six of them were eligible for PK analysis. The geometric mean values of dose-normalized Cmax and AUC0-24 in fed state were 78% and 56% higher than those in fasted state, respectively. One pt experienced Gr 4-infected neoplasm meeting DLT criteria, 1 pt with uterus cancer and PIK3CA mutation had an unconfirmed partial response, and 18 pts had a stable disease.

Conclusions

In this ph1 study of BYL719 in Japanese pts, a positive food effect and preliminary antitumor activity were observed at steady state in pts with PIK3CA mutation status.

Clinical trial identification

NCT01387321

Legal entity responsible for the study

Novartis Pharmaceuticals KK, Tokyo, Japan

Funding

Novartis Pharmaceuticals KK, Tokyo, Japan

Disclosure

H. Saka: Grants from Novartis, KHK, Daiichi Sankyo, Merck, Esai, Bristol-Myers Squibb, Taiho, Ono, Chugai, Eli Lilly, Beyer, MSD, Quintiles, West JCOG. Personal fees Chugai, Kyorin, NU, BI, Eli Lilly, Astellas, Nobelpharma, JSRE, Ono, Chunichi Shimbun, Taiho. S. Takahashi: Grants from Novartis, during the conduct of the study; grants from Chugai, grants from Astrazeneka, grants from Daiichisankyo, grants from Bayer, grants from Parexel, outside the submitted work. K. Natsume, T. Kakizume: Personal fees from Novartis Pharmaceuticals KK, during the conduct of the study; Y. Ando: Grants and personal fee: Chungai, Takeda, KHK, Eisai, Taiho, Nippon, YakultHonsya, Mochida, Merck, Ono, Eli Lilly, Novartis, Janssen, Hisamitsu, GSK, Terumo, Bayel, Meiji, BSC, Boehringer Ingelheim, Bristol-Myers Squibb, Sawai, Otsuka, Shionogi, outside the submitted work. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

258P - Phase 1 study of RX-5902, a novel orally bioavailable inhibitor of phosphorylated P68, which prevents β-catenin translocation in advanced solid tumors (ID 3962)

Presentation Number
258P
Lecture Time
13:15 - 13:15
Speakers
  • J. Diamond
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

RX-5902 is a novel compound that targets phosphorylated p68 RNA helicase (also known as DDX5), a member of the DEAD box family of RNA helicases. Phosphorylated p68 may play a vital role in cell proliferation and tumor/cancer progression through inhibition of β-catenin translocation. We report preliminary results of the Phase 1 study of RX-5902 as a single agent to treat advanced solid tumors.

Methods

The dose finding portion of the Phase 1 study (NCT02003092) was designed to evaluate safety, tolerability and dose limiting toxicities, to identify the maximum tolerated dose and a recommended phase 2 dose and schedule (RP2D). Secondary objectives were pharmacokinetics (PK) and antitumor activity (RECIST v1.1). Eligible subjects (aged ≥ 18 years), with relapsed/refractory solid tumors that had been heavily pretreated, received oral RX-5902 ranging from 25 mg to 350 mg and administered at 1, 3, 5 or 7 times per week for 3 weeks followed by 1 week of rest or for 4 weeks without rest.

Results

As of May 2017, 35 subjects (23 Females, 12 males) were treated with oral RX-5902. The dose limiting toxicities were G4 hyponatremia (n = 1) and G3 fatigue (n = 1) at 300 mg administered daily for 4 weeks. The maximum tolerated dose of 250 mg, will be studied further in the Phase 2a portion. Of the 35 subjects treated, 15 subjects (breast ER+/PR+/Her2-, triple negative breast (n = 2), cervical (n = 2), neuroendocrine (n = 3), paraganglioma, colorectal (N = 3), pancreatic, ovarian, head and neck cancers) experienced stable disease; 2 subjects have received treatment for > 2.5 years. The most common related adverse events were G1/G2 anorexia, G1/G2 nausea, G1/G2 vomiting, G1/G2 diarrhea, G1 weight loss and G1/G2 fatigue. Oral RX-5902 was bioavailable with median Tmax of 2 hours and median elimination half-life of 12 hours.

Conclusions

Data from this study support that RX-5902 is safe and well-tolerated at the doses and schedules tested. The RP2D of 250 mg of RX-5902 administered daily for 5 consecutive days for 4 weeks is being studied further in metastatic triple negative breast cancer in the Phase 2 portion of this study.

Clinical trial identification

NCT02003092

Legal entity responsible for the study

Rexahn Pharmaceuticals, Inc.

Funding

Rexahn Pharmaceuticals, Inc

Disclosure

C. Peterson, R. Pila, E. Benaim: Employee of Rexahn Pharmaceuticals, Inc. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

259P - Adherence to International ESO-ESMO (ABC) guide-lines in HER2-ve metastatic breast cancer (MBC) patients (pts): Preliminary results of the GIM 13 - AMBRA Study (ID 1830)

Presentation Number
259P
Lecture Time
13:15 - 13:15
Speakers
  • M. Cazzaniga
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

ESO/ESMO develops consensus guidelines for MBC treatment every 2 years, potentially applicable worldwide. Aim of the present analysis is to verify the adherence to ABC recommendations for HER2-ve MBC in the context of the AMBRA study.

Methods

AMBRA is a longitudinal cohort study, aiming to describe the choice of first and subsequent lines of treatment in HER2-ve MBC pts receiving at least one Chemotherapy (CHT) (SABCS 2016, P5-15-07 & P5-14-09). For the present analysis, we selected 4 statements from the ABC1 & ABC2 Conferences, comparing them with the clinical choices of 1st-line therapy in all evaluable cases.

Results

So far, 791 pts have been enrolled, of whom 586 (74.1%) have received CHT as 1st-line treatment. 479 pts (81.7%) had Luminal tumours at diagnosis. First-line CHT was monotherapy in 89 pts (25.2%), anthra-based CHT in 45 (7.7%) and platinum-based in 38 (6.5%), mainly TNBC (20/38, 52.6%). Selected recommendations and percentages of adherence are reported in Table.

259P Adherence to ABC recommendations for HER2-ve pts

n/N (%of adherence)
1 - Anthracycline (A)- or Taxane (T)-based regimens, preferably as a single agent, would usually be considered as first-line CHT, in those pts who have not received these regimens as adjuvant treatmentA/T mono 59/ 270 (21.8%)160/270 (59.2%)
A/T poly 101/ 270 37.4%)
2 - In pts with taxane-naive and anthracycline-resistant MBC, taxane-based therapy, preferably as a single agent, would usually be considered as the treatment of choiceT mono 89/ 431 (20.6%)208/431 (48.2%)
T poly 119/ 431 (27.6%)
3 - Other options are Capecitabine and Vinorelbine135/586 (23.0%)
4 - If given in the adjuvant setting, a taxane can be re-used in the metastatic setting, particularly if there has been at least 1 year of disease-free survivalDFI > 12 months 74/79 (93.7%)79/151 (52.3%)
DFI

Conclusions

The adherence to clinical recommendations is very low, being only partially applied in all the clinical situations analyzed. Educational interventions are urgently needed and a confrontation with the clinical community is strongly recommended.

Legal entity responsible for the study

Marina E. Cazzaniga

Funding

Celgene Intl.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

260P - M-bioscore: A practical tool for predicting outcomes among patients with previously untreated metastatic breast cancer (ID 3374)

Presentation Number
260P
Lecture Time
13:15 - 13:15
Speakers
  • O. Abdel-Rahman
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Two prognostic models “bioscore” and “Neo-bioscore” were recently published and validated to help predict the outcomes of patients with non-metastatic breast cancer treated with either upfront surgery or upfront neoadjuvant chemotherapy. A comparable model for metastatic disease is yet to be developed. The current study thus sought to propose and validate a third model “M-bioscore” to help predict the outcomes of treatment-naïve patients with metastatic breast cancer.

Methods

Through SEER*Stat program, surveillance, epidemiology and end results (SEER) database (2010-2013) was accessed. The resulting cohort was equally split into two halves: training set (to guide model development) and validation set (to test the model prediction). Multivariate analysis for the candidate prognostic factors (extent of metastases, estrogen receptor (ER), progesterone receptor (PR), HER2 neu and nuclear grade) was conducted through a Cox proportional model. M-bioscore was then calculated for each patient. Cancer-specific survival analyses according to M-bioscore were conducted through Kaplan-Meier analysis/log-rank testing.

Results

A total of 6655 patients with previously untreated metastatic breast cancer and complete data were identified in the period from 2010-2013. The following factors were associated with better cancer-specific survival in multivariate analysis in the training set (isolated distant nodal metastases, ER positivity, PR positivity, HER 2 neu positivity and lower nuclear grade) (P < 0.01). This has been shown for both training and validation sets. accordingly, the M-bioscore model has been proposed as follows: an M-bioscore point is given for each of ER negativity, PR negativity, HER2 neu negativity and nuclear grade 3. The site/distribution of metastasis are given the following points: 0 for isolated distant lymph nodes, 1 for isolated bone/skin deposits, 2 for isolated liver/lung deposits, 3 for isolated brain deposits or multiple sites of metastases. A total M-bioscore was then calculated for each case in the training cohort (which may range from 0 to 7). After assignment of a total M-bioscore for each patient, cancer-specific survival was compared according to the score. Log rank testing with pair wise comparisons between all different scores was conducted. For cancer-specific survival assessment according to the M-bioscore, P values for pair wise comparisons among different score points were significant (P <0.05) except for the comparison between score 0 and score 1/score 2/score 3 in the training cohort. These findings have been confirmed in the validation and overall cohorts. Table shows the three year cancer-specific survival (CSS) rates for patients in the overall cohort according to M-bioscore.

260P Three-year cancer-specific survival according to M-bioscore

M-bioscoreAll patients
N (%)CSS rate
022 (0.3%)95%
1273 (4.1%)81%
21567 (23.5%)69%
31202 (18.1%)60%
41680 (25.2%)52%
51038 (15.6%)35%
6590 (8.9%)26%
7283 (4.3%)17%

Conclusions

M-bioscore is a novel, easy and reliable tool for predicting the outcomes of patients with previously untreated metastatic breast cancer. Further external validation within the context of other population-based cohorts is recommended.

Legal entity responsible for the study

Omar Abdel-Rahman

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

261P - Is PFS a more relevant endpoint than OS in 1L HR+, HER2– MBC? A systematic literature review (ID 3014)

Presentation Number
261P
Lecture Time
13:15 - 13:15
Speakers
  • A. Forsythe
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Hormone receptor-positive (HR+), human epidermal growth factor 2 receptor-negative (HER2–) metastatic breast cancer (MBC) accounts for 73% of all MBC.1 Endocrine therapy (ET) is the basis of first-line (1L) therapy for patients (pts) with HR+, HER2– MBC; however, efficacy is limited by ET resistance. Novel therapies have demonstrated improvements in progression-free survival (PFS) vs standard ET. The clinical relevance of PFS is debated due to a lack of direct correlation with overall survival (OS) benefit, and cases of asymptomatic progression. We review studies of HR+ HER2– MBC to assess factors that influence OS and treatment response, and changes in health-related quality of life (HRQoL).

Methods

The Embase®, MEDLINE®, and Cochrane databases were systematically searched to identify studies in adult women with HR+, HER2– MBC, published 2006–January 2017, and written in English. Phase (Ph) 2 and 3 randomized controlled-trials (RCTs), observational, and retrospective studies were considered and HRQoL and real-world evidence reviewed.

Results

79 RCTs were identified: 58 (73%) in the 1L setting and 21 (27%) in the ≥second-line setting. PFS data were reported in 61 (77%) studies; 31 (51%) reported significant PFS improvement. OS was reported in 44 (56%) of studies; only 11 (14%) reported a significant OS improvement. Significant improvements in both PFS and OS were reported in only 6 (8%) studies (1 Ph 2; 5 Ph 3). Pts with HER2– MBC received on average ≥5 lines of therapy, with no defined treatment pathway. Baseline characteristics, prior therapies, and the type and number of post-progression therapies significantly impacted OS. PFS, response rates, and HRQoL decreased with each line of therapy (EQ-5D: 0.78 1L vs 0.70 post-progression).

Conclusions

Multiple HR+, HER2– MBC therapies have been investigated yet few CTs have achieved a significant improvement in OS. Multiple factors besides the choice of 1L therapy impact OS, such as post-progression therapies, which cannot be controlled in RCTs. This study emphasizes the importance of PFS improvement coupled with HRQoL maintenance in 1L treatment of HR+, HER2– MBC. 1. Howlader N et al. J Natl Cancer Inst 2014;106:dju055.

Legal entity responsible for the study

Novartis Pharmaceuticals Corporation

Funding

Novartis Pharmaceuticals Corporation

Disclosure

A. Forsythe: Consultant for Novartis. D. Chandiwana, M. Thabane, J. Baeck: Novartis employee and Novartis stocks/shares. J. Barth: Novartis employee. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

262P - PFS/TTP as a potential surrogate for OS in HR+, HER2– MBC (ID 3015)

Presentation Number
262P
Lecture Time
13:15 - 13:15
Speakers
  • A. Forsythe
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Several, recent randomized controlled trials (RCTs) in hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2–) metastatic breast cancer (MBC) have demonstrated a significant improvement in progression-free survival (PFS); however, few have reported an improvement in overall survival (OS). OS may be an imperfect endpoint due to the impact of factors such as baseline characteristics and subsequent therapies. Investigation of the use of PFS or time to progression (TTP) as a surrogate for OS in HR+, HER2– MBC has been limited. This study assesses the correlation of PFS/TTP and OS in HR+, HER2– MBC across all lines of therapy.

Methods

A systematic literature review of RCTs in HR+, HER2– MBC was conducted to identify studies that reported both median PFS/TTP and OS. The correlation between PFS/TTP and OS was evaluated using Pearson’s product-moment correlation and Spearman’s rank correlation. Subgroup analyses were performed to explore possible reasons for heterogeneity. Errors-in-variables weighted least squares regression (LSR) was used to model incremental OS months as a function of incremental PFS/TTP months. An exploratory analysis investigated the impact of 3 covariates (chemotherapy vs other, PFS vs TTP, and 1L vs > 1L) on the use of PFS/TTP in OS prediction. The lower 95% prediction band was used to determine the minimum incremental PFS/TTP months below which there would be no predicted OS benefit (the surrogate threshold effect [STE]).

Results

A total of 39 studies were identified. There was a statistically significant correlation between median PFS/TTP and OS (Pearson=0.741, p < 0.000; Spearman=0.650, p < 0.000). Results were unchanged for chemotherapy and hormonal or targeted therapy, and for line of therapy. Initial LSR analysis yielded an R2 of 0.354; 1 PFS/TTP month corresponded to 1.13 OS months. The addition of 3 covariates improved R2 to 0.569; 1 PFS/TTP month corresponded to 0.78 OS months. The STE for OS benefit was 5–6 months of incremental PFS/TTP.

Conclusions

Results of this study indicate a significant association between PFS/TTP and OS, which may justify the use of PFS/TTP as surrogate for OS benefit during regulatory approval and subsequent reimbursement of new therapies in HR+, HER2– MBC.

Legal entity responsible for the study

Novartis Pharmaceuticals Corporation

Funding

Novartis Pharmaceuticals Corporation

Disclosure

A. Forsythe: Consultant for Novartis. D. Chandiwana, M. Thabane, J. Baeck: Novartis employee and Novartis stocks/shares. J. Barth: Novartis employee. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

263P - Survival gains from advances in first-line systemic therapy for HER2-positive metastatic breast cancer in the U.S., 1995-2015 (ID 5269)

Presentation Number
263P
Lecture Time
13:15 - 13:15
Speakers
  • J. Roth
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Of the approximately 67,000 women diagnosed with metastatic breast cancer (mBC) annually in the U.S., nearly 10,700 are HER2-positive (HER2+). Prior to the advent of HER2 targeted therapy, women with HER2+ mBC had a less favorable prognosis than those with HER2-negative mBC. The objective of this study was to quantify survival gains for women with HER2+ mBC from 1995-2015, a period spanning pre HER2 targeted therapy to the present, where multiple targeted therapies are available.

Methods

We developed a simulation model to estimate overall survival (OS) for successive cohorts diagnosed with HER2+ mBC from 1995-2015. OS data were derived from clinical trials referenced in NCCN guidelines and extrapolated to a lifetime horizon by fitting Weibull curves. Patients were assigned to regimens in each diagnosis year using information from the IPSOS ‘Global Oncology Monitor’- a commercial treatment registry. Results were calibrated with SEER OS data. Outcomes included 5-year OS, expected OS, and total life years (LYs) for all patients with HER2+ mBC. Annual incidence of HER2+ mBC was assumed to be constant over time.

Results

Over the period 1995-2015, expected 5-year OS for women with HER2+ mBC increased by 28.9%. Mean expected per-patient OS improved by 28.1 months (25,000 population LYs) (see Table). The largest gain (15.7 months) occurred between 2010 and 2015–the period over which pertuzumab-based treatment was approved for first-line use and gained substantial market penetration. The smallest gain (2.4 months) occurred between 1995 and 2000—the period over which trastuzumab-based treatment was approved, but had not yet achieved market penetration.

Conclusions

The introduction and expanded use of targeted treatments, along with other advances in care, have provided substantial individual- and population-level survival gains for women with HER2+ mBC. A considerable proportion of the expected OS differences between 1995 and 2015 took place after the introduction and uptake of trastuzumab and pertuzumab-based regimens.

263P

Diagnosis Year5-Year OS (%)Mean Per-Patient OS (Months)Population Life Years
201538.855.349300
19959.927.224300
Difference+28.9+28.1+25000

Clinical trial identification

Not applicable

Legal entity responsible for the study

Genentech

Funding

Genentech

Disclosure

J. Roth, S. Sullivan, S. Ramsey: Research funding from Genentech, Inc. P. Bajaj, C. Reyes, V. Antao, A. Stein: Employee of Genentech, Inc; Stock Ownership in Roche. R. Mahtani: Advisory boards for Genentech, Pfizer, Amgen, Celgene, Sandoz; Research support from Genentech.

Collapse
Poster display session Poster Display session

264P - Progression-free survival (PFS) and site of first progression in HER2+ metastatic breast cancer (MBC) patients (pts) with (w) or without (w/o) brain metastases: A pooled analysis of tucatinib phase I studies (ID 3188)

Presentation Number
264P
Lecture Time
13:15 - 13:15
Speakers
  • S. Moulder
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Brain metastases (BM) are frequent in HER2+ MBC occurring in > 30% of pts and are associated w significant neurologic morbidity and mortality. Current treatment strategies for BM primarily utilize radiotherapy (RT) and in selected instances surgical resection. Tucatinib is a highly selective oral HER2+ tyrosine kinase inhibitor that has shown promising results in HER2+ MBC both in pts w and w/o BM.

Methods

Two Phase 1b studies of tucatinib were pooled to compare PFS and sites of relapse in pts w or w/o BM.

Results

77 pts were analyzed, all treated at the recommended Phase 2 dose of 300mg PO BID of tucatinib, 50 in the 004 trial (tucatinib + T-DM1) and 27 in the 005 trial (tucatinib + trastuzumab + capecitabine). All pts were heavily pretreated w a median of 3 prior therapies including a taxane, trastuzumab, pertuzumab, T-DM1 and lapatinib. Four cohorts of pts were identified: 46% (35) had systemic metastases only, 17% (13) had previously treated (RT w or w/o surgery) and stable BM, 19% (15) had previously treated and progressive BM and 17% (13) had asymptomatic untreated BM demonstrated by screening MRI. Median PFS across cohorts was 8.5, 6.1, 9.0 and 7.1 months, respectively. No statistically significant difference in PFS was seen when comparing the non-BM cohort to all BM cohorts (median of 8.5 vs. 6.7 months; p = 0.65). The risk of progression in brain in pts w baseline BM was 48.8% overall (41.5% in brain only; 7.3% in brain and body) compared to an 11.1% overall (8.3% in brain only; 2.8% in brain and body) in pts w/o baseline BM.

Conclusions

54% of pts entered tucatinib studies w baseline BM, either previously treated (stable or progressive) or untreated. The cohorts of pts analyzed appear to differ only in the site of disease progression. Although pts w/o baseline BM primarily have progression in extraneural sites and pts w baseline BM primarily have progression in the CNS, PFS is comparable across cohorts. Furthermore, pts both w and w/o BM have durable responses w these combination therapies following multiple lines of prior HER2 targeted therapy. These data support the use of tucatinib in both pts w and w/o BM in the accruing HER2CLIMB trial.

Clinical trial identification

ONT-380-004 and ONT-380-005

Legal entity responsible for the study

Cascadian Therapeutics, Inc.

Funding

Cascadian Therapeutics, Inc.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

265P - Survival of patients with aromatase inhibitors sensitive, HR+/HER2- metastatic breast cancer treated with a first-line endocrine therapy or chemotherapy in a multicenter national observational study (ID 3623)

Presentation Number
265P
Lecture Time
13:15 - 13:15
Speakers
  • E. JACQUET
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

For HR+/HER2– metastatic breast cancer (mBC), international guidelines recommend the use of endocrine therapy (ET) as first-line (L1) treatment except in case of “visceral crisis” for which chemotherapy (CT) is advised. Few studies directly compare these two treatment options. In 2014, UNICANCER launched the Epidemiological Strategy and Medical Economics (ESME) Research program to centralize real-world data in oncology. We sought to use this database to study this question.

Methods

All patients (pts) who initiated treatment for a newly diagnosed mBC between January 2008 and December 2014 in all 18 French Comprehensive Cancer Centers were included in the ESME mBC database. ESME Research program centralized all existing data using retrospective data collection. Primary endpoint of the present study was progression free survival (PFS1) and overall survival (OS) according to L1 treatment for aromatase inhibitors sensitive (AIS) HR+/HER2- mBC pts.

Results

6265 pts out of 16703 in ESME, had AIS HR+/HER2- mBC. As L1 therapy, 2733 pts (43.6%) received ET alone, while 3532 received CT (56.4%). Among these 3532 pts, 2073 (58.7%) received ET as maintenance treatment after CT. A Cox multivariate analysis with significant prognostic variables identified a lower risk of death in the patients with L1 ET (HR = 0.839, 95% IC [0.772-0.911], p < 0.0001). Patients receiving CT were younger (median age 56.0 vs 66.0, p < 0.001), more likely to have visceral metastasis (61.6% vs 40.1%, p < 0.001) and SBR III primary tumors (31.3% vs 18.8%, p < 0.001). Median PFS1 was 15.18 months for L1 ET (95% CI, 14.45-16.20) vs 12.58 months for L1 CT +/- hormone maintenance (95% CI, 11.89-13.14), p < 0.0001. Median OS was 60.78 months for L1 ET (95% CI, 57.16-64.09) vs 49.64 months for L1 CT (95% CI, 47.31-51.64), p < 0.0001.

Conclusions

The results show that despite guidelines, a majority of AIS HR+/HER2- mBC pts still received CT as first-line treatment in the past years. PFS1 and OS data do not suggest any advantage of this aggressive strategy over ET alone. Advanced statistical methods using the propensity score will be presented in order to control for potential selection bias.

Legal entity responsible for the study

UNICANCER

Funding

UNICANCER

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

266P - Use of everolimus in advanced hormone receptor positive metastatic breast cancer in a multicenter national observational study (ID 1914)

Presentation Number
266P
Lecture Time
13:15 - 13:15
Speakers
  • A. Lardy-Cleaud
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The everolimus-exemestane combination has been included in the International guidelines for advanced HR+ breast cancer (mBC) since the results of the Bolero-2 trial. Marketing authorization has been granted in France in July 2012. We evaluated the incidence and indication of everolimus (EVE) use before and after marketing authorization and reimbursement.

Methods

All patients who initiated treatment for a newly diagnosed mBC between 01/2008 and 12/2015 in all 18 French Comprehensive Cancer Centers have been included in the real life ESME database, which collects retrospective data using a clinical trial-like methodology.

Results

The ESME program included a total of 16,703 patients of which 9,921 had HR+/HER2- mBC. Median age at metastatic diagnosis was 62.0 year (range 23-96). Visceral metastases were present in 60.3% of cases. Only 4123 patients (41.6%) received endocrine therapy alone as first-line therapy, and 60% were deemed endocrine resistant. Overall, 1,217 (12.3%) pts have received EVE during therapy as of Dec. 2015 (all lines). EVE was given as first line therapy in 117 pts (10% of all EVE pts and 1.2% of pts receiving a first line therapy). In 99/117 pts (85%) EVE was combined with exemestane. Before 2012, EVE was used within clinical trials. After 2012, use of EVE increased steadily. Percentages in the Table refer to the total of pts who received any kind of treatment during a given year of observation (eg 506 pts took EVE in 2015 out of 4435). Median duration of EVE use was 6.0 months (range 0-65) as first line treatment and 3.9 months (range 0-65) in pretreated patients. Patient population and causes of EVE cessation will be detailed at the meeting.

266P

Year20082009201020112012201320142015
N471311133391493506
%0.200.220.300.212.36.68.611.41

Conclusions

In this very large French national and representative cohort of HR+ HER2- mBC, EVE use rose quickly as soon as marketed. EVE was mostly used in pretreated mBC albeit in probably too advanced pts. These data underline the need for physician and patient education for oral therapies.

Clinical trial identification

not applicable

Legal entity responsible for the study

UNICANCER R&D

Funding

UNICANCER

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

267P - Can contemporary trials in HER2-negative metastatic breast cancer (mBC) detect overall survival (OS) benefit? (ID 1547)

Presentation Number
267P
Lecture Time
13:15 - 13:15
Speakers
  • S. Kümmel
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Although several recent trials have demonstrated improved progression-free survival (PFS) or time to progression (TTP) with first-line regimens for HER2-negative mBC, OS benefit is often elusive. We calculated required sample sizes to power for OS based on published data in recent trials.

Methods

Randomised superiority trials of first-line chemotherapy or targeted therapy for HER2-negative mBC including >150 patients, meeting the primary efficacy objective and published between 2000 and 2014 were identified. The sample sizes required to power for PFS (or TTP) and OS were calculated retrospectively for each trial using the observed results and study/recruitment follow-up durations (alpha=0.05, 2-sided log-rank test, 80% power), and summarised as a factor relative to the actual sample size (x < 1 required x-fold fewer cases to show the same gain; x > 1 required x-fold more cases).

Results

Only 8 of the 14 identified trials reported all information required for retrospective sample size calculation (Table). Most would have required a far larger sample size to demonstrate an OS gain (x: 0.5–2479) with the observed results. In 10 of 13 trials, the sample size required to power for OS was at least 5-fold larger than that needed to power for PFS.

267P Summary of results

StudyTotal No. of patients in trial (randomisation if not 1:1)Observed median, months (arm A v B)
Retrospectively calculated sample size
Factor (x)
PFSTTPOSPFS/TTPOSOS sample size/NOS/PFS sample size
Ackland 2001460a6.3 v 8.7b18.2 v 20.1360590612.816.4
Jassem 2001267a6.2 v 8.3b18.3 v 23.3b59217926.73.0
Ejlertsen 2004387a8.2 v 10.1b18.0 v 19.178811 98831.015.2
Bontenbal 20052166.6 v 8.0b16.2 v 22.6b10224762.20.5
Feher 2005397a3.4 v 6.1b11.8 v 19.1b982120.52.2
von Minckwitz 2005364a6.7 v 8.2b820
Paridaens 2005331a3.9 v 7.5b15.6 v 18.38018825.723.5
Albain 20085294.0 v 6.1b15.8 v 18.6b16814042.78.4
Gray 20097225.8 v 11.3b24.8 v 26.572826211.4114.8
Sparano 20097517 v 9.8b20.6 v 20.52941 862 0102479.46333.4
Miles 2010488 (1:1:1)8.2 v 10.1b31.9 v 30.280220 80242.625.9
Robert 2011615 (1:2)5.7 v 8.6b22.8 v 25.723438406.216.4
Gligorov 20141854.3 v 11.9b23.7 v 39.0b342421.37.1
Lorusso 20142337.8 v 9.4b28.0 v 30.1972965441.49.9

Duration of accrual and/or follow-up not reported; accrual period assumed to be 1/3 of study duration.

Statistically significant.

Conclusions

Designing trials to test potential new treatments for HER2-negative mBC is challenging and requires a balance of regulatory acceptability, feasibility and realistic medical assumptions to calculate sample sizes, which can be particularly difficult in heterogeneous study populations with long post-progression survival and heterogeneous subsequent therapies. However, ongoing and future trials of cancer immunotherapy (new mode of action) focusing on triple-negative mBC (a more homogeneous population with shorter OS and post-progression survival, and fewer treatment options) may show a new pattern.

Clinical trial identification

Not applicable

Legal entity responsible for the study

Roche Pharma AG

Funding

Roche Pharma AG

Disclosure

S. Kümmel, C. Jackisch: Membership on advisory board: Roche Pharma AG. V. Müller: Membership on advisory board or board of directors: Amgen, AstraZeneca, Celgene, DaiichiSankyo, Eisai, Pfizer, Pierre-Fabre, Novartis, Roche, Teva, Janssen-Cilag, Genomic Health, Roche, Pierre Fabre, Amgen, Daiichi-Sankyo and Eisai. S. Klawitter: Roche Pharma AG (Employment). M.P. Lux: Membership on advisory board: Roche, Novartis, Pfizer, MSD, AstraZeneca; Corporate-sponsored research: Roche, Novartis, MSD, Celgene, Amgen. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

268P - What are the treatment patterns and overall survival (OS) in patients with metastatic triple-negative breast cancer (mTNBC) in US clinical practice? (ID 1145)

Presentation Number
268P
Lecture Time
13:15 - 13:15
Speakers
  • P. Bajaj
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

mTNBC is a condition with significant unmet medical need due to limited treatment (tx) options and short survival after chemotherapy (chemo) failure. Tx patterns and OS in patients (pts) with mTNBC in US clinical practice were assessed.

Methods

This retrospective chart review study used data collected in October and November 2016 from US oncologists via an online case report form. Data of adult pts who were initiated on a pharmacological tx after a diagnosis of recurrent or de novo mTNBC between January 2012 and June 2015 were reviewed. Tx regimens used in 1st- and 2nd-lines for mTNBC were summarized. Median OS from 1st- and 2nd-line tx initiation were estimated using Kaplan-Meier analyses.

Results

125 oncologists provided data on 411 mTNBC pts; mean age was 57 years; 298 (73%) had ≥2 lines of tx; 256 different tx sequences were identified. Mean duration of tx was 7.5 months (mos) in 1st-line and 7.3 mos in 2nd-line. The most prevalent tx regimens were single agent chemo with taxane (22%) in 1st-line and capecitabine (Cap; 20%) in 2nd-line. Median OS was 16.7 mos in 1st-line and 14.2 mos in 2nd-line.

268P

1st-line, N = 411%Median OS [95% CI], mos
All-16.7 [15.2; 18.0]
Single agent4515.6 [13.2; 18.6]
Combination5517.0 [15.1; 19.1]
Tx regimen
Single agent taxane (docetaxel/paclitaxel (pac)/nab-pac)22
Anthracycline (ATC; doxorubicin [Dox]/epirubicin/liposomal Dox) + cyclophosphamide +/- taxane14
Platinum (carboplatin/cisplatin/oxaliplatin) + taxane13
Bevacizumab-containing10
Gemcitabine (Gem) + platinum10
Cap9
Other21
2nd-line, N = 298
All-14.2 [10.5; 22.3]
Single agent7212.9 [10.5; 22.3]
Combination2816.2 [8.1; -]
Tx regimen
Cap20
Platinum-containing (w/o taxane)13
Single agent taxane13
Taxane-containing combination12
Eribulin9
ATC-containing (w/o taxane or platinum)8
Gem7
Other16

Conclusions

Findings suggest that, in US clinical practice, there is substantial heterogeneity in the tx of pts with mTNBC. Pts had a median OS < 18 mos after tx initiation. A better understanding of the mTNBC pt subpopulation and optimal tx sequencing is warranted to improve tx strategies and prolong survival.

Legal entity responsible for the study

Genentech, Inc.

Funding

Genentech, Inc.

Disclosure

P. Bajaj, C. Reyes, A. Stein, P. Cortazar: Employee of Genentech, Inc. and owner of Roche stock. D. Latremouille-Viau, A. Guerin: Employee of Analysis Group, Inc., which has received consulting fees and research funding from Genentech, Inc. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

269P - Outcomes of systemic therapy for advanced triple-negative breast cancer: A single centre experience (ID 1420)

Presentation Number
269P
Lecture Time
13:15 - 13:15
Speakers
  • N. Battisti
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Systemic treatment outcomes for advanced triple-negative breast cancer (aTNBC) are worse compared to other disease subtypes, due to aggressive behaviour, heterogeneity and lack of molecular targets. Many options are under investigation although most patients receive standard cytotoxic chemotherapy. We aimed to provide better insight into the efficacy of different lines of therapy for aTNBC (overall response rate [ORR], median progression-free survival [mPFS] and median overall survival [mOS]) to better inform discussion with patients, decision-making and referral for clinical trials.

Methods

We retrospectively identified 268 patients diagnosed with aTNBC from 01/12/2011 to 30/11/2016 from our electronic records. Patients’ and tumour characteristics were recorded, along with systemic treatment outcomes. Chi-squared/Fishers exact test and Kaplan-Meier statistical methods were utilised.

Results

186 patients treated with ≥1 line of palliative systemic treatment were eligible for the analysis with a median age at 55 (range 26-91). 53.8% had ECOG Performance Status 0 and 69.9% visceral involvement. 38.6% had a disease-free interval (DFI) <12 months following surgery and 13.4% had de novo advanced disease. 11.4% carried a BRCA mutation. 64.5% received 2 lines of therapy, 37.6% had 3 and 21.5% had 4. ORR and mPFS to first line therapy were respectively 43.9% (95% CI 36.5-51.5) and 3.7 months (95% CI 2.9-5.1), to second line was 40.2% (95% CI 31.2-49.6) and 3.5 months (95% CI 2.6-4.1), to third line 28.8% (95% CI 18.3-41.3) and 2.5 months (95% CI 2.0-3.0) and to fourth line 25.0% (95% CI 12.7-41.2) and 2.1 months (95% CI 1.6-2.8). First line patients with a DFI ≥12 months had ORR of 47.7% (95% CI 36.8-58.7) compared to 35.8% (95% CI 23.1-50.2) for those with a DFI <12 months (p 0.172). mPFS was respectively 5.2 months (95% CI 3.4-6.5) compared to 2.7 (95% CI 1.8-3.6) (p 0.005).

269P Demographics at advanced stage disease diagnosis

Median age55 (range: 26-91)
Age group N (%)< 60 years117 (62.9)
≥ 60 years69 (37.1)
ECOG Performance Status N (%)0100 (53.8)
175 (40.3)
211 (5.9)
Disease sites N (%)Visceral1130 (69.9)
Non-visceral only56 (30.1)
Advanced disease histology N (%)Invasive ductal112 (60.2)
Invasive lobular7 (3.8)
Mixed2 (1.1)
Metaplastic3 (1.6)
Other2 (1.1)
Not available260 (32.3)
De novo advanced disease N (%)25 (13.4%)
No. of lines of systemic therapy received N (%)1186 (100)
2120 (64.5)
370 (37.6)
440 (21.5)
520 (10.7)
65 (2.7)
71 (0.5)
DFI3≤12 months56 (38.6)
>12 months89 (61.4)
BRCAWild-type66 (35.7)
Mutated421 (11.4)
Unknown98 (53.0)

: with or without non-visceral involvement; 2: radiological diagnosis of advanced disease; 3: 145 (80%) patients included in the DFI analysis; 4: either BRCA1 or BRCA2.

Conclusions

The response rates observed in this population of patients are similar to those observed in published clinical trials. However, the PFS rates are short, and as a result early consideration for inclusion in clinical trials of novel approaches can be justified in these patients.

Legal entity responsible for the study

Nicolò Matteo, Luca Battisti

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

270P - Untreated hormone receptor positive/HER2-negative metastatic breast cancer survival with front-line chemotherapy and maintenance endocrine therapy (ID 2286)

Presentation Number
270P
Lecture Time
13:15 - 13:15
Speakers
  • R. Sabatier
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Except some life threatening cases, combination of endocrine therapy and CDK4/6 inhibitors is becoming the standard first line treatment for women with hormone receptor (HR) positive/HER2 negative advanced and metastatic breast cancer (MBC). However cost-effectiveness analyzes are lacking concerning this therapy. As chemotherapy also targets cell cycle we wondered how sequential combination of chemotherapy and maintenance endocrine therapy could be effective as first line treatment for naive HR+/Her2- MBC.

Methods

We retrospectively collected from our institutional database (“Institut Paoli-Calmette”, Marseille, France) cases with naive HR+/HER2- MBC. We selected patients treated with chemotherapy plus maintenance endocrine therapy as first line treatment between January 2000 and December 2015. Progression-free survival (PFS) and Overall Survival (OS) were analyzed using the Kaplan-Meier's method. We also conducted univariate (UV) and multivariate analyzes including menopausal status, visceral disease, pathological subtype, and progesterone receptor expression assessed by immunohistochemistry.

Results

A total of 183 female patients were included with a median age at diagnostic of 56,9 years. Most of them were postmenopausal (n = 114, 65.9%) and 108 (59.7%) had visceral metastases. Anthracyclynes-Taxanes combinations were used for 162 patients (88.5%). Median number of chemotherapy cycles was 6. Endocrine therapy was aromatase inhibitors and tamoxifen for 120 (67.8%) and 56 (31.6%) cases, respectively. Median PFS was 33 months [95CI = 25-38] and median OS was 79 months [95CI = 63-101]. In UV analysis pre-menopausal status (HR = 0.58), non-ductal non-lobular subtype (HR = 0.47), and absence of visceral disease (HR = 0.51) were correlated to better OS. All these features remained significant in multivariate analysis. We observed no death related to treatment.

Conclusions

Following these results, and with the issues of cost-effectiveness related to newly approved therapies, first-line chemotherapy plus maintenance endocrine therapy may be considered for untreated HR+/HER2- MBC.

Legal entity responsible for the study

Institut Paoli-Calmettes

Funding

None

Disclosure

R. Sabatier: Travel grants: Pfizer. Consultant: Novartis, Pfizer. Investigator in clinical trials promoted by Novartis and Lilly. F. Bertucci: Investigator in clinical trials promoted by Novartis and Lilly. A. Gonçalves: Consultant: Novartis. Investigator in clinical trials promoted by Novartis and Lilly. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

271P - Can we predict subsequent brain metastasis in patients with metastatic breast cancer? (ID 2353)

Presentation Number
271P
Lecture Time
13:15 - 13:15
Speakers
  • S. El Zawawy
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The one year over all survival of breast cancer patients with brain metastasis is only 20%–40%. Approximately, 80% of brain metastases occur after the diagnosis of other systemic metastatic lesions. Due to this dismal prognosis, prophylactic approaches as cranial irradiation, high-dose methotrexate, or lapatinib could be evaluated as preventative measures. However, these approaches are usually toxic and cannot be applied to all patients. This study is carried out to evaluate risk factors that have an impact on subsequent development of brain metastasis in metastatic breast cancer patients and thus, those patients can be candidates for prophylactic measures.

Methods

The medical files of 267 metastatic patients were retrospectively reviewed for demographic, clinico pathological, metastatic and treatment characteristics.

Results

46 out of 267 patients developed brain metastasis with an incidence of 17.2%. Significant risks include age <40y 28.7% patients compared to21.6% for age 40 – 50y and 11.1% for age >50y (P = 0.031), 24.2% premenopausal patients compared to 11.4% for postmenopausal (P = 0.013), Her2/neu overexpression (48.5%) and triple negative (35.3%) compared to 11.3% patients with ER positive (P = 0.0001, 0.003), high grade compared to low grade tumors (35.6% vs 12.6% P = 0.005). Patients with N2, 3 had higher risk than N0, 1 (44% vs 13.8%) (P = 0.01), 30.9% patients with disease free duration (DFD) <2 years developed brain metastasis compared to 22.1% for M1 patients and 11.1% patients with DFD >2years (P = 0.019, 0.033). 3.6% patients with bone only metastasis developed brain metastasis compared to 20.6% patients with visceral only metastasis and 27.4% patients with bone and visceral metastasis (P = 0.036, 0.014). 32.3% patients with lung containing metastasis developed brain metastasis compared to 57.3% patients with lung and liver containing metastasis and 9.2% of patients with liver containing metastasis (P = 0.038, 0.022).

Conclusions

Young patients with lung metastases, Her2/neu overexpression or triple negative with disease free duration < 2years carried the highest risk for brain metastases. Such patients may be candidate for screening or prophylactic strategies.

Legal entity responsible for the study

Ethics committee, Faculty of Medicine, Alexandria University.

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

272P - Pathological characteristics and prognosis of a cohort of 57 patients (pts) with de novo oligometastatic breast cancer (OMBC) (ID 4411)

Presentation Number
272P
Lecture Time
13:15 - 13:15
Speakers
  • S. Chretien
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Management of de novo OMBC is controversial with no specific treatment guidelines available.

Methods

We reviewed 57 consecutive de novo OMBC pts defined as ≤ 5 synchronous metastatic lesions, diagnosed between 01/2000 and 12/2012 at Oscar Lambret Center. Clinicopathological (CP) characteristics and survival data were retrospectively collected. Predictors of overall survival (OS) and progression free survival (PFS) were identified by univariate analysis before being introduced into a stepwise cox regression model.

Results

Median primary tumor size was 40mm (0-150) and 79% of the cases were invasive ductal carcinoma. Estrogen and progesterone receptors were detected in respectively 76% and 56% of the cases. HER2 overexpression was observed in 26% and 12.5% were triple negative. Median KI67 was 40% (5-80). On SBR grading, 3 (6%) cases were grade I, 31 (66%) grade II and 12 (26%) grade III. 16% pts had ≥ 4 histologically involved axillary lymph nodes. The median number of metastatic lesions was 2 (1-5). 31 pts had bone metastases. 65% pts were treated surgically for the primary tumor, 42% had neoadjuvant (NA)/adjuvant chemotherapy (CT), 45% had radiotherapy (RT) for primary BC and 58% were locally treated for their metastatic lesions, while 17 pts were treated with palliative CT only. The median follow-up period was 6.4 years (0.2–12). 2 and 5-year OS and PFS were respectively 90.8%/52.1% and 45.7%/21.4%. Significant predictors of both OS/PFS in univariate analysis were SBR grade, surgery and RT of primary BC, use of NACT and hormonotherapy. In multivariate analysis (MV) both SBR ≤ 2 at diagnosis and the administration of NA CT were identified as significant predictors for better OS (p = 0.005 and 0.006 respectively). High SBR grade independently predicted for worse PFS (p = 0.006).

Conclusions

This retrospective work further highlights the CP characteristics and predictors of outcome of de novo OMBC pts. Only SBR grade at diagnosis and use of NA CT were identified as significant predictors in MV analysis. A molecular analysis of this cohort is planned to complete these findings and characterize the subset of OMBC pts who could benefit from curative therapy and those for whom alternative strategies should be adopted.

Legal entity responsible for the study

Centre Oscar Lambret

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

273P - External validation of modified breast graded prognostic assessment for breast cancer patients with brain metastases (ID 2108)

Presentation Number
273P
Lecture Time
13:15 - 13:15
Speakers
  • G. Griguolo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Several prognostic indexes have been developed to estimate survival of breast cancer (BC) patients with brain metastases (BM). The Modified Breast Graded Prognostic Assessment (GPA) has been proposed as refinement of the Breast GPA, based on a single-institution cohort of 1,552 patients. In addition to age, tumor subtype and Karnofsky PS, the modified breast GPA comprises number of BM. The aim of this study is to validate the modified breast-GPA in an external multinational cohort.

Methods

Clinical and biological data of 668 BC patients diagnosed with BM at four European institutions between 1996 and 2016 were reviewed. Patients were classified according to breast GPA and modified breast GPA. OS was calculated from time of BM diagnosis to death or last follow up. Cox proportional models were used to calculate Hazard Ratio and 95% Confidence Intervals (CI). Performances of breast-GPA and modified breast-GPA were compared using Harrell's concordance index.

Results

At last follow-up, 632 patients (94.6%) had died. Median OS was 8.1 months (95% CI 6.9-9.4 months). Median age at BM diagnosis was 56 years (range 24-85). Tumor phenotype distribution was: triple negative (20.1%), hormone receptor (HR)-HER2 + (21.6%), HR+HER2 + (20.4%) and HR+HER2- (33.4%). KPS distribution was: 90-100 (19.6%), 70-80 (49.0%), 60 (12.8%) and ≤50 (18.6%). 355 patients (53.5%) had >3 BM. Number of BM (1,2,3,>3) was significantly associated with OS (p < 0.001). Both breast-GPA and modified breast-GPA predicted OS (Table). Concordance indices were 0.641 (95% CI, 0.6405 to 0.6422) and 0.667 (95% CI, 0.6662 to 0.6678) for breast-GPA and modified breast-GPA, respectively (p < 0.001).

273P

Breast GPA categoryNumber of patients (%)Median OS, months (95% CI)Hazard Ratio (95% CI)p
3.5-486 (13.5%)18.8 (14.5-22.6)ref<0.001
2.5-3248 (38.8%)10.3 (8.8-11.8)1.45 (1.12-1.88)
1.5-2194 (30.4%)6.2 (4.9-7.6)2.04 (1.56-2.66)
0-1.0111 (17.4%)2.5 (1.8-3.2)4.97 (3.67-6.71)
Modified breast GPA categoryNumber of patients (%)Median OS, months (95% CI)Hazard Ratio (95% CI)p
3.5-437 (5.8%)18.9 (17.2-20.5)ref<0.001
2.5-3209 (32.8%)15.2 (12.1-18.3)1.43 (0.98-2.09)
1.5-2257 (40.3%)7.9 (5.9-9.9)2.30 (1.59-3.33)
0-1.0135 (21.2%)2.3 (1.9-2.8)7.03 (4.72-10.46)

Conclusions

Number of BM is a significant prognostic factor in BC patients with BM and modified breast-GPA performs better than breast-GPA in predicting prognosis of these patients.

Legal entity responsible for the study

Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

274P - Baseline characteristics and first-line (1L) treatment of patients with HER2+ metastatic breast cancer (MBC) from the SystHERs registry (ID 3973)

Presentation Number
274P
Lecture Time
13:15 - 13:15
Speakers
  • P. Kaufman
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The addition of pertuzumab (P) to trastuzumab (H) and docetaxel improves survival in clinical trials of patients with HER2+ MBC, and is a guideline-recommended standard of care for this population. In the real world, however, various factors may influence treatment decisions. Systematic Therapies for HER2-positive Metastatic Breast Cancer Study (SystHERs) is a fully enrolled (June 2012–June 2016), ongoing, US-based, observational study that captures real-world data for patients with HER2+ MBC. Here, we describe the baseline characteristics and treatment patterns of patients who received 1L PH or H without P.

Methods

Eligible patients had HER2+ MBC diagnosed within 6 months of enrollment and were ≥18 years of age. Patients were compared descriptively by 1L treatment (PH vs H without P), defined as any therapy received up to first progression.

Results

As of February 10, 2017, among 978 eligible patients, 949 had received 1L treatment (PH, n = 711; H without P, n = 174; no H, n = 64) (Table). 476 (67%) and 88 (51%) of patients in the 1L PH and H without P cohorts, respectively, remain on study. Median follow-up from 1L treatment start was 22 and 25 months, respectively. In patients treated with PH, median duration of treatment with H and P were 15 and 13 months, respectively. In the H without P cohort, median duration of H was 15 months. Among all patients, 68% (648/949) received 1L PH + taxane.

274P

PH (n = 711)H without P (n = 174)
Baseline characteristics
Median age at MBC diagnosis, years (range)55 (21–89)61 (28–90)
White, n (%)565 (79)137 (79)
Eastern Cooperative Oncology Group performance score 0–1, n (%)613 (86)146 (84)
Private insurance, n (%)367 (52)83 (48)
Urban or suburban living location, n (%)556 (78)138 (79)
De novo, n (%)379 (53)88 (51)
Estrogen receptor positive and/or progesterone receptor positive, n (%)496 (70)126 (72)
Visceral, n (%)476 (67)108 (62)
≥3 metastatic sites, n (%)232 (33)43 (25)
Liver metastasis, n (%)300 (42)47 (27)
Central nervous system (CNS) metastasis, n (%)45 (6)a15 (9)a
Prior cardiovascular disease, n (%)80 (11)31 (18)
Treatment for early breast cancerb, n (%) Any neoadjuvant or adjuvant therapy Any Hn = 332 293 (88) 187 (56)n = 86 72 (84) 46 (53)
1L treatment patterns, n (%) (Treatments are not mutually exclusive)
With chemotherapy Taxane Docetaxel Paclitaxel Platinum673 (95) 648 (91) 479 (67) 195 (27) 69 (10)117 (67) 87 (50) 34 (20) 51 (29) 40 (23)
With hormonal therapy Aromatase inhibitor Tamoxifen282 (40) 221 (31) 61 (9)94 (54) 76 (44) 14 (8)

 In patients who did not receive H, 25% (16/64) had CNS metastasis.

 In patients with recurrent disease.

Conclusions

Of patients with HER2+ MBC in the real-world SystHERs study, 68% were treated with PH + taxane. Compared with patients who received PH, those who received H without P were older, less commonly had liver metastasis, and more commonly had prior cardiovascular disease, suggesting that these characteristics may have influenced the treatment choice between PH vs H without P.

Clinical trial identification

NCT01615068

Legal entity responsible for the study

Genentech/Roche

Funding

Genentech/Roche

Disclosure

P.A. Kaufman: Consulting fees from Genentech. Contracted research paid to institution from Genentech/Roche. S.A. Hurvitz: Research funding from Amgen, Bayer, Boehringer Ingelheim, Genentech, GSK, Pfizer, Roche, Biomarin, Merrimack, PUMA, Dignitana, Medication. Research and travel from Lilly, Novartis, OBI Pharma. J. O\'Shaughnessy: Consulting fees from Genentech. G. Mason: Advisory Board: Avon Foundation for Women Scientific. D.A. Yardley: Fees for services from Novartis and Genentech. Contracted research paid to institution from Genentech and Novartis. A. Brufsky: Consulting fees from Roche. H.S. Rugo: Fees for services from Genomic Health. Ccontracted research paid to institution from Genentech, Pfizer, Novartis, Lilly, OBI Pharma, GTX, Macrogenics, Merck. M. Cobleigh: Royalties and Receipt of IP rights from Genomic Health. Consulting fees from Genentech/Roche and GSK. Contracted research from Genentech/Roche, Merrimack, Macrogenics, NRG foundation. S.M. Swain: Consulting fees from Genentech/Roche, Novartis, and Pieris. Contracted research paid to institution from Genentech/Roche, Pfizer, Merrimack, and Lilly. D. Tripathy: Consulting fees from Puma Biotech, NEKTAR Pharma, Novartis. Contracted research paid to institution from Novartis. L. Chu, V. Antao, B. Yoo: Employment at Genentech. Stock ownership in Roche. M. Jahanzeb: Consulting fees from Roche. Data and Safety Monitoring Board for Puma.

Collapse
Poster display session Poster Display session

275P - First-line treatment patterns by age for patients (pts) with HER2+ metastatic breast cancer (MBC) in the SystHERs registry (ID 3817)

Presentation Number
275P
Lecture Time
13:15 - 13:15
Speakers
  • M. Jahanzeb
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Clinical outcomes for pts with HER2+ MBC may differ by age, potentially influenced by differences in treatment regimens administered to older vs younger pts. Systematic Therapies for HER2+ Metastatic Breast Cancer Study (SystHERs) captures real-world treatment patterns and outcomes in pts with HER2+ MBC. Here, we examine baseline characteristics, first-line treatments, and breast cancer–specific survival (BCSS) by age.

Methods

SystHERs is a fully enrolled (Jun 2012–Jun 2016), ongoing, US-based, observational study. Pts were ≥18 years old and had HER2+ MBC diagnosed within 6 months of enrollment. Pts were grouped by age at MBC diagnosis (<50, 50–69, or ≥ 70 years) and compared descriptively. BCSS was defined as time from the date of MBC diagnosis to date of death due to MBC progression.

Results

As of Feb 10, 2017, of 978 eligible pts, 287 were <50 years old, 563 were 50–69, and 128 were ≥70 at MBC diagnosis. Median follow-up from MBC diagnosis was 23, 22, and 19 months, respectively. Baseline characteristics, first-line treatments, and BCSS are shown (Table). In pts who received chemotherapy, docetaxel was the most common agent in pts <50 (67%) and 50–69 (64%) followed by paclitaxel (29% in both groups), whereas in pts ≥70, 43% and 45% received docetaxel and paclitaxel, respectively.

275P

<50 years (n = 287)50-69 years (n = 563)≥70 years (n = 128)
Baseline characteristics
Median age at MBC diagnosis, years (range)43 (21–49)59 (50–69)75 (70–90)
Eastern Cooperative Oncology Group performance score 0–1, n (%)249 (87)476 (85)101 (79)
De novo, n (%)158 (55)278 (49)52 (41)
Median time from early breast cancer diagnosis to MBC diagnosisa, months (range)36 (9–166)50 (5–329)42 (4–452)
Estrogen receptor positive and/or progesterone receptor positive, n (%)203 (71)387 (69)96 (75)
Visceral disease, n (%)196 (68)378 (67)75 (59)
≥3 metastatic sites, n (%)87 (30)183 (33)32 (25)
Bone metastasis, n (%)165 (57)282 (50)57 (45)
Liver metastasis, n (%)117 (41)216 (38)33 (26)
First-line treatments, n (%) (Treatments are not mutually exclusive)
Any HER2-targeted therapy PH H without P275 (96) 229 (80) 37 (13)531 (94) 414 (74) 93 (17)116 (91) 68 (53) 44 (34)
Any chemotherapy PH + taxane H without P + taxane254 (89) 207 (72) 18 (6)489 (87) 386 (69) 53 (9)86 (67) 55 (43) 16 (13)
Any hormonal therapy With PH With H without P113 (39) 91 (32) 18 (6)230 (41) 161 (29) 48 (9)67 (52) 30 (23) 28 (22)
Breast cancer -specific survival, % (95% confidence interval)
Breast cancer–specific survival rate 12 months 24 months 36 months95 (92–97) 86 (81–90) 78 (71–84)92 (89–94) 80 (76–84) 68 (63–73)92 (85–95) 80 (71–87) 64 (48–76)

In pts with recurrent MBC only. H, trastuzumab; P, pertuzumab.

Conclusions

In this preliminary real-world analysis of pts with HER2+ MBC, pertuzumab (P) + trastuzumab (H) was more commonly used than H without P across all age groups. Pts <50 and 50–69 years old more commonly received PH + taxane than those ≥70 (72% and 69% vs 43%, respectively). Compared with younger pts, those ≥70 received regimens with chemotherapy less commonly (89% and 87% vs 67%), and more commonly received regimens with H without P (13% and 17% vs 34%) or hormonal therapy (39% and 41% vs 52%). Pts <50 had longer BCSS than those ≥70 (78% vs 64% at 3 years).

Clinical trial identification

NCT01615068

Legal entity responsible for the study

Genentech/Roche

Funding

Genentech/Roche

Disclosure

M. Jahanzeb: Consulting fees from Roche. Data and Safety Monitoring Board for Puma. D. Tripathy: Consulting fees from Puma Biotech, NEKTAR Pharma, Novartis. Contracted research paid to institution from Novartis. S.A. Hurvitz: Research funding from Amgen, Bayer, Boehringer Ingleheim, Genentech, GSK, Pfizer, Roche, Biomarin, Merrimack, PUMA, Dignitana, Medication. Research and travel from Lilly, Novartis, OBI Pharma. J. O\'Shaughnessy: Consulting fees from Genentech. G. Mason: Advisory Board: Avon Foundation for Women Scientific. D.A. Yardley: Fees for services from Novartis and Genentech. Contracted research paid to institution from Genentech and Novartis. A. Brufsky: Consulting fees from Roche. H.S. Rugo: Fees for services from Genomic Health. contracted research paid to institution from Genentech, Pfizer, Novartis, Lilly, OBI Pharma, GTX, Macrogenics, Merck. M. Cobleigh: Royalties and Receipt of IP rights from Genomic Health. Consulting fees from Genentech/Roche and GSK. Contracted research from Genentech/Roche, Merrimack, Macrogenics, NRG foundation. S.M. Swain: Consulting fees from Genentech/Roche, Novartis, and Pieris. Contracted research paid to institution from Genentech/Roche, Pfizer, Merrimack, and Lilly. L. Chu, V. Antao, B. Yoo: Employment at Genentech. Stock ownership in Roche. P.A. Kaufman: Consulting fees from Genentech. Contracted research paid to institution from Genentech/Roche.

Collapse
Poster display session Poster Display session

276P - Defining priorities for research: Interim results of the Canadian metastatic breast cancer priority setting partnership (ID 4089)

Presentation Number
276P
Lecture Time
13:15 - 13:15
Speakers
  • N. Nixon
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Research priorities are generally determined by funders and researchers without direct involvement and input from patients and caregivers. Certain disease areas have incorporated the patient voice to determine patient driven priorities. In this study, this approach was employed to better understand the needs and priorities of metastatic breast cancer patients and their caregivers.

Methods

This study was conducted using methodology outlined by the James Lind Alliance. A steering committee of patients, physicians, patient advocates, and allied health care professionals was assembled to oversee the research study. The initial survey collected unanswered research questions from patients, caregivers, and clinicians. Responses were collected and categorized by consensus of the steering committee. Here we present the results from the national survey.

Results

Between November 2016 and April 2017, 733 responses from 311 individuals were collected (62% patients, 11% physicians, 9% caregivers or relatives, 5% nurses/allied health professionals, 2% patient organization representatives, and 10% other). The main themes for key patient priorities are: 136 (19%) related to treatment and monitoring, 78 (11%) linked lifestyle and alternative therapy, 58 (8%) regarded tumour biology, 53 (7%) regarded psychosocial aspects, 46 (6%) to diagnosis, 35 (5%) to toxicity, 24 (3%) to prevention, and 17 (2%) to young or pre-menopausal population. Two hundred and eighty-six (39%) were considered out of scope. The most frequently identified priorities included the role of alternative therapies for improving survival, the role of immune therapy for treating metastatic breast cancer, and the potential for improving outcomes with early detection/surveillance with modern treatment and diagnostic modalities.

Conclusions

Patient derived research priorities in advanced breast cancer point to an improved understanding of alternative therapies, integration of immune therapy and a focus on early detection of relapse. These priorities should be addressed by the research community to meet the needs of our patients with advanced breast cancer.

Legal entity responsible for the study

Nancy Nixon

Funding

None

Disclosure

S. Verma: Advisory Boards for: Roche, Pfizer, Novartis, Eli Lilly, Merck, Amgen. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

277P - Real-world treatment patterns and outcomes among elderly metastatic triple negative breast cancer patients in the United States (ID 4168)

Presentation Number
277P
Lecture Time
13:15 - 13:15
Speakers
  • S. Satram-Hoang
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Triple negative breast cancer (TNBC) is more aggressive than other breast cancer (BC) subtypes and accounts for up to 20% of all BC. Despite the poorer prognosis, there are no approved targeted treatments available and chemotherapy (chemo) remains the only choice. We examined treatment patterns and outcomes among elderly metastatic TNBC patients (pts) in routine clinical practice.

Methods

Patients were identified from the linked SEER-Medicare database (1/1/2001-12/31/2013). The analysis included TNBC de novo Stage IV pts (n = 776) and pts with distant recurrences following an initial diagnosis of Stage I-III (n = 1851). Pts were ≥66 years and continuously enrolled in Medicare Parts A/B in the year prior to diagnosis. The analysis was stratified by age <70 (n = 359) and ≥70 (n = 2268). Kaplan Meier analyses and time-varying Cox proportional hazards regression assessed overall survival (OS).

Results

The mean age at metastatic diagnosis was 77.6 years and 1259 (48%) pts received treatment with chemo. Compared to < 70 year olds, ≥70 year olds were less likely to be diagnosed with de novo Stage IV disease (28% vs. 42%), had worse performance status indicators, higher comorbidity burden, were less likely to receive chemo (45% vs. 66%), but were more likely to have had BC surgery (81% vs. 70%). Pts treated with chemo had improved OS compared to untreated pts, and the survival advantage was more pronounced in the <70 year olds with a 6 month longer unadjusted OS compared to the ≥70 cohort (log rank p < 0.0001). This finding was supported in the adjusted multivariate model which showed a 46% (HR = 1.46; 95% CI: 1.08-1.96) increased risk of death for untreated pts in the <70 year olds and a 17% (HR = 1.17; 95% CI: 1.06-1.29) increased risk of death for untreated pts in the ≥70 year olds (vs. treated).

Conclusions

In this real-world analysis, 52% of elderly TNBC pts did not receive chemo following their metastatic diagnosis. Although the survival benefits of chemo were stronger in the younger cohort, the benefits of treatment were maintained among ≥70 year olds who were also less likely to receive chemo. These findings suggest opportunities exist to improve the clinical treatment of elderly TNBC pts.

Legal entity responsible for the study

Q.D. Research, Inc

Funding

Genentech, Inc.

Disclosure

S. Satram-Hoang, K.Q. Hoang, F. Momin: Research funding to Institution from Genentech. P. Bajaj, A. Stein, P. Cortazar, C. Reyes: Genenetech employment and Roche stock ownership.

Collapse
Poster display session Poster Display session

278P - Effect of neoadjuvant chemotherapy on disease free survival and over all survival in triple-negative breast cancer patients (ID 4379)

Presentation Number
278P
Lecture Time
13:15 - 13:15
Speakers
  • J. Gill
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Breast cancer (BC) is the most common cancer in India with 150000 new cases are diagnosed and 70000 women dies of it every year. Triple-negative breast cancer (TNBC) is an aggressive subtype that lack ER and PR expression and absence of overexpressed or amplified HER2. TNBC accounts for 15%-25% of all invasive BC, occurs more in younger women and is associated with higher histologic grade and advanced disease. Our goal was to study the relation between triple-negative receptor status and major determinants of clinical outcome, such as response to neoadjuvant chemotherapy (rate of pathologic complete response [pCR]), progression free survival (PFS), site-specific distribution of recurrence, postrecurrence survival (PRS) and overall survival (OS).

Methods

We included 2658 patients who received neoadjuvant chemotherapy at Jawaharlal Nehru Cancer Hospital Bhopal for stage I-III breast cancer from 1990 to 2010 and for whom complete receptor information were available. Clinical and pathologic parameters, pCR, survival measurements and organ-specific relapse rates were compared between patients with TNBC and non-TNBC.

Results

505 patients (19%) had TNBC. Mean age for TNBC (42 years) was lesser than non-TNBC (56 years; P=.002). Patients with TNBC had significantly higher pCR rates (34% v 14%; P=.028) but decreased 5 year PFS rates (P<.0001) and 5year OS rates (P <.0001). TNBC was associated with increased risk for distant metastases (P =.0005), lower risk for bone recurrence (P =  .034) and shorter PRS (P < .0001). Recurrence and death rates were higher for TNBC only in the first 5 years. If pCR was achieved, patients with TNBC and non-TNBC had similar survival (P = .36). Patients with residual disease (RD) had worse OS if they had TNBC compared with non-TNBC (P < .0001).

Conclusions

TNBC patients have increased pCR rates (excellent survival) compared with non-TNBC. However TNBC patients with RD have significantly worse survival after neoadjuvant chemotherapy in first 5 years.TNBC patients may be best treated with 3rd generation adjuvant or neoadjuvant chemotherapy regimens that achieve the highest possible pCR rates. With high risk of distant metastases, these patients require closer surveillance in initial years of follow-up.

Clinical trial identification

IND16434A

Legal entity responsible for the study

Jawaharlal Nehru Cancer Hospital, Bhopal, India

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

279P - Real-world everolimus experience in postmenopausal HR+ HER2- advanced breast cancer women: Treat ER+ight Canadian prospective observational study 2nd subgroup analysis (ID 1023)

Presentation Number
279P
Lecture Time
13:15 - 13:15
Speakers
  • C. Doyle
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Treat ER+ight is the 1st Canadian real-world study enrolling patients (pts) previously exposed to letrozole or anastrozole and currently receiving endocrine therapy (ET) alone or in combination with targeted therapy.

Methods

At data cut-off (13 Mar '17), 35 pts were enrolled in the everolimus + exemestane (EVE+EXE) subgroup out of 100 total enrolled pts since Mar '16 from 24 active sites. This sub-analysis describes baseline characteristics, treatment duration and stomatitis prevention outcomes in EVE+EXE pts.

Results

Baseline characteristics: median age – 65 (39-80); family history of breast cancer (BC) – 37%; ECOG 0-1 – 83%; median time since primary BC diagnosis – 6 yrs (2-12); median time since advanced BC diagnosis – 1 yr (0-3.5). Sites of metastases (%): bone (B) only – 26; visceral (V) only – 43; B+V – 23. Line (L) of metastatic therapy (%): 17 - 1L, 51 - 2L, 31 - 3L. EVE start dose (%): 10mg (80), 7.5mg (3), 5mg (17). Therapy ongoing n (%): 23 (66) (78% 1L & 2L). Therapy discontinued n (%): 12 (34) (50% 3L). Reason for discontinuation n (%): 9 (75) progression, 2 (17) adverse event, 1 (8) death. Median follow-up time at data cut-off 3.4 mths (0.5-9.1). Median time to treatment discontinuation (TTD) 7.0 mths (95% CI, 3.4-NR) in overall subgroup. Median TTD NR (95% CI, 2.4-NR) in 20 (57%) pts receiving prophylactic/proactive mouthwash (P/P MW) compared to 5.7 mths (95% CI, 3.2-NR) in 15 (43%) pts receiving reactive/no MW (p = 0.140, Log-rank). 1st stomatitis event related to EVE n (%): overall subgroup 10 (29) – any Grade (Gr), 7 (20) – Gr 1, 2 (6) – Gr 2, 1 (3) – Gr 3 and in P/P MW subgroup 3 (15) – any Gr. Median time to 1st stomatitis event in P/P MW subgroup (mths) NR (95% CI, 1.64-NR) and NR (95% CI, 0.33-NR) in reactive/no MW subgroup (p = 0.334, Log-rank) with majority of stomatitis events occurring early within the 1st 2 months of therapy.

Conclusions

Compared to BOLERO-2, EVE+EXE pts in Treat ER+ight had lower ECOG 0-1 (83 vs 96%), more V disease (66 vs 56%), 20% received lower EVE start dose, similar treatment duration (7.0 mths median TTD vs 7.8 mths median PFS) and lower any Gr stomatitis (29 vs 59%). This represents the 1st observation of a trend toward improved TTD in pts receiving P/P MW upon EVE initiation.

Clinical trial identification

NCT02753686

Legal entity responsible for the study

Novartis Pharmaceuticals Canada Inc.

Funding

Novartis Pharmaceuticals Canada Inc.

Disclosure

E. Chouinard: Research support received from Novartis. S.R. Perri: Employee of Novartis Pharmaceuticals Canada Inc. S. Chia: Honorarium from Novartis and Pfizer. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

280P - FICHE-YOUNG: FIrst-line treatment CHoicE in hormone receptor positive (HR+)/HER2- negative metastatic breast cancer patients (MBC) ≤45 years old. A large observational multicenter cohort survival analysis (ID 2554)

Presentation Number
280P
Lecture Time
13:15 - 13:15
Speakers
  • B. PISTILLI
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Metastatic breast cancer (MBC) in young patients (pts) is traditionally considered at poor prognosis. Although current guidelines recommend endocrine therapy (ET) as first line treatment (1st trt) for HR+ HER2- MBC, younger age can lead to more extensive use of first line chemotherapy (CT). In the present analysis, we aimed to assess overall survival (OS) of younger MBC pts compared to older ones, and to explore 1st trt choices in a large real-life multicenter cohort.

Methods

The Epidemiological Strategy and Medical Economics (ESME) Research program aims to collect high-quality real-world data in oncology from 18 French Comprehensive Cancer Centers. Pts who started treatment for a newly diagnosed MBC between Jan 2008 and Dec 2014 were selected in the MBC ESME database. The primary end point of the FICHE-Young study was to compare adjusted OS in pts diagnosed with endocrine-sensitive HR+ HER2- MBC and aged ≤45 vs > 45 at diagnosis. We also evaluated 1st trt choices in both categories and its correlation with OS. Analyses will be adjusted on a propensity score, in order to control selection biases associated with non-randomization.

Results

6265 pts out of 16703 in ESME had HR+/HER2- MBC. Characteristics and 1st trt choices are listed in the Table. Median OS was 62.3 months (mos) (95% CI 56.5-69) in pts ≤45 and 52.8 mos in those >45 (95% CI 50.7-55), p<.0001. In pts ≤45, we did not show any statistically significant difference in OS between first line ET and CT+/-ET (68.5 mos for ET (95% CI, 56.8-NE) vs 59.0 mos for CT+/-ET (95% CI, 55.9-69), p = 0.3208).

280P

≤ 45 yrs old>45 yrs old
N8515414
Median age40.0 [23;44]63 [45;95]
Visceral metastases56.3%51.6%
De novo MBC41%42.4%
Median time to onset of MBC3.28 yrs [0.50;19.53]9.18 yrs [0.50;43.02]
1st trt: ET alone19.4%47.4%
Chemo +/- maintenance ET80.6%52.6%

Conclusions

With the limitations of a nonrandomized study population, in this real-world setting, younger HR+MBC pts did not show a poorer prognosis compared to older patients. Many young pts received CT as first line treatment, with no demonstrated benefit over ET alone.

Clinical trial identification

NOT APPLICABLE

Legal entity responsible for the study

UNICANCER

Funding

UNICANCER

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

281P - Cell-free circulating DNA as independent prognostic markers in metastatic breast cancer (ID 2461)

Presentation Number
281P
Lecture Time
13:15 - 13:15
Speakers
  • J. Cheng
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Blood-based biomarkers like microRNAs, cell-free DNA and circulating tumor cells hold great promise as they are reproducible and easily accessible in cancer patients. Cell-free DNA variables, such as cell-free DNA concentrations (cfDNA conc) and cell-free DNA integrity (cfDI), have great potential as diagnostic and prognostic markers in breast cancer patients. Here we investigated the potential prognostic ability of cfDNA conc and cfDI in a prospective study cohort of metastatic breast cancer (MBC) patients.

Methods

Blood was collected for cfDNA extraction from patients when enrolled about to start the first cycle of systematic therapy at baseline (MBCBL) and after the first cycle of systematic therapy (MBC1C). cfDNA conc and cfDI in blood plasma were evaluated by measuring the short and long fragments of two repetitive DNA elements, ALU (ALU-111bp, ALU-260bp) and LINE1 (LINE1-97bp, LINE1-266bp), by quantitative PCR. In total, 268 patients were included in this study. cfDNA conc and cfDI was compared between these two groups. The prognostic ability of cfDNA variables was evaluated by univariate and multivariate Cox regression model.

Results

A significant increase of cfDI (P = 1.21E-7 for ALU and P = 1.87E-3 for LINE1) and decrease of cfDNA conc (P = 1.17E-3 for ALU and P = 1.60E-2 for LINE1) were observed between patients at MBCBL and patients at MBC1C. Multiple Cox regression model indicated that cfDI and cfDNA conc can be used as independent prognostic markers in patients after one cycle of therapy with odds ratio (OR) and 95% confidence interval (CI) of 0.70 (0.48 - 1.01) for ALU cfDI, 0.63 (0.44 - 0.92) for LINE1 cfDI, 2.44 (1.68 - 3.53) for ALU cfDNA conc, 2.12 (1.47 - 3.06) for LINE1 cfDNA conc for overall survival. When four cfDNA variables were combined, it can reach an OR of 2.53 (1.77-3.62) for overall survival analysis of patients.

Conclusions

In summary, our results showed a decreased cfDNA conc and increased cfDI from the enrollment of the study to the first cycle of systematic therapy in MBC patients. cfDNA conc and cfDI can serve as independent prognostic markers in MBC patients after the first cycle of systematic therapy.

Legal entity responsible for the study

Molecular Biology of Breast Cancer, Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany

Funding

The University Hospital of Heidelberg, Heidelberg, Germany; the German Cancer Research Center (DKFZ), Heidelberg, Germany

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

282P - Gene alteration in triple negative breast cancer patients in a phase I/II study of combination therapy with eribulin and olaparib (ID 1010)

Presentation Number
282P
Lecture Time
13:15 - 13:15
Speakers
  • A. Shimomura
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Olaparib (Lynparza®) shows efficacy in patients with triple negative breast cancer (TNBC). Eribulin is one of the standard therapies for metastatic breast cancer. A phase I/II study of combination therapy with eribulin and olaparib capsule (EO study) was conducted on patients with TNBC. We investigated the correlation between response to combination therapy and homologous recombination deficiency (HRD).

Methods

Tissue samples were collected from patients who participated in the EO study. Archival tissue samples were examined for gene alterations using the Foundation Medicine, Inc. (FMI) gene panel. Pathogenic or likely pathogenic gene alterations were extracted from all detected gene alterations using the FMI data dictionary. This dictionary uses the COSMIC database, relevant literature, and internal evidence to determine the reportable status of an alteration. HRD-related genes were defined as previously described (Konstantinopoulos PA, et al. Cancer Discovery, 2015). Correlation between presence of HRD and response to combination therapy was tested using chi-square test.

Results

A total of 32 tissue samples were collected. Nineteen samples were collected from the phase I and 13 samples from the phase II. Seventeen patients were treated at the recommended dose. Thirty-three gene mutations were detected. The most frequent gene mutations were TP53 (n = 27, 84.4%), PIK3CA (n = 7, 21.9%), BRCA1 (n = 5, 15.6%), MLL3 (n = 5, 15.6%), and AKT1 (n = 4, 12.5%). We detected 32 gene amplifications, with MYC being the most common (n = 6, 18.8%). Eight homozygous deletions were detected, and the most frequent was loss of PTEN (n = 4, 12.5%). We detected 10 gene rearrangements. HRD, including BRCA1/2 mutations, was observed in nine patients. The overall response rate (RR) and clinical benefit rate (CBR) were 31.3% and 78.1%, respectively. The RRs in patients with HRD and without HRD were 44.4% and 26.1%, respectively (p=.4072). The CBRs for the HRD and non-HRD groups were 66.7% and 82.6%, respectively (p =.3702).

Conclusions

Eighty-three gene alterations were detected in TNBC pts receiving combination olaparib/eribulin therapy. Patients with HRD had numerically higher response rate.

Clinical trial identification

UMIN000018721

Legal entity responsible for the study

Japan

Funding

Japan Agency for Medical Research and Development, AstraZeneca

Disclosure

A. Shimomura: Research fund from AstraZeneca, Inc. K. Yonemori: Lecture fee from Eisai. N. Masuda: Research Funds from Chugai, Eisai. Honorarium from Chugai, AstraZeneca. K. Aogi: Honoraria from Chugai, Eisai, Sanofi, SRL, AstraZeneca, Taiho, Novartis, Daiichi Sankyo, Mochida Pharmaceutical, Ono Pharmaceutical, Otsuka Pharmaceutical, Nihon Medi-Physics and Eli Lilly. M. Takahashi: Lecture fee from Eisai, AstraZeneca, Chugai. Y. Naito: Honoraria from Eisai, Chugai, Taiho, Novartis, Eli Lilly, Meiji Seika, Bayer Yakuhin, Roche Diagnostics. Research funds from Merck Serono, AstraZeneca, Eli Lilly, Nippon Kayaku. C. Shimizu: Research funds from Chugai, Pfizer, Eli lilly, MSD. K. Tamura: Research funds from AstraZeneca, Daiichi Sankyo, Pfizer, MSD. Y. Fujiwara: Lecture fee from AstraZeneca, Eisai, Daiichi Sankyo, Taiho, Chugai, Eli Lilly, Yakut. Research funds from Taiho, Takeda, Chugai, Eli Lilly, Nippon Kayaku. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

283P - Neutrophil-lymphocyte ratio (NLR) as a prognostic factor in metastatic breast cancer (ID 2925)

Presentation Number
283P
Lecture Time
13:15 - 13:15
Speakers
  • F. Ayala de la Peña
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Neutrophil-lymphocyte ratio (NLR) might be a surrogate marker of the tumor microenvironment immune balance and has been proposed as a prognostic factor for different tumors. A metanalysis of NLR for breast cancer (BC) showed that higher values at diagnosis were associated with lower survival. However, these results were mainly derived from the analysis of early BC cases: only three of twelve articles included women with metastatic breast cancer (MBC), sample size was small and no differential statistical analysis was performed for MBC. The aim of this work was to determine the prognostic value of NLR for MBC.

Methods

We retrospectively collected clinical and analytical data from a series of consecutive MBC patients treated in one center between 2009 and 2016. NLR (=neutrophil count/lymphocyte count) was obtained from differential white blood cell count at diagnosis of metastasis, before starting any treatment. Non-parametric tests (Mann Whitney U, Kruskal-Wallis) were used to evaluate differences of NLR between groups. Kaplan-Meier curves and Cox regression models (univariate and multivariate) were used for overall survival analysis.

Results

265 consecutive patients with MBC were included, 117 of them (44%) with metastatic disease at diagnosis. Median age: 59 (19-95); ECOG 0-1 (69%), 2-3 (12%); site: bone only (37%), visceral only (18%), bone+visceral (30%); tumor subtype: HR (hormone receptor)+/HER2- (59%), HR+/HER2 + (17%), HR-/HER2 + (7%), HR-/HER2- (14%); disease free interval in recurrent MBC: <24 months (48, 32%), > 24 months (100, 67%). Outcomes: 135 deaths; median overall survival (OS): 35 months (IC95%: 27.4-42.6). Median NLR was 2.31 (range: 0.70-44.33), with significant higher values in women with ECOG 2-3 (p = 0.008) or negative estrogen receptors (p = 0.03). Univariate Cox model of OS showed a HR = 1.07 (95%: 1.03-1.11; p < 0.001) for NLR as a continuous variable; using the median value as cut-off, HR = 1.47 (95%CI: 1.05-2.07; p = 0.024). A multivariate Cox model showed the independent value of NLR for OS (Table).

283P Multivariate Cox proportional hazard regression model of MBC overall survival including NLR at diagnosis of distant disease

HR (95%CI)p
MBC at diagnosis (M1)1.48 (1.02-2.14)0.036
Negative estrogen receptors2.56 (1.75-3.76)<0.001
LNR (continuous variable)1.07 (1.03-1.11)0.001
Age (continuous variable)1.02 (1.00-1.03)0.008

Conclusions

A higher NLR at diagnosis is a negative prognostic factor for overall survival in metastatic breast cancer. These data, if prospectively validated, may support the addition of NLR to MBC prognostic models and may lead to differential therapeutic approaches in patients with higher NLR.

Legal entity responsible for the study

Francisco Ayala de la Peña

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

284P - Real-life study of BRCA genetic screening in metastatic breast cancer (ID 4458)

Presentation Number
284P
Lecture Time
13:15 - 13:15
Speakers
  • G. Meynard
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Genomic instability is a hallmark of cancers and mutations in the DNA repair BRCA1 and BRCA2 genes predispose to breast and other cancers. Prevalence of BRCA1/2 mutations is known in general population, but the frequency of these mutations in patients with metastatic breast cancer has not been established.

Methods

Prospective BRCA1 and BRCA2 genetic testing was proposed to all patients with metastatic breast cancer treated in 7 centers (in Franche-Comte, France) between February 19th 2015 and November 30th. BRCA TrueTM test (Pathway Genomics®, San Diego CA, USA) was used to analyze the coding and flanking regions of BRCA1 and BRCA2 genes associated with hereditary breast and ovarian cancer by next-generation sequencing-base and Sanger sequencing.

Results

Of the 407 metastatic breast cancer patients, 11 (2.7%) had pathogenic germline BRCA1/2 mutations. BRCA2 (n = 8) mutations were the most frequent. Five of 11 patients (45%) would not have been candidate for BRCA1/2 mutation screening according to genetic counseling recommendations. All patients with a BRCA2 mutation presented a luminal metastatic breast cancer whereas all patients with BRCA1 mutation had a triple-negative metastatic breast cancer.

Conclusions

This is the first study assessing the prevalence of germline BRCA1 and BRCA2 mutations in an unselected population of patients with metastatic breast cancer. These patients with BRCA1/2 germline mutation represent the targeted population for poly(ADP-ribose) polymerase (PARP) inhibitors based therapy.

Legal entity responsible for the study

University Hospital Jean Minjoz of Besancon, France

Funding

BioMarin

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

285P - Higher MCTS1 mRNA level in breast cancer may associate with an unfavorable outcome (ID 1583)

Presentation Number
285P
Lecture Time
13:15 - 13:15
Speakers
  • Y. Shi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The oncogene MCTS1 was originally discovered as an amplified product in a subset of T-cell lymphoma lines. It has been involved in cell cycle progression and conferring a growth advantage in lymphomas. However, the role of MCTS1 in predicting the outcome of breast cancer patients remains unclear.

Methods

GEPIA was a newly–developed web server for gene expression profiling and interactive analyses. We analyzed the gene expression profile across the clinical and RNA-seq data of 1085 breast cancer and 291 normal breast tissue based on TCGA and GTEx data.

Results

The results showed that MCTS1 was one of the most differential survival genes, and the median expression levels of MCST1 in breast cancer and normal tissue were 51.3 and 32.7 respectively. The overall survival of breast cancer patients with high MCTS1 mRNA level was inferior to that with low MCTS1 mRNA level (Logrank p = 2.3e−06). Cox Proportional Hazards Model analysis showed that MCST1 mRNA level in tumor was an independent predictor for overall survival status in breast cancer patients (HR = 2.2, p(HR)=4.1e−06). We inquired MCST1 on UCSC Genome Browser on Human Dec. 2013 (GRCh38/hg38) Assembly, and the result showed that H3K27ac, an active enhancer mark associated with the activation of transcription, was highly modified in promoter subdomain. We further analyzed the functional protein association networks on the String database, which showed that DENR involved in the translation of target mRNAs by recognizing the initiation codon were related to breast cancer with high MCTS1 mRNA expression.

Conclusions

In conclusion, higher MCTS1 mRNA level in breast cancer may associate with an unfavorable outcome due to H3K27ac modified MCTS1 promoter hyperacylation, which promotes its expression to inhibit apoptosis and cell cycle progression. To further confirm it, the experiment about the transfection of the shRNA on target gene is undergoing.

Legal entity responsible for the study

Guangdong Academy of Medical Sciences & Guangdong General Hospital, Guangzhou, China Guangdong Academy of Medical Sciences & Guangdong General Hospital, Guangzhou, China

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

286P - Holistic therapeutic strategy of TNBC necessitates in depth molecular classification: A prospective study (ID 3638)

Presentation Number
286P
Lecture Time
13:15 - 13:15
Speakers
  • R. Bhattacharyya
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Triple negative breast cancer (TNBC), the most heterogeneous and aggressive breast cancer has always remained a global burden. To understand the molecular pathogenesis, we stratified the seven subgroups of TNBC propounded by Lehmann et al (2011) as: [1] TNBC with alterations of the DNA damage repair and cell cycle checkpoint pathways (Basal Like 1 & 2 (BL1, BL2)), [2] with upregulation of cell signalling and cell motility pathways (mesenchymal (M), mesemchymal stem like (MSL)), [3] with upregulated cell survival pathways (BL2, M, MSL) [4] With upregulated angiogenesis pathways (BL2, MSL), [5] With upregulation of pathways associated with T cell signalling, [6] With upregulated Androgen receptor signalling pathways. Our objective was to prioritize the basic molecular heterogeneity of TNBC in redefining our choice of drugs.

Methods

Lehmann’s TNBC subgroups showed deregulation of diverse molecular pathways necessitating targeted therapeutics. We conducted a Meta-Analysis on12 randomized reported trial cases (n = 1170), solely under the following classes of drug regimens: [1] DNA destabilizers, [2] PARP inhibitors, [3] Microtubule stabilizers, [4] Angiogenesis inhibitors, [5] Antimetabolite, [6] T cell targeted therapy; as single or combinational therapies. Radiotherapy recipients were excluded.

Results

Best therapeutic efficacies of DNA destabilizers with angiogenesis inhibitors in combination than monotherapy with either (OR: 5.011-7.286; p value< 0.001) indicated a significant prevalence of basal like TNBCs in populations. Statistical significance with antimetabolites as combination therapy (OR: 2.343; p value: 0.018) and not with microtubule stabilizer (OR: 0.377) were remarkable, indicating probability of less predominance of M or MSL type TNBC in a population. PARP inhibitors or T cell targeted therapies were also found promising (OR: 1.120, 1.040 respectively), warranting their targeted usage for BRCA1 deficient and IM type TNBCs respectively.

Conclusions

For TNBC treatment, personalized medicine and not a generalized treatment strategy should be considered.

Clinical trial identification

NA

Legal entity responsible for the study

Netaji Subhas Chandra Bose Cancer Research Institute

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

287P - Neutrophil-to-lymphocyte ratio in metastatic breast cancer: Association with clinico-pathological features and outcome (ID 4689)

Presentation Number
287P
Lecture Time
13:15 - 13:15
Speakers
  • M. Bartoletti
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Tumors are closely linked with systemic inflammation, of which neutrophil-to-lymphocyte ratio (NLR) represents a simple and inexpensive tool of evaluation. Previous data suggested that a high NLR is associated with poor prognosis in several tumors, including breast cancer (BC). However, few studies involved patients (pts) with metastatic breast cancer (mBC).

Methods

We retrospectively analyzed clinico-pathological features and treatment outcome of 595 consecutive mBC pts treated at the Department of Oncology of Udine, Italy, between 2004 and 2014. NLR was calculated from the blood count performed before first line therapy start. Differences in NLR according to clinico-pathological characteristics were investigated through chi-square test. Cox regression was used to determine the prognostic impact of NLR.

Results

A statistically significant higher NLR was found in pts whose tumor had the following features: high grade (P = 0.009), ductal isotype (P = 0.02), ER negativity (P = 0.003), PgR negativity (P = 0.0001), high Ki-67 (P = 0.03). There were no statistical differences in NLR between HER2-positive and HER2-negative BC (P = 0.33). Among subtypes, triple-negative BC were associated with higher NLR, while luminal HER2+ BC with lower NLR (P = 0.004). No statistical differences in NLR were found according to visceral disease (P = 0.13) nor according with bone-only disease (P = 0.24). At univariate analysis, a NLR ≥2.64 was associated with worse progression-free survival after first line therapy (HR 1.41, 95%CI 1.11-1.79, P = 0.005) and with worse overall survival (HR 1.76, 95%CI 1.32-2.36, P < 0.0001). The statistical significance was lost at multivariate analysis (P = 0.08 and P = 0.13, respectively). Of note, a subgroup analysis showed a significant prognostic value of NLR in HER2-positive subtype (HR 4.89, 95%CI 1.13-21.23).

Conclusions

High NLR was associated with pathological features of BC, but did not represent an independent prognostic factor at multivariate analysis. Further investigation is warranted to identify the appropriate cut-off value of NLR and the BC subtypes in which its prognostic role could be more useful.

Legal entity responsible for the study

University of Udine

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

288P - Prognosis after loco-regional recurrence of breast cancer: 35 years longitudinal data from the Stockholm cancer register (ID 2206)

Presentation Number
288P
Lecture Time
13:15 - 13:15
Speakers
  • C. Falato
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Loco-regional recurrence (LRR) of breast cancer is a significant cause of morbidity and mortality. It is poorly described how prognosis after LRR has evolved over time at the population level.

Methods

2272 patients diagnosed with LRR between 1980-2014 were identified within the Stockholm cancer registry and divided in 7 cohorts by the year of LRR diagnosis. Post-relapse event free survival (EFS) and overall survival (OS) were analyzed separately in local and loco-regional relapses and compared across the cohorts by Cox regression method. Primary tumor size, axillary node status, estrogen receptor (ER) status, type of surgery, adjuvant chemotherapy, LRR free survival, and age at LRR were the covariates for Cox model adjustment.

Results

In 1615 patients diagnosed with local relapse, 903 post-LRR events were registered (Table). A significant improvement in median EFS (p < .001) and OS (p < .001) was observed in patients diagnosed 2010-14 compared with previous time periods. Among 657 patients with loco-regional recurrences, 476 experienced a post-LRR event (Table). EFS and OS independently improved over time (p <.001 and p <.001, respectively). Smaller primary tumors, negative axillary lymph nodes, ER positive status, breast-conserving surgery, longer LRR-free interval and younger age at LRR occurrence were independently associated with longer survival after LRR. No association was observed between survival and type of surgery or LRR-free interval in LRRs. An improvement in survival over time was also demostrated when cohorts 1980-84 and 2010-14 were excluded from the model.

288P

Local recurrenceLocoregional recurrence
Type of post-ILRR relapseN (%)N (%)
Loco-regional117 (13%)57 (12%)
Distant438 (49%)305 (64%)
Loco-regional + distant13 (1%)3 (1%)
Death335 (37%)111 (23%)
Total903 (100%)476 (100%)

Conclusions

Survival after LRR has gradually improved over the last 35 years regardless of other recognized prognostic factors.

Legal entity responsible for the study

Karolinska Institutet

Funding

Dagmar Ferbsminnesfond

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

289P - The CAN BEAR study: A systematic review and meta-analysis investigating adverse events (AEs) of targeted agents added to endocrine therapy (ET) in patients (pts) with hormone-receptor positive (HR+) metastatic breast cancer (MBC) (ID 4526)

Presentation Number
289P
Lecture Time
13:15 - 13:15
Speakers
  • S. Martel
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Combining targeted agents and ET improves outcomes in pts with HR+ MBC but increases the risk of AEs. However, the specific additional toxicity burden caused by these agents remains unknown. Our meta-analysis aims to better estimate the comparative risk of AEs with the combination of ET and CDK4/6 inhibitors, PI3K inhibitors, mTOR inhibitors and anti-HER2 agents in pts with HR+ MBC.

Methods

A systematic literature search of MEDLINE, EMBASE, Cochrane Library and proceedings from major conferences up to March 31st 2017 was conducted to identify randomized controlled trials investigating ET plus CDK4/6, PI3K, mTOR inhibitors and anti-HER2 agents as compared to ET alone in pts with HR+ MBC. For each class of targeted agents, two groups were considered: ET plus targeted agent vs. ET alone. Summary risk estimates (odds ratio, OR) and 95% confidence intervals (CI) were calculated for each side effect within each class of targeted agents for each trial. Pooled analysis was conducted using the random and fixed effects models.

Results

A total of 7865 pts from 15 studies were included in our meta-analysis. Overall, the addition of targeted agents to ET was associated with significant higher risk of grade 3-4 AEs: OR 2.95 (95% CI 2.47-3.53) for CDK4/6 inhibitors, 2.05 (95% CI 1.63-2.58) for PI3K inhibitors, 1.89 (95% CI 1.40-2.56) for mTOR inhibitors, and 2.33 (95% CI 1.17-4.63) for anti-HER2 agents. Anti-HER2 agents, CDK4/6 and PI3K inhibitors significantly increased the risk of grade 3-4 fatigue, but not mTOR inhibitors (OR 1.48; 95% CI 0.64-3.43). Anti-HER2 agents, PI3K and mTOR inhibitors significantly increased the risk of grade 3-4 diarrhea, but not CDK4/6 inhibitors (OR 1.15; 95% CI 0.46-2.87). Other AEs and class specific toxicities will be reported at the conference.

Conclusions

In pts with HR+ MBC, the combination of targeted agents and ET is associated with significant increased risk of AEs. The risk of developing different AEs varies largely according to the type of agent used. Potential specific toxicities should be taken into account and discussed with patients when deciding to opt for combination regimens.

Clinical trial identification

PROSPERO registration number: CRD42017058278

Legal entity responsible for the study

Samuel Martel

Funding

None

Disclosure

E. De Azambuja: Honoraria from Roche and travel grants from Roche and GlaxoSmithKline outside the submitted work. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

290P - OlympiAD: Health-related quality of life (HRQoL) in patients with HER2-negative metastatic breast cancer (mBC) and a germline BRCA mutation (gBRCAm) receiving olaparib monotherapy vs standard single-agent chemotherapy treatment of physician’s choice (TPC) (ID 4542)

Presentation Number
290P
Lecture Time
13:15 - 13:15
Speakers
  • M. Robson
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The Phase III OlympiAD study showed a statistically significant and clinically meaningful PFS survival benefit with olaparib monotherapy, compared with standard of care chemotherapy (median 7.0 vs 4.2 months, respectively, hazard ratio 0.58; 95% CI 0.43, 0.80; P = 0.0009) in patients (pts) with HER2-negative mBC and a gBRCAm. A key predefined secondary objective was to assess the effect of olaparib on HRQoL.

Methods

The randomized, open-label, Phase III OlympiAD study (NCT02000622) enrolled pts with HER2-negative mBC and a gBRCAm, after ≤2 chemotherapy lines for mBC. Pts were randomized 2:1 to olaparib tablets (300 mg bid) or single-agent treatment of physician’s choice (TPC; capecitabine, vinorelbine or eribulin). Pts were asked to complete an EORTC QLQ-C30 questionnaire (analysis focused on the Global HRQoL scale with range 0–100, and higher scores indicating a better QoL), at baseline and every 6 weeks until disease progression. Changes in Global HRQoL scores were analyzed descriptively, and mean change from baseline (cfb) by a mixed model for repeated measures.

Results

302 pts (ITT) were randomized to olaparib (n = 205) or TPC (n = 97). Overall QLQ-C30 compliance rate was 93% for olaparib vs 77% for TPC. HRQoL was better preserved with olaparib than TPC (mean cfb in Global HRQoL score across all visits was 3.9 [n = 191] vs -3.6 [n = 73], respectively, difference 7.5; 95% CI 2.48, 12.44; P=0.0035). The proportion of pts (ITT) who were free of Global HRQoL deterioration (cfb decrease in ≥ 10 points) was 81.5% in the olaparib arm vs 61.2% in the TPC arm at 6 months, and 64.0% vs 53.5% at 12 months, respectively. The median time to Global HRQoL deterioration was not reached in olaparib pts, and was 15.3 months for TPC pts. A best HRQoL response of ‘improved’ (cfb increase in ≥ 10 points over two visits ≥21 days apart) was observed in 34% olaparib pts vs 13% TPC.

Conclusions

Pts receiving olaparib experienced significantly less and later deterioration in Global HRQoL vs TPC. HRQoL was modestly and consistently greater in patients receiving olaparib compared with TPC.

Clinical trial identification

Clinical trials no: NCT02000622

Release date: 18 November 2013

AstraZeneca name: OlympiAD

AstraZeneca number: D0819C00003.

Legal entity responsible for the study

AstraZeneca

Funding

AstraZeneca

Disclosure

M. Robson: Consultancy: AstraZeneca and McKesson. Travel, accommodation and expenses and honoraria: AstraZeneca. Research funding: AstraZeneca, AbbVie, Myriad Genetics and Medivation. S-A. Im: Research grant from AstraZeneca. E. Senkus-Konefka: Honoraria, Consulting/Advisory: Amgen, AstraZeneca, Pfizer, Pierre Fabre, Roche. Travel, accommodation, expenses: Amgen, AstraZeneca, Pfizer, Novartis, Roche. S.M. Domchek: Honorarium from EMD Serrano.The University of Pennsylvania has received research funding from AbbVie and Clovis. N. Masuda: Personal honoraria from: Chugai Pharma and AstraZeneca. Institution research funding from: Chugai Pharma, AstraZeneca, Kyowa Hakka Kirin, MSD, Novartis, Pfizer and Lilly. S. Delaloge: AstraZeneca advisory board member, and institution funded for sponsored research from AstraZeneca. N. Tung: Research funding from Myriad and Ambry. A. Armstrong: Consulting/advisory: Roche, Syndax. W. Wu, C. Goessl, A. Degboe: AstraZeneca employee with stocks. P.F. Conte: Speakers\' bureau and advisory board: AstraZeneca. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

291P - PALOMA-2: Neutropenia (NP) patterns in patients (Pts) with estrogen receptor−positive (ER+)/human epidermal growth factor receptor 2−negative (HER2–) first-line advanced breast cancer (ABC) receiving palbociclib + letrozole (P+L) (ID 4016)

Presentation Number
291P
Lecture Time
13:15 - 13:15
Speakers
  • V. Diéras
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

PALOMA-2 demonstrated efficacy of P+L vs placebo (PBO) + L in pts with treatment-naive ER+/HER2– ABC (Finn NEJM 2016). We describe clinical patterns of hematologic adverse events (AEs), with an emphasis on NP, in pts receiving P+L.

Methods

Postmenopausal women (N = 666) with no prior systemic therapy for ABC were randomized 2:1 to receive P+L (P, 125 mg/d, 3 wk on/1 wk off; L, 2.5 mg/d continuously) or PBO+L (L, 2.5 mg/d continuously) until disease progression, unacceptable toxicity, or consent withdrawal. Hematological AEs are reported based on lab results.

Results

As of 2/26/2016, median follow-up was 23.0 mo in pts receiving P+L (n = 444). Median age of P+L pts was 62.0 (range, 30–89) years; ECOG status was 0, 1, and 2 in 57.9%, 40.1%, and 2.0%, respectively; and 213 (48.0%) received prior chemotherapy. 423 (95.3%) P+L pts experienced any grade (gr) NP, including 298 (70.4%) with gr 3/4 NP, manageable with dose modification. Among pts with gr 3/4 NP, 65 (15.4%), 41 (9.7%), and 192 (45.4%) experienced 1, 2, or ≥ 3 episodes, respectively. 92 (20.7%) and 84 (18.9%) pts experienced 3–5 episodes of any grade anemia and thrombocytopenia, respectively. Median (range) times to first episode of gr ≥ 3 NP, anemia, and thrombocytopenia were 28.0 d (12 − 854 [median duration, 31.5]), 182.0 d (14 − 760 [11.5]), and 283.5 d (21 − 617 [26.5]), respectively. Although NP is associated with increased risk of infection, the rate of gr 3/4 infections was 3.5% in P+L pts with NP. Of pts with gr 3/4 NP, 68.8% did not have any overlapping infections. Febrile NP was reported in 1.8% of P+L pts and did not result in therapy discontinuation. In univariate analysis, risk of developing gr 3/4 NP was associated with Asian ethnicity (P=0.0002) and low baseline absolute neutrophil counts (P<0.0001). NP resulting in dose reduction or interruption had no impact on PFS.

Conclusions

NP occurred early during therapy, and was manageable with dose modification. Febrile NP was reported in 1.8% of P+L pts and did not result in therapy discontinuation. Withholding dose, or dose reduction does not negatively impact PFS. Funding, Pfizer.

Clinical trial identification

NCT01740427

Legal entity responsible for the study

Pfizer Inc

Funding

Pfizer Inc

Disclosure

V. Diéras: Consulting and advisory role: Genentech, Lilly, Pfizer, AbbVie, Novartis Pharma KK, Roche-Peru. Speakers bureau: Pfizer, Novartis Pharma KK, Roche-Peru. N. Harbeck: Honoraria: Lilly, Novartis, Pfizer. A.A. Joy: Honoraria: Pfizer, Novartis, Roche, Eli Lilly, AstraZeneca, Bristol-Myers Squibb, Boehringer Ingelheim. Consulting or Advisory: Pfizer, Novartis, Roche, Eli Lilly, AstraZeneca, Bristol-Myers Squibb, Boehringer Ingelheim. K.A. Gelmon: Consulting or Advisory Role: Pfizer, Novartis, AstraZeneca, NanoString Technologies, Merck. J. Ettl: Honoraria: Pfizer, Novartis Pharma KK, Roche-Peru. Consulting or advisory role: Pfizer, Novartis Pharma KK. Speakers bureau: Pfizer, Novartis Pharma KK, Roche KK. S. Verma: Consulting and advisory role: Genentech/Roche, Lilly, Pfizer, Novartis, Amgen. D. Lu, E.R. Gauthier, P. Schnell, A. Mori: Pfizer employee and shareholder. H.S. Rugo: Speakers bureau and honoraria: Genomic Health. Research funding: Plexxikon Macrogenics, OBI Pharma, Eisai, Pfizer, Novartis, Lilly, GlaxoSmithKline, Genentech, Celsion, Merck, Clovis Oncology. R.S. Finn: Honoraria: Bayer, Pfizer, Bristol-Myers Squibb, Novartis, Eisai. Consulting or advisory role: Pfizer, Bayer, Novartis, Bristol-Myers Squibb, Merck. Research funding: Pfizer.

Collapse
Poster display session Poster Display session

292P - Prospective observational study of peripheral neuropathy in breast cancer patients treated with eribulin (ID 958)

Presentation Number
292P
Lecture Time
13:15 - 13:15
Speakers
  • Y. Sakata
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Eribulin mesylate (ERI), a nontaxane microtubule dynamics inhibitor, has antitumor activity and can prolong overall survival in patients with recurrent breast cancer (BC). As data lack on peripheral neuropathy (PN) when using ERI as primary or secondary therapy for recurrent BC, we assessed the incidence of PN, the times to PN onset and recovery, and the risk factors for PN in patients with HER-2-negative recurrent or metastatic BC treated with ERI.

Methods

We analyzed data from an ongoing 2-year multicenter prospective observational study where the same number of patients who started ERI administration as primary or secondary therapy and as tertiary or later therapy were enrolled. PN events were defined as new onset or existing PN worsened after ERI administration. Logistic regression was run to assess the risk factors.

Results

The analysis set comprised 458 patients who had completed 6-month follow-up after ERI administration. Mean age ± standard deviation was 59.4 ± 10.9 years, and 171 patients (37.3%) had a history of PN from previous chemotherapy and 190 (41.5%) had existing PN at start of ERI administration (baseline). PN events were observed in 115 patients (25.1%), with severity of Grade 1 in 55 (12.0%), Grade 2 in 51 (11.1%), and Grade 3 in 9 (2.0%) in accordance with the Common Terminology Criteria for Adverse Events version 4.0. After the PN events, 91 patients (79.1%) continued ERI, and 16 (13.9%) reduced the dose or underwent a drug holiday. Within 6 months after ERI administration, 39.0% recovered to the state at baseline. In 266 patients without PN at baseline, new PN appeared in 71 patients (26.7%), and median time to PN onset was 57.0 days (95% confidence interval, 43.0–66.0 days). Results also showed that “history of radiotherapy”, “hemoglobin level”, and “history of PN from previous chemotherapy” were significantly associated with PN events, but no evident association was found with the number of chemotherapy prior to baseline.

Conclusions

PN events were observed in about one-quarter of patients treated with ERI, most of which were mild, and about one-third of patients who developed PN recovered early. This suggests that ERI is well tolerated.

Clinical trial identification

NCT02371174 (First release on November 21, 2014)

Legal entity responsible for the study

NA

Funding

Eisai Co., Ltd., Tokyo, Japan

Disclosure

Y. Sakata, H. Iwata, H. Ikezawa, R. Fudetani, D. Tonoda, Y. Uchida, H. Hasegawa, T. Matsuoka: Employee of Eisai Co., Ltd.

Collapse
Poster display session Poster Display session

293P - Overall survival and quality of life in patients with metastatic breast cancer treated with nab-paclitaxel: Final results of the non-interventional study NABUCCO (ID 3409)

Presentation Number
293P
Lecture Time
13:15 - 13:15
Speakers
  • K. Potthoff
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Nab-paclitaxel (Nab-P) is approved for the treatment of metastatic breast cancer (MBC) after first line therapy and when anthracyclines are not indicated. Clinical trials proved high efficacy and reduced toxicity of nab-P compared to standard taxanes. Real world data of nab-P in MBC, however, are still limited.

Methods

The prospective, multicenter, non-interventional NABUCCO study was designed to collect data on effectiveness including overall survival, safety, treatment patterns and quality of life (QoL) in patients (pts) with MBC in real world. QoL was assessed with the validated questionnaires Functional Assessment of Cancer Therapy-General (FACT-G) and the breast cancer and taxane specific modules (FACT-B and FACT-Taxane) at baseline (BL), 3, 6 months. Data were analyzed descriptively. Survival was analyzed with the Kaplan-Meier method.

Results

697 of 705 pts with MBC enrolled at 128 sites in Germany from 4/2012 to 4/2015 were evaluable (median age 62.3 years (yrs) (min-max 29.2-89.3); age ≥65 yrs n = 291 (41.8%), ECOG 0/1 n = 628 (90.1%), prior taxanes n = 419 (60.1%)). 194 pts (27.8%) received 220-260 mg/m2 q3w, 491 pts (70.4%) received weekly nab-P at ≤ 150 mg/m2 (physician’s discretion; 1.7% other). Median overall survival (mOS, months [95% CI]) was 15.6 [14.2-17.2]. No difference was observed with regard to treatment pattern (15.1 [12.3-17.5] q3w vs 16.3 [14.4-18.5] weekly) and age (15.7 [14.0-18.1] <65 yrs vs 15.1 [12.8 - 17.3] ≥ 65 yrs). mOS was significantly shorter in pts receiving prior taxanes (13.7 [11.7-15.5] vs. 18.3 [16.4-22.2]) or prior chemotherapy in general (19.2 [16.9-22.2], 15.1 [12.5-17.3], 14.1 [10.3-17.2], 11.3 [9.1-12.7] with 0, 1, 2, ≥ 3 prior palliative lines). Consistent with safety data, pts reported increased taxane-related symptoms after start of nab-P (BL vs 6 months; [range]: FACT-Taxane subscale score 52.3 [4.0-64.0] vs 40.0 [3.0-64.0]). Global and breast cancer related QoL were not affected (FACT-G 71.3 [27.0-105.0] vs 67.0 [14.0-104.0]; FACT-B subscale score 23.6 [0.0-35.0] vs 22.0 [2.0-35.0]).

Conclusions

The NABUCCO study confirms survival data from clinical trials in real world without deteriorated global and breast cancer related QoL.

Clinical trial identification

IOM-02240

Legal entity responsible for the study

iOMEDICO AG

Funding

Celgene

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

294P - Assessment of gastric pH changes and food intake on ribociclib bioavailability: In silico and clinical evaluations (ID 4008)

Presentation Number
294P
Lecture Time
13:15 - 13:15
Speakers
  • T. Samant
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Ribociclib (KISQALI®) is a CDK4/6 inhibitor that has been approved recently in the United States for use in combination with an aromatase inhibitor as a first-line therapy for HR+, HER2− advanced or metastatic breast cancer. The recommended ribociclib dose is 600 mg/day (3-weeks-on/1-week-off) with no restrictions on food intake or concomitant proton pump inhibitor (PPI) use. Here we examine the influence of gastric pH and food intake on ribociclib bioavailability using physiology-based pharmacokinetics (PBPK) modelling, observed clinical pharmacokinetic (PK) data, and population PK (pop-PK) analysis.

Methods

In silico PBPK models based on in vitro and preclinical data were built and fitted with clinical PK data from healthy volunteers. Sensitivity analyses were performed to evaluate the effect of gastric pH (range, 0.5-8.0) on ribociclib PK and absorption. A descriptive statistical analysis of clinical PK data from patients with and without concomitant PPI use and a pop-PK analysis were used to examine the effect of PPI dose intensity on ribociclib bioavailability. The effect of a high-fat meal on ribociclib exposure was evaluated in a bioequivalence trial in healthy volunteers.

Results

Sensitivity analyses using validated PBPK models predicted no effect of varying stomach pH on ribociclib absorption. PK data (AUC0-24h and Cmax) from several clinical studies showed similar ribociclib exposure regardless of PPI use (Table). The pop-PK analysis supported the PBPK models and clinical findings by showing that PPI use is a statistically insignificant and clinically unimportant covariate on ribociclib bioavailability. Food intake did not affect the rate or extent of ribociclib absorption.

294P Ribociclib PK parameters by PPI usea

Study No.PK ParameternPPI UseGeometric Mean (Geometric Coefficient of Variation, %)
X2107AUC0-24h (ng*hr/mL)8Yes24,700 (30.6)
10No21,100 (57.2)
Cmax (ng/mL)10Yes1,780 (34.6)
13No1,620 (53.2)
X2101AUC0-24h (ng*hr/mL)12Yes25,900 (79.1)
46No23,700 (61.3)
Cmax (ng/mL)13Yes2,050 (74.7)
48No1,870 (60.3)
X1101AUC0-24h (ng*hr/mL)2Yes42,600 (28.7)
6No55,100 (68.6)
Cmax (ng/mL)2Yes2,700 (53.0)
6No3,500 (65.8)

AUC0-24h, area under the concentration-time curve from time zero to 24 hours; Cmax, maximal concentration; PK, pharmacokinetics; PPI, proton pump inhibitor. aDefined by PPI use prior to and on the day of sampling on C1D15 for AUC0-24h and Cmax, and on the dosing date corresponding to the AUC0-24h or Cmax. “Yes” was defined as PPI use for at least 5 consecutive days; “No” was defined as no PPIs use for at least 13 consecutive days.

Conclusions

In silico models and clinical PK data indicate that ribociclib can be administered without regard to PPI use or food intake. This lack of dosing restriction may facilitate greater patient compliance and clinical benefit.

Legal entity responsible for the study

Novartis Pharmaceuticals Corporation

Funding

Novartis Pharmaceuticals Corporation

Disclosure

T. Samant, M. Elmeliegy, Y. Lu, S. Yang, M. Miller, C. Germa: Employee of Novartis Pharmaceuticals Corporation. S. Dhuria: Was an employee Novartis Pharmaceuticals Corporation at the time this study was conducted; currently a consultant for Novartis Pharmaceuticals Corporation. M. Laisney, A. Grandeury, M. Mueller-Zsigmondy, K-I. Umehara, F. Huth: Employee of Novartis Pharma AG.

Collapse
Poster display session Poster Display session

295P - Pneumocystis jiroveci pneumonia (PCP) in patients receiving weekly chemotherapy for metastatic breast cancer (ID 1061)

Presentation Number
295P
Lecture Time
13:15 - 13:15
Speakers
  • S. Wijaya
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Pneumocystis jiroveci pneumonia (PCP) is thought to be a rare phenomenon in the solid tumour population, particularly in patients with breast cancer. However, it may be increasing within this population as the type and intensity of chemotherapy used changes. There is currently a lack of consensus on PCP prophylaxis in patients who are immunocompromised due to chemotherapy.

Methods

The EPIC electronic health record system was searched for metastatic breast cancer (MBC) patients treated with a weekly chemotherapy (epirubicin/paclitaxel) regimen from October 2014 – February 2016 at Addenbrooke’s hospital (n = 49). A subset of patients diagnosed with PCP (n = 5) was identified. A retrospective analysis was performed on the charts of all patients.

Results

Patients received a mean of 21 weeks (SD = 15, min=1, max=62) of chemotherapy. An overall of 16% (n = 8) of patients had profound lymphopaenia (absolute lymphocyte count <0.5*10ˆ9/L) at some point during their treatment. 10% (n = 5) of the patients were diagnosed with confirmed (n = 3) and probable (n = 2) PCP.

Conclusions

A high incidence of PCP was observed in MBC patients receiving weekly epirubicin/paclitaxel treatment. Additional investigation is needed to define the population of patients at the greatest risk of PCP infection, and to identify those who might benefit from antibiotic prophylaxis.

Legal entity responsible for the study

Cambridge Cancer Centre

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

296P - Detection of early cardiac effects of docetaxel plus trastuzumab and pertuzumab through strain rate imaging in patients with HER2-positive metastatic breast cancer (ID 3097)

Presentation Number
296P
Lecture Time
13:15 - 13:15
Speakers
  • V. Michalaki
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Dual anti- HER2 therapy with trastuzumab and pertuzumab in combination with taxane-based chemotherapy improves overall survival in patients with metastatic HER2-positive breast cancer. There is a critical need to investigate the potential cardiotoxicity of dual anti-HER2 blockade, given the importance of HER2 signaling in cardiac homeostasis and stress response. Sequential left ventricular (LV) ejection fraction (EF) assessment has been mandated to detect myocardial dysfunction. Changes in cardiac function induced by this therapy, however, are subtle and difficult to quantitate by conventional imaging methods. Doppler myocardial imaging-based velocity, strain, and strain rate measurements have been shown to sensitively quantify abnormalities in cardiac function in other settings. The aim of this study was to determine if sensitive indices of left ventricular (LV) dysfunction, specifically strain rate imaging, would be useful for addressing the early detection of dual anti-HER2 mediated cardiotoxicity.

Methods

Patients with 0-1 lines of prior therapy were treated with 8 cycles of docetaxel (75mg/m(2) plus trastuzumab (8 mg/kg loading dose followed by 6 mg/kg) and pertuzumab (840 mg loading dose followed by 420 mg every 3 weeks. Conventional and Doppler myocardial imaging echocardiography were obtained at baseline and every 2 cycles of treatment. Segmental peak systolic longitudinal and radial velocity, strain, and strain rate (SR) were measured.

Results

Twenty-seven women (median age 52.2 years) were enrolled in the study. There was no overall change in Left ventricular dimensions, ejection fraction, and systolic myocardial velocity. In contrast, a significant reduction in longitudinal and radial strain and strain rate was found after 8 cycles (longitudinal strain -12.8% +/- 2.2% vs baseline (P = .001); radial strain 29.3% +/- 7.1% vs 50.3% +/- 10.6%, P < .001 vs baseline). Changes in radial function appeared earlier and were more pronounced than in longitudinal direction.

Conclusions

In contrast with conventional echocardiography myocardial velocity measurements allowed detecting subtle changes in longitudinal and radial left ventricular function after 8 cycles of therapy. We suggest that strain rate imaging identifies preclinical myocardial dysfunction earlier than conventional measures in women undergoing treatment with dual anti-HER2 therapy for metastatic breast cancer and could be used for cardiac function monitoring.

Legal entity responsible for the study

Vasiliki Michalaki

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

297P - Evaluation of drug-drug interactions (DDI) between tucatinib and capecitabine (C) in patients with advanced HER2+ metastatic breast cancer from a phase 1b study (ID 4219)

Presentation Number
297P
Lecture Time
13:15 - 13:15
Speakers
  • A. Vo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Tucatinib is an orally bioavailable, potent HER2 selective tyrosine kinase inhibitor. Based on the combination activity with chemotherapy and trastuzumab (Tz) in preclinical HER2+ tumor models, tucatinib was evaluated in combination with C and Tz in a Phase 1b study in patients with HER2+ metastatic breast cancer (mBC).

Methods

A phase 1b 3 + 3 dose escalation study (ONT-380-005) was conducted to evaluate the safety and tolerability of tucatinib in combination with C and Tz. Tucatinib (300 mg PO BID), C (1000 mg/m2 PO BID 14 days of a 21-day cycle), and Tz (8 mg/kg IV loading; then 6 mg/kg IV once every 21 days), were administered to HER2+ mBC patients previously treated with Tz and T-DM1. Pharmacokinetic (PK) assessments were conducted on cycle 1 day 14 (+C) and on cycle 1 day 21 (-C). PK of C and its major catabolites/metabolites were also measured. In vitro assessments of the activation of C were determined in the presence of tucatinib. The enzymes evaluated were carboxyesterase (CES), cytidine deaminase (CDA), thymidine phosphorylase (TP), and dihydropyrimidinephosphorylase (DPD).

Results

Tucatinib did not inhibit conversion of C to 5’-DFCR in vitro; at 10 µM tucatinib reduced ∼30% of CES activity. Similarly, tucatinib did not have any significant effect on the activity of CDA, TP, or DPD. Results from in vitro inhibition studies suggested tucatinib does not have a measurable effect on the conversion of C to its active antimetabolite. The clinical PK of tucatinib was unchanged in the presence or absence of C. The PK of C and its catabolites/metabolites were also unaffected in the presence of tucatinib, and were consistent with reported literature.

Conclusions

The overall in vitro and clinical results indicate there is no evidence for DDI between tucatinib and C, including when the combination is given with Tz. The tucatinib-Tz-C triplet combination has been reported to be well tolerated and supports the evaluation of the efficacy of the combination regimen in an ongoing controlled, randomized, double-blinded registrational study (HER2CLIMB).

Clinical trial identification

ONT-380-005

Legal entity responsible for the study

Cascadian Therapeutics, Inc.

Funding

Cascadian Therapeutics, Inc.

Disclosure

A. Vo, D. Leviten, T. Sierra, A. Dozier, L. Walker, S. Peterson: Minor stockholder and employee of Cascadian Therapeutics, Inc. M. Insko: Minor stockholder of Cascadian Therapeutics, Inc., BLPH, and Faraday Pharmaceuticals Inc. Employee of Faraday Pharmaceuticals Inc.

Collapse
Poster display session Poster Display session

298P - Outcomes of intracranial stereotactic radiotherapy (SRT) in metastatic breast cancer (BC) (ID 5132)

Presentation Number
298P
Lecture Time
13:15 - 13:15
Speakers
  • G. Kothari
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Brain metastases (BM) are a significant cause of morbidity and mortality. Advancement of systemic therapies for patients with BC has improved control of extra-cranial metastases and survival. Intracranial control however continues to be challenging. SRT has been shown to provide excellent local control (LC) with minimal toxicity, although breast specific data is comparatively lacking.

Methods

This study aims to describe outcomes of first SRT including LC, distant brain control (DC), time to intracranial progression (TTP) and overall survival (OS) in a cohort of patients with BC who received SRT from 2001 to 2016 at the Royal Marsden Hospital. Kaplan Meier and log-rank methods were used for statistical analysis.

Results

64 patients underwent SRT for 129 BM. Median age was 52.4 years. 58 (91%) were ECOG 0/1. 18 (28%) were hormone receptor positive (HR+)/HER-2 negative, 38 (59%) were HER-2 enriched (HER-2+), and 8 (13%) were triple negative (TN). The median number of BM treated with SRT was 1 (range 1 - 12). Median dose and range was 20 Gy (12 – 35 Gy) in 1 fraction (1 – 10). 29 (45%) were treated using a linear accelerator, 34 (53%) with CyberKnife and 1 with GammaKnife. 27 (42%) had prior whole or partial brain radiotherapy (WBRT). 21 (33%) had prior surgery. 30 (49%) had concurrent endocrine therapy, 25 (40%) had targeted therapy and 8 (13%) had chemotherapy. Follow-up imaging was available for 57 patients. Median follow up was 15 months. 1 year LC was 54% and DC was 56%. Median TTP was 7.1 months (95%CI: 6.1 – 10.8) and was significantly worse for TN compared to HER-2+ patients (5.3 vs 9.9 months, p = 0.046). Salvage radiotherapy or surgery was performed after SRT in 13 (23%) patients for local failure, and 20 (35%) for distant failure. 7 (12%) patients had radionecrosis following SRT. OS from SRT was 19.6 months (95%CI: 14.9 – 23.1), and was significantly worse for TN patients (13.6 vs 22.1 months, p = 0.003), age ≥ 60 years (14.9 vs 19.8 months, p = 0.028) and multiple brain metastases (14.0 vs 22.6 months, p = 0.010).

Conclusions

Outcomes in patients with BC and BM treated with SRT are excellent overall. Outcomes are worse in TN disease, older patients and patients with multiple BM, and should be considered in SRT decision making.

Legal entity responsible for the study

The Royal Marsden NHS Foundation Trust

Funding

The Royal Marsden NHS Foundation Trust/Ross Smith Cridlan Trust

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

299P - Eribulin is safe and efficient in metastatic breast cancer in elderly patients. Results from the REPROLINE multicentric retro-prospective cohort (ID 2555)

Presentation Number
299P
Lecture Time
13:15 - 13:15
Speakers
  • J. MARTIN-BABAU
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Treating metastatic breast cancer (MBC) in women of 70 years old or more is a frequent problem however few data are available describing the safety and efficacy of chemotherapy in elderly patients. Eribulin is validated for MBC from the 2nd line treatment onwards since two phase III studies. We present here a focus on safety and efficacy of eribulin in patients ≥ 70 years old compared to the results of younger patients in a real life cohort.

Methods

From Oct 2014 to Feb 2017, a multicentric retro-prospective study (REPROLINE) was conducted. Data concerning patient, tumor characteristics, previous treatments administered, tolerance, efficacy and outcome of eribulin were retrieved for patients treated in real life for MBC in 12 different French hospitals between Dec 2013 and Jan 2016. Data from 446 MBC patients were collected. This database was splitted in two cohorts: comparing the results from the cohort of 363 patients < 70 years old (group A) to the cohort of 83 patients ≥70 years old or more (group B).

Results

Median age for each cohort was 56.3 and 75.4 years old. Both cohorts had similar tumour characteristics, number of metastatic sites and the median number of prior chemotherapy lines was 2. Albumine serum levels were lower in the group B with 21% of patients with albumine < 30g/L versus 13% in group A, without any statistical difference. Outcomes were similar in group A and B with respectively: median PFS of 3.67 months versus 3.7 months; HR 0.972(CI 95% 0.762-1.241), p = 0.821. median OS 10.7 months vs 10.7 months; HR 0.997 (CI95% 0.752-1.323), p = 0.984. Patients in both groups received a median number of 4 cycles. The most frequent grade 3 adverse events were neutropenia (22.9% in group A and 15.7% in group B), fatigue (6.5% group A and 13.3% group B) and neurotoxicity (4.4% and 3.6%, respectively). No statistical difference between elderly and younger patients was demonstrated. 9.6% of the total patients stopped the treatment due to fatigue.

Conclusions

We present here the first study focusing retro-prospectively on the tolerance and efficacy of eribulin in elderly patients in real life. In this study, eribulin in patients ≥70 years old is as effective and safe as in younger patients.

Clinical trial identification

NCT02393287

Legal entity responsible for the study

Anne Patsouris

Funding

EISAI

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

300P - Long-term responders to trastuzumab monotherapy in the first-line metastatic setting: characteristics and survival data (SAKK 22/99 Trial) (ID 3538)

Presentation Number
300P
Lecture Time
13:15 - 13:15
Speakers
  • S. Schmid
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The prospective randomized trial SAKK 22/99 compared first-line trastuzumab plus chemotherapy with the sequential use of trastuzumab followed by chemotherapy plus trastuzumab at progression in patients with stage 4 HER2-positive breast cancer. The main results were recently published in Annals of Oncology. Here we report characteristics and outcomes of patients with long-lasting disease control with trastuzumab alone.

Methods

Long-term responders to trastuzumab monotherapy were defined as patients achieving disease control for ≥ 6 months. A risk score was defined as sum of negative prognostic factors (NPFs): ER negative (neg), PgR neg and visceral disease. The HER2:CEP17 FISH ratio was correlated with duration of response. Standard descriptive statistics were used. Survival analysis was done using the Kaplan-Meier method.

Results

Of 175 enrolled pts, 86 were randomised to receive trastuzumab monotherapy until PD. 24 patients (28%) were long-term responders (≥6 months). In a landmark analysis excluding patients who died within 6 months, 5y-overall survival (OS) in long-term responders was 54% (95% CI 31–72) compared to 18% (95% CI 10–30) in short-term responders (log-rank p = 0.02). Baseline characteristics were well-balanced except for visceral disease (see Table, Fisher’s exact test p = 0.01). With each additional NPF the proportion of long-term responders decreased: 0 NPF, 42%; 1 NPF, 40%; 2 NPFs, 35%; 3 NPFs 17%. Median FISH ratio was 4.8 (IQR 4.1–5.3) in long-term responders and 4.7 (IQR 2.5–5.3) in short-term responders; no long-term responders were in the lowest quartile of all FISH ratios.

300P Baseline characteristics

HER2-long Responders (N = 24)HER2-short Responders (N = 62)
Age (med)57 years52 years
Visceral disease (yes)46%76%
ER-Status (pos)59%60%
PR-Status (pos)48%41%
FISH (med. Ratio)4.84.7
ALP U/l (med)74103
Hb g/l (med)130134
WBC G/l (med)5.86.8
Endocrine treatment adj (yes)33%48%

Conclusions

Long response to trastuzumab monotherapy is prognostic with significantly higher 5y-survival rates in this cohort. Negative prognostic factors including lower FISH ratio may reduce the chance for long-term response to trastuzumab monotherapy as first line therapy.

Clinical trial identification

Subanalysis of SAKK 22/99 trial; NCT00004935

Legal entity responsible for the study

Swiss Group of Clinical Cancer Research (SAKK)

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

301P - Metronomic chemotherapy (mCHT) in HER2-ve advanced breast cancer (ABC) patients (pts): Old drugs, new opportunities Preliminary results of the VICTOR-6 study (ID 2175)

Presentation Number
301P
Lecture Time
13:15 - 13:15
Speakers
  • M. Cazzaniga
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

mCHT is the minimum biologically effective dose of a chemotherapeutic agent, given at regular dosing regimen with no prolonged drug free interval, that leads to anti-tumor activity. Old regimens included Cyclophosphamide-Methotrexate (CM), whereas in the last years new regimens, such as Vinorelbine (VRL) and Capecitabine (CAPE)-based have been developed. Aim of this observational retrospective ongoing study is to describe the use of mCHT in ABC pts across 5 years and the clinical characteristics of the pts together with efficacy of old (CM-like) vs new (VRL/CAPE-based) metronomic regimens in terms of response and disease control.

Methods

We retrospectively identified from clinical records those HER2-ve ABC pts who have received any kind of mCHT in the years 2011-2015, alone, or in combination with a non-metronomic drug. Standard statistical approaches were used for describing the sample characteristics. Logistic and non proportional hazard analysis were used to identify factors associated with response, and time to treatment failure and survival, respectively. This preliminary analysis focuses on Response Rate (RR) and Disease Control Rate (DCR).

Results

From June 2011 to December 2015, 267 pts have been identified till now and 233 are fully evaluable. Median age at mCHT start was 67 years. 81% was HR+ and 33% had non-visceral metastatic disease. 22% of the pts received CM, 55% VRL-based and 23% mCAPE-based regimens. mCHT use increased over the time from 15.0% (2011) to 30% (2015). As 1st-line treatment, CM was administered in 27% of compared with more than 48% of patients receiving CAPE/VRL-based regimens. Overall Response Rate (ORR) was 28% and Disease Control Rate (DCR) was 79%. Median duration of mCHT was 6.2 months. New generation metronomic regimens produced higher ORR in comparison to old ones (32% vs 13.5%), with similar duration of treatment (6.4 vs 5.4 months, respectively).

Conclusions

The use of mCHT in the treatment of HER2-ve ABC pts has deeply changed across the last 5 years, being new generation regimens used in earlier lines of treatment, producing interesting results in terms of objective response and disease control.

Legal entity responsible for the study

Marina Elena Cazzaniga

Funding

A&Q Consorzio per la riqualificazione agro-alimentare

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

302P - Factors associated with prolonged time to treatment failure with fulvestrant 500 mg in patients with postmenopausal estrogen receptor-positive advanced/metastatic breast cancer (JBCRG-C06; Safari): A subgroup analysis (ID 1449)

Presentation Number
302P
Lecture Time
13:15 - 13:15
Speakers
  • H. Kawaguchi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

This subanalysis of a retrospective, multicenter, cohort study of fulvestrant 500 mg (F500) in advanced/metastatic breast cancer (AMBC) patients in Japan (UMIN000015168) sought to identify clinical factors associated with prolonged time to treatment failure (TTF).

Methods

We analyzed ER+/human epidermal growth factor receptor 2 (HER2)-negative and ER+/HER2-positive patients who received F500 as 2nd- or later-line treatment. Factors investigated were age (≥65/≤65 years), treatment line (≥4th/3rd/2nd), time from AMBC diagnosis to F500 use (<3/≥3 years), prior palliative chemotherapy (no/yes), nuclear or histological grade (1/2/3), visceral metastasis (no/yes), ER expression (+/−), and progesterone receptor expression (+/−). TTF was determined using Kaplan–Meier analysis. TTF data were analyzed using univariate and multivariate analyses with a Cox proportional hazards model.

Results

We registered 1072 patients who received F500 between November 2011 and December 2014 at 16 sites in Japan. In the ER+/HER2− group (n = 828), median TTF was 5.4 months. By univariate analysis, higher age, earlier F500 use and no prior chemotherapy were associated with significantly longer TTF. By multivariate analysis, higher age, longer time from AMBC diagnosis to F500 use, no prior palliative chemotherapy and F500 treatment line were correlated with prolonged TTF (Table). In the ER+/HER2+ group (n = 132), treatment line was correlated with TTF (median 4.6 months) in the univariate analysis (P = 0.042); no factors significantly correlated with TTF in the multivariate analysis.

302P Factors correlated with TTF in ER+/HER2− AMBC patients

ER+/HER2−Hazard ratio95% confidence intervalP
Age (≥65/<65 years)0.850.73–0.990.035
Treatment line (≥4th/3rd/2nd)1.361.22–1.52<0.001
Time from diagnosis (≥3/<3 years)0.650.54–0.79<0.001
Prior chemotherapy (yes/no)1.341.13–1.58<0.001

Conclusions

In ER+/HER2− patients who received F500 as a ≥ 2nd-line treatment, treatment line, advanced age, no prior palliative chemotherapy and a longer time from AMBC diagnosis to F500 use were associated with longer TTF.

Clinical trial identification

UMIN000015168

Legal entity responsible for the study

Japan Breast Cancer Research Group (JBCRG)

Funding

Japan Breast Cancer Research Group (JBCRG) and AstraZeneca

Disclosure

H. Kawaguchi: Leadership Position/Advisory Role: Chugai, AstraZeneca. Consulting fee/honorarium: Chugai, AstraZeneca, Eisai, Kyowa Kirin, Novartis, Taiho. K. Aogi: Personal fees as honoraria: Chugai, Eisai, Sanofi, SRL, AstraZeneca, Taiho, Novartis, Daiichi Sankyo, Mochida, Ono, Otsuka, and Eli Lilly Japan, and the institution received research funds from Chugai, Eisai and Sanofi. N. Masuda: Personal fees as honoraria: Chugai Pharmaceutical and AstraZeneca, and the institution received research funds from Chugai Pharmaceutical and Eisai. T. Nakayama: Lecture\'s Fee: AstraZeneca, Chugai Pharmaceutical Co., Ltd. Novartis Pharma K.K. Y. Ito: Research Funds: MSD, AstraZeneca, Novartis, Parexel, Chugai, Lilly. T. Takano: Research Funds: Chugai, Takeda, Novartis. S. Saji: Honoraria from AstraZeneca, Chugai Pharmaceutical, Eisai, Novartis Pharma, and research funds from AstraZeneca and Chugai Pharmaceutical. S. Morita: Lecture\'s Fee and Consulting fee/honorarium: AstraZeneca. H. Yamashita: Research Funds: Taiho Pharmaceutical, Daiichi Sankyo, Chugai Pharmaceutical, Takeda Pharmaceutical. T. Yamashita: Consulting fee/honorarium: Chugai, Eisai, Novartis Pharma, Taiho, Nippon Kayaku, AstraZeneca, Takeda, Kyowa Kirin, Pfizer. Y. Yamamoto: Consulting fee/honorarium: Chugai, AstraZeneca, Novartis, Esai, KyowaHakko-Kirin, Taiho, Nipponkayaku, Takeda. S. Ohno: Consulting fee/honorarium: Chugai, AstraZeneca, Eisai, Novartis. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

303P - Durable complete response in HER2-positive breast cancer: A multicenter retrospective analysis (ID 3594)

Presentation Number
303P
Lecture Time
13:15 - 13:15
Speakers
  • A. Shimomura
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Though advanced and metastatic epidermal growth factor receptor 2 (HER2)-positive disease is not curable, a small proportion of patients with HER2-positive metastatic breast cancer remain in prolonged complete remission with anti-HER2 treatment. We hypothesized that some cases of human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer may be curable by trastuzumab. In this large, multicenter retrospective study, we aimed to assess the long-term outcomes for patients with a durable response to trastuzumab.

Methods

We retrospectively evaluated the data of patients diagnosed with HER2-positive metastatic breast cancer who received trastuzumab for more than 2 years as the first-line treatment. Patients diagnosed between April 1, 2001 and December 31, 2014 at 19 institutions in Japan were included in the analysis.

Results

A total of 108 patients were evaluated. Sixteen were met to exclusion criteria. The median follow-up length was 7.7 years. Disease progression occurred in 44/108 (40.7%) patients and 13/108 (12%) patients died. The median progression-free survival was 11.2 years, and as more than 80% of patients were alive 10 years after metastatic breast cancer diagnosis. Of the 108 patients, 57 achieved a clinical complete response. Trastuzumab therapy was interrupted for 27 (47.4%) of these patients (based on the doctor’s recommendation for 19 patients, owing to adverse events for 4 patients, owing to unknown reasons for 3 patients, and at the request of 1 patient). Disease progression occurred in 4 of the 27 patients after the interruption of trastuzumab treatment. The median duration of trastuzumab therapy for all 27 patients was 5.1 years (0.9-9.3 years).

Conclusions

In conclusion, we found that some patients showed no evidence of disease after the interruption of trastuzumab therapy. Discontinuation of maintenance trastuzumab in this patient population after a limited time should be explored cautiously while awaiting a global collaborative effort for a randomized trial.

Legal entity responsible for the study

Japan

Funding

None

Disclosure

A. Shimomura: Research funds from AstraZeneca. J. Watanabe: Honoraria from AstraZeneca Japan, Chugai Pharmaceuticals, Eisai, Novartis Pharma Japan, Taiho pharmaceutical. Advisory board member of AstraZeneca Japan, Eisai. A. Matsui: Lecture Fee: Chugai, Daiichi Sankyo, Eisai, AstraZeneca, Kyowa Kirin Pharmaceutical. Research Funds: Chugai, Daiichi Sankyo, Eisai, Takeda Pharmaceutical, Taiho Pharmaceutical. T. Shien: Lecture fee from AstraZeneca, Novartis, Eizai, Chugai. K. Tamura: Research funds from AstraZeneca, Daiichi Sankyo, Pfizer, MSD. S. Saji: Honoraria from AstraZeneca, Chugai Pharmaceutical, Eisai, Novartis Pharma. Research funds from AstraZeneca and Chugai Pharmaceutical. N. Masuda: Research funds from Chugai and Eisai. Honorarium from Chugai and AstraZeneca. Y. Tokuda: Lecture fee: Pfizer, AstraZeneca, Novartis, Galderma, Kyowa Hakko Kirin. Manuscript fee: Kyowa Hakko Kirin, Daiichi Sankyo. Research funds: Taiho, Eisai, Nippon Kayaku, Chugai, Kyowa Hakko Kirin, Takeda, Novartis, CSPOR. Consulting fee: Meiji. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

304P - Characterization of breast cancer responses to metformin (ID 4225)

Presentation Number
304P
Lecture Time
13:15 - 13:15
Speakers
  • A. Makhtar
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Over the last six decades, metformin has become one of the most widely prescribed oral medications for type II diabetes. It has recently received considerable attention because of its potential role in reducing the risk of cancer development and its antineoplastic properties. However, the mechanism behind the growth-inhibitory effect of metformin on breast cancer cells remains unclear, with little consensus on which tumour subtypes benefit from treatment. Furthermore, it should be noted that much of the in vitro work published to date has used drug concentrations greatly exceeding the recommended clinical dose, and therefore may not translate directly into clinical practice.

Methods

Non-tumorigenic (MCF10A), pre-malignant (MCF10AT), pre-invasive (DCIS), the three-invasive breast cancer [MCF7, T47D and MDA-MB-231] and the fully bone-homed variant of MDA-MB-231(BM) were treated with metformin and effects on cellular proliferation were evaluated by trypan-blue exclusion and clonogenic assays. The expression levels of metformin transporters mRNA and proteins (OCT1-3, MATE1-2 and PMAT) were evaluated by qRT-PCR, western blot.

Results

The growth-curtailing effect of metformin was achieved at 0.3mM for MDA-MB-231(P = 0.0198) in cell counting assays. Colony-forming capacity was inhibited in all cell lines tested at 0.03mM, (P < 0.0001). The Invasive cancer cells demonstrated strong expression of OCT2, PMAT and MATE1 but minimal positivity for OCT1 and no expression of MATE2. OCT3 is only expressed by the triple negative MDA-MB-231. The mRNA and tissue expression of metformin transporters followed the same pattern.

Conclusions

Clinically relevant doses of metformin inhibited the proliferation and colony formation of different breast cancer subtypes regardless of their receptor status and aggressiveness, including the hard to treat triple negative subtype MDA-MB-231 cells. Expression of various influx and efflux metformin-transporters is essential for cellular response and sensitivity to treatment.

Legal entity responsible for the study

Libyan higher ministry of education

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

305P - Paclitaxel every-3-weeks versus weekly paclitaxel and versus weekly vinorelbine in metastatic breast cancer (ID 690)

Presentation Number
305P
Lecture Time
13:15 - 13:15
Speakers
  • L. Katselashvili
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Single-agent chemotherapy (CT) is widely used in the management of HER2-negative breast cancer patients (pts). As both Paclitaxel (P) and Vinorelbine (V) have demonstrated efficacy in the treatment of Metastatic Breast Cancer (MBC), they are recommended among the standard available CT agents for MBC patients. This study compares the efficacy and safety profile of most frequently used three treatment regimens: Paclitaxel every-3-weeks (3-w-P) versus weekly Paclitaxel(w-P) and versus weekly Vinorelbine (w-V) in MBC. Primary objective: Time to progression (TTP). Secondary objectives: evaluation of safety profiles, clinical benefit and response rate (RR) of all arms.

Methods

In this open-label randomized prospective study, pts were randomized (2:2:1) to receive either: intravenously 3-w-P every 21 days, w-P 80 mg/m2/week (day 1, 8, 15) every 28 days or w-V 25 mg/m2/week (day 1, 8, 15) every 28 days. Main eligibility criteria: age ≥18 years, documented metastatic disease previously untreated by CT for metastatic setting, ER/PR positive and HER2-negative disease, or triple negative disease. ECOG≤2.

Results

From April 2014 to April 2015, 95 pts were included. 39 received 3-w-P; 38 received w-P and 18 received w-V per protocol. Median age was 58 years (range 38-79), median duration of treatment 11.5 weeks (range 9-24). Efficacy: with a median follow up of 24 months (m), median time to progression (primary endpoint) was 10.3m, 9.8m and 9.6m in 3-w-P arm, w-P and in w-V arm respectively (p = 0.006). Safety: w-V was much better tolerated with fewer G 3/4 toxicity events (n = 2) than w-P and 3-w-P (n = 23 and 16). Neuropathy G3/4 was mostly reported in 3-w-P and w-P arm than in V arm (75% vs. 69% vs. 17%). G3/4 alopecia was reported in both P arms (94%) when in V arm G3 alopecia was only in 6% of pts.

Conclusions

Weekly Paclitaxel appeared as effective as every-3-weekly regimen and weekly Vinorelbine, however neurotoxicity is a treatment-limiting toxicity for both Paclitaxel regimen. Vinorelbine had fewer significant grade 3-4 toxicities than both Paclitaxel arms and had better RR. Larger randomised studies are needed to determine the efficacy and overall survival of Paclitaxel versus Vinorelbine.

Legal entity responsible for the study

Lika Katselashvili

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

306P - All oral combination of vinorelbine and capecitabine as a first line treatment in patients (pts) with metastatic breast cancer (MBC) (ID 2859)

Presentation Number
306P
Lecture Time
13:15 - 13:15
Speakers
  • S. Shehata
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Oral chemotherapy (OCT) represents a step forward in the management of MBC. It has gained an increased importance over the past years. In Egypt, cancer pts living in rural areas are often hours away from the closest treatment center. For these pts, OCT offers a convenient option and seems to be preferred. In first line MBC, oral vinorelbine (OV) with capecitabine (C) is an active full oral combination with response rates (RR) ranging from 48 to 70% in published data. Based on that, we evaluated efficacy and safety of OV-C in first line Her2 negative MBC pts.

Methods

26 patients were treated. Eligible pts had no previous treatment for their advanced disease. All pts had measurable disease relapsing after (neo) adjuvant AC ± taxane based treatment, WHO PS ≤ 2. Pts were treated with OV 60 mg/m2 D1, D8 for the first cycle and thereafter 80mg/m2 D1, D8 in combination with (C) 825mg/m2 twice daily from D1 to D14, every 21 days for 6 cycles. Primary endpoint (EP) was Disease Progression Rate (DPR) (%); secondary EPs were RR, 3 year survival (3YS) and safety.

Results

All 26 pts were included in the analysis. Median age was 53.2 years (range 38.8-77.1); median WHO PS 1 (range 0-2). 58% of the pts were post-menopausal. All pts were treated with AC-based therapy in the (neo)adjuvant setting and 61.5% with an AC+taxane based treatment. 21 (81%) pts had 2 or more metastatic sites; liver (39%), bone (31%) and lung (31%) being the most frequent sites. A median of 4 cycles were given (range: 1-6) with a total number of 102 cycles delivered. ORR was achieved in 14 pts (54%), including 1 complete (4%) and 13 partial responses (50%). In pts who received 6 cycles of treatment, DPR was 40%, while 3 YS was 64.3%. G3-4 neutropenia was noted in 2 (8%) of pts. G3 hand-foot syndrome, nausea-vomiting and neuropathy were seen in 1 (4%), 2 (8%) and 1 (4%) respectively. Dose escalation was possible in 83% of the pts.

Conclusions

In addition to all the benefits of OCT including convenience and prolonged infusion-free survival, our results show that OV-C is also an effective and well tolerated regimen, making it an attractive option for our pts. OCT appears to be a valid alternative to I.V treatment especially for pts and countries where accessibility to treatment centers remains an issue.

Legal entity responsible for the study

Samir Shehata

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

307P - Retrospective observational study to evaluate the use of halaven plus trastuzumab for the treatment of HER2(+) metastatic breast cancer (MBC) in Spain: HALATRUST study (ID 3000)

Presentation Number
307P
Lecture Time
13:15 - 13:15
Speakers
  • M. Echarri González
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Eribulin is a widely used drug for the management of HER2(-) MBC. Given HER2(+) MBC is routinely treated with combinations of anti-HER2 agents plus chemotherapy, frequent use of eribulin in this context has been published in small case series. As today, clinical trial data of eribulin plus trastuzumab comes from phase 2 study in which 1st line patients achieved objective response rates (ORR) of 71% and median progression free survival (PFS) of 12 months, with median overall survival (OS) not yet achieved. HALATRUST is a retrospective, multicentre study designed to evaluate the clinical benefit rate (CBR) and safety profile of this combination in daily clinical practice.

Methods

All known HER2+ MBC patients treated with eribulin plus trastuzumab for a minimum of 1 dose and whose medical records were available and initiated from April 2011 to May 2016 were included. No statistical hypothesis was pre-established.

Results

Of the 48 patients identified in 19 Spanish public and private centres, 47 fully complied with the inclusion criteria and were analysed for efficacy and safety. Relevant baseline characteristics of the population are as follows. Median age: 55 (35 – 86) year-old, women: 45 (96%), positive estrogen and progesterone receptor: 30 (64%) and 19 (40%) respectively, disease burden of ≥ 5 lesions: 35 (75%), and median number of previous treatments for MBC: 7. Efficacy and safety results of the study population (n = 47) are summarized in the Table.

307P

CategoryVariable
CBR, n (%)25 (53.2)
ORR, n (%)11 (23.4)*
PFS, median (range)3.6 (3.2 – 4.9)
OS, median (range)12.1 (9.6 – 19.1)
Dose adjustments, n (%)9 (19)
TRAE withdrawals, n (%)3 (6)**

TRAE: treatment related adverse events.

Including 1 complete response.

Most TRAE were of hematologic origin.

Conclusions

HALATRUST is, to the best of our knowledge, the largest reported case series on the use of eribulin plus trastuzumab for the treatment of late-line HER2(+) MBC. Results found provide clear evidence on the efficacy and safety of the combination, and highlight the need for its inclusion within the earlier treatment options used in this patient population.

Clinical trial identification

EIS-ERI-2016-01

Legal entity responsible for the study

Eisai Pharmaceuticals Spain

Funding

Eisai Pharmaceuticals Spain

Disclosure

L. Orcajo Rincon, J. Rodríguez-Villanueva: Employee of Eisai Pharmaceuticals. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

308P - Lower response to T-DM1 in metastatic breast cancer patients with HER2 IHC score of 2 and FISH positive compared with IHC score of 3 (ID 3356)

Presentation Number
308P
Lecture Time
13:15 - 13:15
Speakers
  • S. Yazaki
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Ado-trastuzumab emtansine (T-DM1) is the standard second line chemotherapy for HER2 overexpressed metastatic breast cancer (MBC). Tumor HER2 status is measured by either immunohistochemistry (IHC) or fluorescence in situ hybridization (FISH). A previous study showed no difference in objective response rate (ORR) with trastuzumab monotherapy between IHC 3+ and IHC 2+/FISH positive groups. It is not known whether response to T-DM-1 differs between IHC 3+ and IHC 2+/FISH positive patients. The aim of this study is to compare the efficacy of T-DM1 in IHC 3+ group to that of IHC 2+/FISH positive group.

Methods

We retrospectively identified and reviewed the medical records of all patients with HER2 positive MBC who received T-DM1 in our hospital from October 2013 to December 2016. In the efficacy analysis, we excluded five patients who had HER2 negative tumors at metastatic sites.

Results

A total of 44 patients were identified and 36 patients were available for efficacy analysis of ORR. Median age was 58 years old (range 28-80). 95.5% received prior trastuzumab. 45.5% received at least one chemotherapy for MBC, 29.5% received more than four lines of chemotherapy. 79.5% had IHC 3+ and 20.5% had IHC 2+/FISH positive. ORR was 16/30 (53.3%) in IHC 3+ group and was 0/6 (0%) in IHC 2+/FISH positive group (P = 0.024). Median progression free survival (PFS) was 7.0 months (95% CI, 5.58 to 8.42) in IHC 3+ group and was 2.0 months (95% CI, 0.00-4.57) in IHC 2+/FISH positive group.

Conclusions

ORR and PFS were significantly worse in HER2 IHC 2+/FISH positive patients compared with IHC 3+ patients. This is the first report to demonstrate the difference of T-DM1 efficacy by HER2 test results.

Legal entity responsible for the study

St. Luke's International Hospital

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

309P - Nab-paclitaxel (Nab-P) in HER2-ve advanced breast cancer (ABC) patients (pts): From randomized trials to real-life setting: Results from GIM13 - AMBRA study (ID 3388)

Presentation Number
309P
Lecture Time
13:15 - 13:15
Speakers
  • G. Mustacchi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Two randomized studies demonstrated that Nab-P produces a significantly higher overall response rate (ORR), longer Time to Progression (TTP), and greater overall survival (OS) in ABC pts treated with second-line or greater therapy compared with patients who receive conventional Paclitaxel. However, few data are available in the real-life setting, especially for the weekly schedule (wNab-P).

Methods

AMBRA is a longitudinal cohort study, aiming to describe the choice of first and subsequent lines of treatment in HER2-ve ABC pts receiving at least one CHT (SABCS 2016, P5-15-07 & P5-14-09) in the years 2012-2015. For the present analysis, we focused on the use of Nab-P, describing efficacy results according to pts’ characteristics.

Results

So far, 791/1500 pts have been registered into the study and 107 (13.5%) received Nab-P in any line of treatment. Median age was 54 years, 88 (82.2%) had Luminal tumours. Twenty-two pts (20.6%) received Nab-P as 1st line, 48 (44.8%) as 2nd-line, the remaining as 3rd-line or greater. Most pts (47.7%) received the every 3 weeks (Q21) schedule, whereas 30 pts (28%) were treated with the weekly (wNab-P) schedule (days 1,8,15 Q28) at different doses: £100 mg/mq: 11 (10.3%), 125 mg/mq,: 15 (14%); 150 mg/mq: 4 (3.7%). The remaining received different schedules or doses. Median number of cycles received was 5 (1-17) and median duration of treatment was 3.5 months in the whole population. No difference has been observed in terms of number of cycles or duration of treatment according to the schedule.

Conclusions

Our results are similar to those obtained in randomized clinical trials and in a recent large real-life study, confirming that Nab-P is currently one of the most promising choice of treatment for ABC pts.

Legal entity responsible for the study

Marina Elena Cazzaniga

Funding

GIM - Gruppo Italiano Mammella

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

310P - Everolimus-exemestane (EE) vs palbociclib-letrozole (PL) or palbociclib-fulvestrant (PF) in the treatment of metastatic HR+, HER2- breast cancer. An indirect comparison with network meta-analysis (ID 4568)

Presentation Number
310P
Lecture Time
13:15 - 13:15
Speakers
  • C. Cherubini
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

To compare the efficacy of EE to PF or PL in the treatment of metastatic HR+, HER2- breast cancer pre-treated or untreated with aromatase-inhibitors (AI) for advanced disease.

Methods

An indirect comparison with a network meta-analysis comparing EE with PL or PF in the treatment of metastatic HR+, HER2- breast cancer pre-treated or untreated with AI for advanced disease was performed. The Progression-Free-Survival (PFS) was the primary end point of all our indirect comparisons. The indirect comparison was performed both for patients pre-treated with AI and for patients never treated with AI for advanced disease. Efficacy data were expressed as Hazard Ratio (HR) and 95% Confidence Interval (95CI), assuming an α-error of 5% as index of statistical significance.

Results

All the data of the BOLERO-2 trial, the Bachelot et al network meta-analysis (Breast Cancer Treat Rep 2014), the PALOMA-2 and the Paloma-3 trial were analyzed and indirectly compared in a network meta-analysis. 2 orders of comparison were performed: EE vs PL for patients never treated with AI for advanced disease and EE vs PF for patients pre-treated with AI for advanced disease. The pooled HR and 95%CI were respectively 0.597 (0.355-1.005, p = 0.89) and 1.1 (0.7-1.6, p = 0.97) for EE vs PL (never treated with AI) and EE vs PF (pre-treated with AI). No major reasons of clinical and methodological heterogeneity were detected in an independent qualitative analysis, while a moderate quantitative heterogeneity was detected using the I2 test.

Conclusions

Till today EE and PL or PF represent active treatments for patients with metastatic HR+, HER2- breast cancer treated or untreated with AI, and no direct comparisons between EE and PL or PF exist in literature. Although our data have not the power to detect any definitive difference in PFS between EE and PL or PF (probably with the exception of EE vs PL, where a trend in favor of EE could bedetected), EE, PL or PF seem to be comparable in terms of PFS; it follows that the better safety or the economic profile could help physicians in daily clinical practice.

Legal entity responsible for the study

Davide Tassinari

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

311TiP - FRIEND: A randomized pilot study to compare the efficacy and tolerability of fulvestrant 500mg with exemestane as first line endocrine therapy for post-M ER positive HER2 negative ABC patients relapse after adjuvant non-steroidal aromatase inhibitors (NSAI) (ID 881)

Presentation Number
311TiP
Lecture Time
13:15 - 13:15
Speakers
  • J. Wang
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Breast cancer is one of the most common malignancies in women. It has long been acknowledged that oestrogen acts as an endocrine growth factor for hormone-dependent breast cancer. Fulvestrant is a selective estrogen receptor degrader – an ER antagonist with a novel mode of action. Confirm & China Confirm study demonstrated that the efficacy of Fulvestrant 500mg is superior to 250mg. FIRST&FALCON study results confirmed the superior efficacy of fulvestrant over anastrozole in postmenopausal women who have not received prior hormonal therapy. But in the clinical practice, AI are widely used as adjuvant ET for postmenopausal ER+ breast cancer patients. To date there are no randomized trials to compare Fulvestrant 500mg with AI in patients who have relapsed during or after adjuvant non-steroidal AI.

Trial design

The FRIEND trial is a parallel-group, multi-centre study designed to compare the efficacy and tolerability of fulvestrant 500 mg with exemestane 25 mg as first line endocrine therapy in post-M women with ER positive HER2 negative ABC who have relapsed on or after at least 2 years of adjuvant NSAI therapy. Approximately 148 postmenopausal women with ER positive HER2 negative advanced breast cancer who have relapsed whilst on adjuvant NSAI (treatment duration ≥2 years) or after completed adjuvant NSAI treatment will enter this study. Eligible patients will be randomized 1:1 to the following treatment groups: Fulvestrant 500 mg i.m. every 28 (± 3) days plus an additional 500 mg on day 15 (± 3) of first month only; Exemestane 25 mg, orally, once daily. Treatment will continue until disease progression or treatment discontinuation. The primary endpoint is progression-free survival. Secondary endpoints include objective response rate, disease control rate, time to treatment failure, duration of response and overall survival. Efficacy will be determined based on tumor assessments performed by each investigator according to RECIST version 1.1. Safety will be monitored based on the frequency and severity of adverse events (AEs). This study is currently recruiting patients.

Clinical trial identification

NCT02646735

Legal entity responsible for the study

NA

Funding

AstraZeneca China

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

312TiP - VinoMetro: Phase II study of metronomic daily oral vinorelbine as first-line chemotherapy in advanced/metastatic hormone receptor positive (HR+)/human epidermal growth factor receptor 2 negative (HER2-) breast cancer resistant to endocrine therapy (ID 1282)

Presentation Number
312TiP
Lecture Time
13:15 - 13:15
Speakers
  • T. Elger
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Chemotherapy (CTx) is a cornerstone in HR+/HER2- advanced/metastatic breast cancer (a/mBC) after endocrine failure. In this indication, vinorelbine (VRL) is a well-established cytotoxic drug. There is a high medical need for new options that prolong the time between endocrine failure and intensive CTx, which is commonly associated with impaired quality of life and serious side effects. Metronomic CTx was shown to induce disease control in a/mBC with a favorable safety profile. This innovative approach involving continuous daily dosing of oral VRL, which could provide anti-angiogenic and immune-modulatory properties, has not been investigated so far in this indication.

Trial design

VinoMetro is an open-label, single-arm, phase II study (Simon two-stage minimax) of metronomic daily oral VRL (30 mg/day) as first-line CTx. The study involves strict safety monitoring with an initial safety run-in. It is accompanied by a steering committee and supervised by an independent monitoring board. The main objectives are to estimate efficacy in terms of clinical benefit rate after 24 weeks of treatment (primary endpoint) and the progression-free survival, amongst others, as well as the assessment of safety and quality of life. Patients with HR+/HER2- a/mBC having failed or being no candidate for endocrine therapy (targeted combinations allowed) and being naïve to palliative CTx are eligible, if they exhibit ECOG 0-1. The main exclusion criteria are prior vinca-alkaloids, aggressive disease requiring combination CTx and CNS involvement. Until 2017-04-30, 5 patients were enrolled. It is planned to include 45 (39 evaluable) patients at 8 German sites until 09/2018. Scheduled completion date is 09/2019. Two interim analyses are planned (first analysis: safety evaluation based on the 10 initial patients with predefined stopping rules). VinoMetro is an investigator initiated trial (NCT03007992) sponsored by the University Medical Centre of Johannes Gutenberg-University Mainz, Germany, and supported by an unrestricted grant provided by Pierre Fabre Pharma GmbH (Freiburg, Germany).

Clinical trial identification

EudraCT 2016-000284-17

Legal entity responsible for the study

University Medical Centre of Johannes Gutenberg-University Mainz, Germany

Funding

Pierre Fabre Pharma GmbH, Freiburg, Germany

Disclosure

T. Elger, M. Seehase, L. Schollenberger, C. Ruckes, M. Schmidt: Unrestricted study grant for VinoMetro provided by Pierre Fabre Pharma GmbH (Freiburg, Germany). All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

313TiP - Open-label phase II study of everolimus plus endocrine therapy in post-menopausal women with ER+, HER2- metastatic breast cancer (Chloe trial) (ID 1804)

Presentation Number
313TiP
Lecture Time
13:15 - 13:15
Speakers
  • T. Shien
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

BOLERO-2 trial demonstrated that the mTOR inhibitor everolimus was effective in overcoming resistance to endocrine therapy, and BOLERO-4 trial is evaluating the efficacy and safety of combined use of everolimus with letrozole as initial therapy for ER positive HER2 negative metastatic breast cancer (MBC). However, there is no study to evaluate the efficacy of everolimus which can prolong the administration period of aromatase inhibitor (AI) through postponing the acquisition of drug-resistance for MBC with sensitivity to aromatase inhibitor.

Trial design

This study is conducted to examine whether additional administration of everolimus significantly prolongs progression-free survival period in post-menopausal patients with ER-positive HER2-negative MBC which have sensitivity to AI. The inclusion criteria are MBC pts with histologically confirmed ER positive and HER2 negative invasive breast cancer with one or more measurable distant metastatic lesions diagnosed by radiological examination. All patients are receiving AI as the first line hormone therapy for 5-7 months. The pts who are sensitive to AI are randomized to everolimus plus AI arm or the AI alone arm. After randomization, the same AI are continued until progression of diseases and next appropriate regimens are started after that. The primary endpoint is the progression free survival, and the secondary endpoints are overall survival, response rate, disease control rate, adverse events, time to treatment failure and the proportion of patients who continued administration of AI agents for 1 year after the randomized allocation. Sample size for randomized pts was determined to attain at least 80% of power to detect a 5.4 months’ difference (10 vs. 15.4 months, HR:0.65) with one-sided alpha of 0.1. Enrollment of 130 pts for randomization is planned over a 2-year accrual period from April 2017.

Clinical trial identification

This trial was registered at UMIN-CTR[umin.ac.jp/ctr/] as UMIN 000025156.

Legal entity responsible for the study

Comprehensive Support Project for Oncological Research of Breast Cancer

Funding

Novartis

Disclosure

T. Toyama: Research funds from Novartis, Kyowa Hakko Kirin, Diichi Sankyo, Ezai, Chugai, Hippon Kayaku and Takeda Co. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

314TiP - Selecting patients with oligo-metastatic breast cancer harboring homologous recombination deficiency (HRD) for intensified chemotherapy: The OLIGO-study (ID 1947)

Presentation Number
314TiP
Lecture Time
13:15 - 13:15
Speakers
  • T. Steenbruggen
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

About 5% of patients with metastatic breast cancer (MBC) survive more than 10 years. Long-term survival is mostly seen in patients with limited, maximum of 3-5, distant metastases, often referred to as 'oligo’-MBC. Oligo-metastatic cancer can be treated with curative intent using a multidisciplinary approach that targets the detected metastases, circulating micro-metastases, and any locoregional disease if present. Optimal patient selection is of vital importance.

Intensified chemotherapy in the treatment of breast cancer is controversial, as older studies have not shown a survival benefit in unselected patients. Recent retrospective analyses, however, have suggested that patients with HRD derive significant benefit from intensified chemotherapy compared to conventional chemotherapy.

Trial design

This study will evaluate the difference in event-free survival (EFS) between intensified chemotherapy and conventional chemotherapy as part of a multimodality treatment approach in patients with oligo-MBC harboring HRD. Patients are eligible if they have pathological proven oligo-MBC, defined as 1-3 distant metastases, either as de novo or recurrence for which no chemotherapy is given. All lesions must be amenable to surgery or radiotherapy with curative intent. No progression on induction chemotherapy is allowed. Lastly, the tumor has to be HRD by array comparative genomic hybridization. Patients start with 3 cycles of induction chemotherapy, which includes anthracyclines and taxanes in treatment-naïve patients and is adapted according to previously received (neo)adjuvant treatment in others. Patients are 1:1 randomized to another 3 cycles of conventional chemotherapy or 2 cycles of intensified chemotherapy (carboplatin, thiotepa and cyclophosphamide) with stem cell support. Following systemic treatment, all patients receive maximal surgery and/or radiotherapy of locoregional and distant disease. The primary endpoint is EFS at 3 years. Toxicity, time to progression, and overall survival are secondary clinical endpoints. In total 86 patients are required. At the time of abstract submission, 33 patients were randomized.

Clinical trial identification

NCT01646034

Legal entity responsible for the study

The Netherlands Cancer Institute

Funding

Dutch Cancer Society (KWF)

Disclosure

S.C. Linn: Grants and non-financial support from AstraZeneca, Roche, Genentech, Cergentis. Advisory support from Novartis, PhilipsHealth and IBM outside the submitted work. A BRCA-like signature-patent (WO/2015/080585 and PCT/NL2014/050813) is pending. G.S. Sonke: Institutional research support funding from Roche, AstraZeneca, Merck and Novartis. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

315TiP - AGATA molecular screening program: Implementing precision medicine in patients with advanced breast cancer in Spain (ID 2712)

Presentation Number
315TiP
Lecture Time
13:15 - 13:15
Speakers
  • S. Pernas Simon
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Metastatic breast cancer continues to be a major cause of cancer death among women globally. In recent years, a better understanding of tumor biology, and the availability of high-throughput technologies has enabled the emergence of precision medicine bringing new expectations and giving rise to molecular screening programs worldwide. Recently, the MOSCATO trial has shown for the very first time that prospectively sequencing a large panel of genes and utilizing this information to guide treatment choices may improve the outcome of a subset of patients. Some institutions are implementing such strategy as part of the routine treatment decision-making process. However, SOLTI, as a collaborative Spanish network, runs AGATA, the first multi-institutional molecular screening program ever implemented in this country. Patient recruitment started in October 2014 and is expected to conclude in June 2017.

Trial design

Up to 260 patients with metastatic breast cancer will be recruited in 10 participating sites in Spain. Mutation testing is performed prospectively in the genomic laboratories of Vall d´Hebron Institute of Oncology in Barcelona, 12 de Octubre University Hospital in Madrid, and the University Clinical Hospital of Valencia. Upon molecular characterization and collection of key clinical data, each case is reviewed by a multidisciplinary advisory board, which recommends potential experimental treatments, mainly in the context of clinical trials. During this pilot stage, our primary objective is to determine the success rate in including patients in trials based on their molecular profile. Additional aims are to identify technical and logistical barriers to the implementation of a nationwide program, describe the genomic profiles of the tumors, and assess patient outcomes. Retrospective gene expression (PAM50 + 110 genes and 20 miRNAs) and proteomic analysis (40 markers) will be performed to provide a more comprehensive molecular profile of the tumors that may help explain sensitivity or resistance to administered therapies. Data collected within this program is expected to generate hypotheses for further investigations directed to improve precision medicine.

Clinical trial identification

NCT02445482

Legal entity responsible for the study

SOLTI Breast Cancer Research Group

Funding

Novartis, Mutua Madrileña, Instituto de Salud Carlos III

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

316TiP - BREAKOUT: A cross-sectional, prospective, observational study of germline BRCA mutation (gBRCAm) prevalence and real-world outcomes among patients (pts) with HER2-negative (HER2-ve) metastatic breast cancer (mBC) (ID 3131)

Presentation Number
316TiP
Lecture Time
13:15 - 13:15
Speakers
  • T. Dalvi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Several poly (ADP-ribose) polymerase (PARP) inhibitor therapies are currently under investigation to assess their potential for the management of breast cancer. Olaparib (Lynparza) is being evaluated in a Phase III study in HER2-ve mBC pts with a gBRCAm (OlympiAD; D0819C00003; NCT02000622). The prevalence of germline and somatic BRCA mutations or other somatic homologous recombination repair gene mutations (HRRm) in HER2-ve mBC pts is not well defined. Treatment and clinical outcomes data for pts with HRRm are also limited. The primary objective of this real-world evidence study (NCT03078036) is to estimate gBRCAm prevalence in a population of HER2-ve mBC pts not selected by phenotype, age or family history. Treatment patterns and survival outcomes for pts with HRRm will also be examined.

Trial design

BREAKOUT is a prospective, cross-sectional, non-interventional, observational cohort study recruiting HER2-ve mBC pts who have started first-line chemotherapy within 90 days prior to enrollment. If hormone receptor positive, eligible pts will have exhausted all hormone therapy options prior to enrollment and will not have received prior PARP inhibitor treatment. If unavailable from medical records, gBRCAm status will be locally determined by blood test and, if negative, archival tumor specimens may be examined with a FoundationOne Dx genomic profile. Pts with a gBRCAm, somatic BRCA mutation or other HRRm (in ATM, RAD51B, RAD51C, RAD51D, RAD54L, BRIP1, FANCL, PALB2, BARD1, CHEK1, CHEK2, CDK12 or PPP2R2A) will be followed for ≥18 months to determine the distribution of standard-of-care treatments, progression-free survival (investigator assessed) and overall survival by line of therapy. Approximately 2000 women in 17 countries in North America, the EU and the Asia Pacific region will be enrolled to provide a global gBRCAm prevalence estimate with a precision of no more than ±2%. An interim analysis focusing on gBRCAm prevalence will be carried out once testing has been completed for all pts. The study is expected to conclude in 2019.

Clinical trial identification

NCT03078036, June 2020

Legal entity responsible for the study

AstraZeneca

Funding

AstraZeneca

Disclosure

T. Dalvi: Employee of MedImmune and AstraZeneca, stock ownership in AstraZeneca, Bristol-Myers Squibb, Roche, Achillion, Medivation, Inovio and Gilead. K.A. Gelmon: Consulting fees from Novartis, Pfizer, AstraZeneca, Merck, Genentech and Nanostring. R. Dent: Honoraria and consulting fees from AstraZeneca, Merck, Pfizer, Roche and Eisai, and travel and accommodation expenses from Pfizer. M. Pegram: Received consulting fees from Pfizer, Genentech and Amgen. Y. Tazir, A. Milner, J. Lewis: Employee of AstraZeneca. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

317TiP - IMpassion131: Phase III study comparing 1L atezolizumab with paclitaxel vs placebo with paclitaxel in treatment-naive patients with inoperable locally advanced or metastatic triple negative breast cancer (mTNBC) (ID 3851)

Presentation Number
317TiP
Lecture Time
13:15 - 13:15
Speakers
  • D. Miles
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Chemotherapy (including paclitaxel [pac]) remains the main 1L treatment for metastatic TNBC but brings limited clinical benefit, highlighting the need for new treatments. Atezolizumab (atezo) blocks the interaction of PD-L1 with receptors PD-1 and B7.1, thus restoring anti-tumor immunity. TNBC is a rational target for atezo due to high PD-L1 expression, elevated T-cell tumor infiltration and high mutational burden. Atezo alone and in combination with nab-pac was well tolerated, with no exacerbation of chemo-associated adverse events, and demonstrated promising clinical activity in mTNBC. Atezo + nab-pac is being further investigated as 1L TNBC treatment in IMpassion130. IMpassion131, a global, multi-center, randomized, double-blind, placebo (pbo)-controlled study, is comparing the efficacy and safety of 1L atezo + pac vs pbo + pac in patients (pts) with untreated, inoperable, locally advanced or metastatic TNBC.

Trial design

Eligible pts are those with inoperable, locally advanced or metastatic TNBC, histologically confirmed; de novo or recurrent disease after early BC chemo treatment completed ≥ 12 mo prior; taxane monotherapy eligible; no prior chemo or targeted systemic therapy for inoperable locally advanced or metastatic disease; ECOG PS 0-1 and measurable disease by RECIST v1.1. Exclusion criteria include known symptomatic CNS disease, prior immunotherapy and history of autoimmune disease. Approximately 495 pts will be randomized 2:1 to receive atezo (840 mg) or pbo (q2w; days 1 and 15 of 28-day cycle) plus pac (90 mg/m2; days 1, 8, 15 of 28-day cycle) until disease progression. Stratification factors are PD-L1 expression on tumor-infiltrating immune cells (IC; IC0 [< 1%] vs IC1/2/3 [≥ 1%] with VENTANA SP142 IHC assay), prior taxane therapy, presence of liver metastases and geographical region. The primary endpoint is PFS measured by RECIST v1.1. Key secondary endpoints include OS, 12- and 18-month OS rates, 12-month PFS rate, ORR, DOR, and safety. Tumor biopsies will be investigated at baseline, on treatment and at progression to assess biomarkers of response and immune escape.

Clinical trial identification

NCT03125902

Legal entity responsible for the study

F. Hoffmann-La Roche Ltd.

Funding

F. Hoffmann-La Roche Ltd.

Disclosure

D. Miles: Honoraria for Advisory Boards from Roche-Genentech. F. Andre: Research grants: AstraZeneca, Novartis, PF, Lilly. J. Gligorov: Advisory boards: Eisai, GenomicHealth, Novartis, Pfizer, Roche. Granted research: Eisai, Roche. D. Cameron: Employer paid for consultancy work done with Roche. Hospital has also been reimbursed for costs incurred in doing clinical research by Roche. C.H. Barrios: Research: Pfizer, Novartis, Amgen, AstraZeneca, Boehringer Ingelheim, Roche, Lilly, Sanofi, GSK, Taiho, Mylan, Merrimack, Merck, Astellas, Bristol-Myers Squibb. Consulting: Boehringer-Ingelheim, GSK, Novartis, Pfizer, Roche-Genentech, Eisai. A. Schneeweiss: Member of Roche advisory boards. Member of IMpassion 130 steering committee. Honoraria and financial support from Roche for presentations at academic meetings. V. Easton, I. Dolado: Roche employee and stock. J. O\'Shaughnessy: Consultant to Genentech and Roche. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

319TiP - CompLEEment-1: Phase 3b study of ribociclib + letrozole for the treatment of hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2–) advanced breast cancer (ABC) in patients with no prior endocrine therapy (ET) for ABC (ID 4272)

Presentation Number
319TiP
Lecture Time
13:15 - 13:15
Speakers
  • M. De Laurentiis
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

CDK4/6 inhibitor ribociclib was recently approved in the United States in combination with letrozole for the treatment of HR+, HER2– ABC in postmenopausal women with no prior therapy for advanced disease, based on the significantly prolonged PFS versus placebo plus letrozole observed in the pivotal phase 3 MONALEESA-2 trial (Hortobagyi et al. NEJM 2016). The phase 3b CompLEEment-1 study will further evaluate the safety and efficacy of ribociclib plus letrozole as first-line therapy in an expanded patient population.

Trial design

In this open-label study, men or women of any menopausal status with HR+, HER2– ABC will receive ribociclib (600 mg/day, 3 weeks on/1 week off) + letrozole (2.5 mg/day); men and premenopausal women will receive concomitant goserelin (3.6 mg subcutaneous implant every 28 days). Treatment will continue until disease progression or unacceptable toxicity. Patients are limited to ≤ 1 line of chemotherapy and no prior ET for advanced disease; patients receiving (neo)adjuvant ET with a nonsteroidal aromatase inhibitor must have a disease-free interval of > 12 months. Exclusion criteria include Eastern Cooperative Oncology Group performance status >2, or prior CDK4/6 inhibitor treatment. Planned hematologic and chemistry laboratory assessments will be completed every 2 weeks for the first 2 months, then monthly to Cycle 6, and as clinically indicated to Cycle 36. Tumor assessments are recommended every 12 weeks or at intervals per local standard of care during the treatment phase. The primary outcome is safety and tolerability. Secondary outcomes include time to progression, clinical benefit rate, overall response rate, safety, and patient-reported outcomes (PROs). Adverse events and drug-drug interactions will be monitored using CT Scholar; PROs will be collected for female patients using the FACT-B questionnaire to better understand health-related quality of life and treatment side effects. Global recruitment of the planned ∼3,000 patients is ongoing, with the majority occurring in Europe.

Clinical trial identification

NCT02941926

Legal entity responsible for the study

Novartis Pharmaceuticals

Funding

Novartis Pharmaceuticals

Disclosure

M. De Laurentiis: Provided consulting or advisory role and participated in a speakers\' bureau for Novartis, Roche, Pfizer, AstraZeneca, Celgene and Eisai. M. Martin Jimenez: Consulting or Advisory role from Roche/Genetech, Novartis, Amgen, Pfizer, Lilly. The institution received funding from Novartis. A. Ring: Consulting or advisory role for Novartis, Roche, and Genomic Health. P. Cottu: Consulting or advisory role and received honoraria from Pfizer, AstraZeneca, Novartis and Roche. Conducted research project funded by Novartis, Pfizer and Genotech. K. Zhou, J. Wu, J.P. Zarate: Employee of Novartis Pharmaceuticals Corporation. C. Zamagni: Consulting, advisory role, travel, accomodations, expense and/or research funding from Roche, Genomic Health, Pierre Fabre, Eisai, Novartis, AstraZeneca, Celgene, Medivation, Abbvie, Pfizer, Array BioPharma, Morphotek.

Collapse
Poster display session Poster Display session

320TiP - A phase II trial of mirvetuximab soravtansine in patients with localized triple-negative breast cancer (TNBC) with tumors predicted insensitive to standard neoadjuvant chemotherapy (NACT) including a lead-in cohort to establish activity in patients with metastatic TNBC (ID 4285)

Presentation Number
320TiP
Lecture Time
13:15 - 13:15
Speakers
  • S. Damodaran
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

While TNBC patients with pCR/RCB-0 or RCB-1 have excellent survival, those with extensive residual disease (RCB-II or RCB-III) after NACT have poor prognosis. At MD Anderson, through the Moonshot Initiative, we have created a biomarker-driven drug development strategy, ARTEMIS (A Randomized, TNBC Enrolling trial to confirm Molecular profiling Improves Survival) to identify novel targeted therapies for tumors that are predicted to be insensitive to standard NACT. Molecular profiling along with imaging is used to identify patients with chemo-insensitive disease and inform second phase of therapy incorporating targeted agents to improve responses. Folate receptor α (FRα) is a GPI-anchored surface protein encoded by FOLR1 gene that is overexpressed in multiple cancers including TNBC. Mirvetuximab soravtansine is an antibody-drug conjugate that consists of a monoclonal antibody against FRα conjugated to maytansinoid, a microtubule inhibitor. Nearly 40% of TNBC express high levels of FRα, suggesting that FRα directed therapy is a viable therapeutic strategy.

Trial design

The study will include a lead in cohort (Cohort A) to establish efficacy in metastatic TNBC patients and a neoadjuvant cohort (Cohort B) to determine activity in chemo-insensitive, localized TNBC patients. If > 2 patients in Cohort A have response, the neoadjuvant cohort will be activated. Patients deemed to have chemo-insensitive, FRα+ TNBC identified through the ARTEMIS are eligible for Cohort B. The primary objectives are to determine the response rate of single agent mirvetuximab in metastatic FRa+ TNBC (> 2 lines of therapy) and to determine if mirvetuximab would improve the rates of neoadjuvant pathologic response (pCR or RCB-I) from 5% to 20% in patients with high risk, chemo-insensitive, FRa+ TNBC. A two-stage Gehan-type design with 14 patients in the first stage will be employed for the neoadjuvant cohort (n = 37). Mirvetuximab will be given IV every 21 days (6 mg/kg). Correlatives include assessment of FOLR1/FRα and characterization of biomarkers of immune modulation.

Clinical trial identification

NCT03106077

Legal entity responsible for the study

M.D. Anderson Cancer Center

Funding

NCCN

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

321TiP - SOLTI-1303 PATRICIA: A phase II study of palbociclib and trastuzumab (with or without letrozole in ER+) in previously trastuzumab-pretreated, postmenopausal patients with HER2-positive metastatic breast cancer (ID 4434)

Presentation Number
321TiP
Lecture Time
13:15 - 13:15
Speakers
  • P. Villagrasa Gonzalez
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Despite the high efficacy of anti-HER2 agents, HER2+ metastatic breast cancer remains incurable and in need of additional therapeutic options. Persistent activation of the cyclin D1/CDK4 axis has been identified as a mediator of resistance to anti-HER2 therapy but clinical data of the benefit of CDK4/6 inhibitors in combination with trastuzumab is lacking. PATRICIA is a Simon 2-Stage study to evaluate the efficacy of combining trastuzumab plus palbociclib, with or without letrozole, assessed by progression-free survival (PFS) in pretreated HER2-positive patients.

Trial design

Postmenopausal HER2-positive patients treated with 2-4 prior systemic anticancer treatment lines that must involve trastuzumab or another anti-HER2 treatment in the metastatic setting are included in three cohorts: A: HR-negative, receiving trastuzumab and palbociclib; B1: HR-positive, receiving trastuzumab and palbociclib; B2: HR-positive, receiving trastuzumab, palbociclib and letrozole. Palbociclib is administered at 200 mg/day for 14 days of 21-day cycles. Trastuzumab and letrozole are administered at usual doses. As these combinations have not been tested in phase I trials, we incorporated a 2 cycles-safety run-in phase with the first 6 patients of each regimen. The primary objective is to assess clinical efficacy measured as PFS at 6 months (PFS6). Assuming an increase of at least 20% in PFS6 by the addition of palbociclib +/- letrozole to trastuzumab, PFS6 should be ≥ 50% for a cohort to be successful and proceed to stage 2. According to this, it will be necessary to include 15 patients in each cohort in stage 1. In stage 2, each cohort may continue recruitment for up to 46 patients. Translational research searching for predictive biomarkers will be implemented. To date, 43 patients, 13 in A and 15 in each B cohort, have been included in 14 sites across Spain. An independent safety data committee was held twice during the study. The committee recommended that study continue enrollment as planned. The first stage efficacy analysis is intended for December 2017.

Clinical trial identification

NCT02448420

Legal entity responsible for the study

SOLTI Breast Cancer Research Group

Funding

Pfizer

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

322TiP - PYTHIA: A phase II study of palbociclib plus fulvestrant for pretreated patients with ER+/HER2- metastatic breast cancer (ID 4560)

Presentation Number
322TiP
Lecture Time
13:15 - 13:15
Speakers
  • D. Zardavas
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Palbociclib is an oral, potent, selective inhibitor of CDK4/6, blocking cell cycle progression from G1 into S phase. Preclinical data indicate that palbociclib has enhanced activity against luminal ER+ human breast cancer (BC) cell lines in vitro, combined with endocrine treatment. Randomized clinical trials showed significant PFS prolongation in patients with newly diagnosed and pretreated metastatic luminal BC, when palbociclib was combined with letrozole and fulvestrant respectively (PALOMA-1/2 and -3 trials). Predictive biomarkers for patient selection to receive palbociclib plus endocrine treatment are still missing.

Trial design

PYTHIA (IBCSG 53-14/BIG 14-04) is a phase II, single-arm, multicenter, study of fulvestrant and palbociclib in postmenopausal women with ER+/HER2-, advanced BC, who progressed after prior endocrine treatment (1st or 2nd line; up to 1 line of prior chemotherapy is allowed). Patients are enrolled concurrently in the AURORA program (NCT02102165), a longitudinal cohort study with extensive molecular characterization of matched primary-metastatic BC, and plasma samples. The primary endpoint is PFS, based on local assessment as per RECIST 1.1. Secondary endpoints are safety and tolerability, as well as disease control rate. Correlative objectives will assess the potential predictive value of: i) mutations and copy number aberrations in a panel of cancer-related genes, ii) gene signatures inferred by RNA sequencing, iii) early FDG-PET/CT assessment performed for a subset of 30 patients, at baseline and Day 28, and iv) a serum thymidine kinase-1 (TK1) assay, performed at baseline, Day 14 and after Cycle 1. The sample size of 120 patients was selected to have 80% power to detect a HR of 2.0 for biomarker-positive patients, with 30-50% prevalence (two-sided α = 0.05). Enrollment opened in May 2016, with the target-recruitment being 120 patients at 21 sites in Belgium, Italy and the UK.

Clinical trial identification

NCT02536742

Legal entity responsible for the study

International Breast Cancer Study Group (IBCSG)

Funding

Pfizer

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

323TiP - EarLEE-1: A phase 3 study of ribociclib + endocrine therapy (ET) for adjuvant treatment of patients (pts) with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2–), high-risk, early breast cancer (EBC) (ID 4834)

Presentation Number
323TiP
Lecture Time
13:15 - 13:15
Speakers
  • M. Martin Jimenez
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Adjuvant ET and chemotherapy reduce the risk for recurrence of HR+, HER2– EBC. However, recurrence is still common in pts with adverse clinical and pathologic features. In the phase 3 clinical trial MONALEESA-2, the cyclin-dependent kinase 4/6 inhibitor ribociclib in combination with letrozole prolonged progression-free survival versus letrozole plus placebo in postmenopausal women with HR+, HER2– advanced breast cancer and no prior therapy for advanced disease (HR = 0.56, 95% CI, 0.43-0.72; P = 3.29 × 10−6; Hortobagyi et al. N Engl J Med. 2016). EarLEE-1 (NCT03078751) investigates the efficacy and safety of ribociclib with ET versus placebo with ET as adjuvant treatment in pts with high-risk EBC.

Trial design

In this double-blind, placebo-controlled, phase 3 trial, ∼2000 women and men with fully resected, high-risk, HR+, HER2– EBC (defined as AJCC 8th ed. Prognostic Stage Group III for pts who received adjuvant chemotherapy, or > 2 mm residual disease in axillary lymph nodes and > 10 mm in breast after neoadjuvant chemotherapy) are being randomized 1:1 to ribociclib (600 mg/day, 3 weeks on/1 week off for ∼24 months) with ET or placebo with ET. Adjuvant ET may include tamoxifen, letrozole, anastrozole, or exemestane for ≥ 60 months with ovarian suppression for premenopausal women. Randomization is stratified by menopausal status, risk group, and region. Eligible pts must have tumor tissue from the surgical specimen, adequate bone marrow and organ functions, normal serum electrolytes, QTc interval < 450 msec, and completed and recovered from acute toxicities of adjuvant radiotherapy and (neo)adjuvant chemotherapy. The primary endpoint is invasive disease-free survival. Secondary endpoints include recurrence-free survival, distant disease-free survival, overall survival, quality of life, and safety. Global recruitment is ongoing.

Clinical trial identification

NCT03078751

Legal entity responsible for the study

Novartis Pharmaceuticals

Funding

Novartis Pharmaceuticals

Disclosure

M. Martin Jimenez: The institution has received research funding from Novartis. T. Bachelot: Grants, personal fees and non-financial support from Roche, grants, personal fees and non-financial support from Novartis, grants and personal fees from AstraZeneca, outside the submitted work. C. Barrios: Grants and/or personal fees: Pfizer, Novartis, Amgen, AstraZeneca, B, GSK, Roche-Genentech, Lilly, Sanofi, Taiho, Mylan, Merrimack, Merck, Abbvie, Astellas, Biomarin, Bristol-Myers Squibb, Daiichi, Abraxis, AB, Asana Bios, Medivation, Exelixis, ImClone, LEO, Millennium, Pfizer, Eisai. K. Blackwell: Grants and/or personal fees: Novartis, Amgen, AstraZeneca, Advaxis, Bayer, Celgene, Coherus, Eisai, Eli Lilly, Genentech, GI Thera, Hospira, MacroGenics, Merck, Pfizer, Pierian Bio, Puma, Roche, Rockwell, Sandoz, Spectrum. M. De Laurentiis: Consulting and/or advisory role and speakers\' bureau from Novartis, Roche, Pfizer, AstraZeneca, Clegene and Eisai. S. Hurvitz: Grants from Amgen, Bayer, BI, Genetech, GSK, Pfizer, Roche, Biomarin, Merrimack, PUMA, Dignitana, Medivation, Lilly, Novartis and OBI Pharma, outside of the submitted work. S. Loi: Grants from Novartis, Merck, Roche-Genentech, Pfizer, during the conduct of the study. P. Schmid: Personal fees from Pfizer, Boehringer Ingelheim, Bayer, Puma, Eisai, Celgene, Roche-Genentech, during the conduct of the study. K. Hazell, S. Mondal, M. Shilkrut, C. Germa: Employee of Novartis Pharmaceuticals Corporation. All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

324TiP - PATINA: A randomized open label phase III trial to evaluate the efficacy and safety of palbociclib + anti HER2 therapy + endocrine therapy vs anti HER2 therapy + endocrine therapy after induction treatment for hormone receptor positive, HER2-positive metastatic breast cancer (ID 4918)

Presentation Number
324TiP
Lecture Time
13:15 - 13:15
Speakers
  • O. Metzger
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Pre-clinical data and initial results from clinical studies point to the added benefit of CDK4/6 inhibition when combined with anti-HER2 tx. The current study is designed to evaluate the added benefit of Palbociclib when given in combination with anti-HER2 and endocrine tx maintenance in the 1st line setting of metastatic HER2+HR+ breast cancer.

Trial design

PATINA is an international, open-label, pivotal Phase III study. Primary objective is to demonstrate that the combination of Palbociclib with anti-HER2 plus endocrine tx is superior to anti-HER2 plus endocrine tx in prolonging PFS. Sample size is 496 pts. The study starts after completion of 6-8 cycles of chemotherapy-containing anti-HER2 tx for metastatic breast cancer in the 1st line setting. Pts are eligible provided they are without evidence of disease progression by local assessment (i.e. CR, PR or SD). To account for the need for less intense tx regimens for a subset of pts diagnosed with HER2+ER+ disease, clinicians may recommend the combination of trastuzumab with either a taxane or vinorelbine prior to study initiation. Clinicians might also choose a non-Pertuzumab option for pts previously treated with pertuzumab in the neo(adjuvant) setting. Secondary objectives include measures of tumor control (OR, CBR, DOR), OS, safety and QOL. The translational science main objective is to compare PFS estimates according to PIK3CA mutation status assessed by cfDNA analysis. Endocrine tx options are AI or fulvestrant. Premenopausal pts must receive ovarian suppression. The study has a 90% power to detect a hazard ratio of 0.667 in favor of the palbociclib arm. Pts approached to participate in AFT-38 will be asked to indicate on the informed consent forms whether remaining biospecimens and clinical data from the control arm of the study can be shared with the Mastering Breast Cancer (MBC) Initiative. The overarching purpose of the MBC is to create a mechanism for understanding the natural history of metastatic breast cancer by cataloguing longitudinally studied tumor-specific markers and treatment effects.

Clinical trial identification

NCT02947685

Legal entity responsible for the study

Alliance Foundation Trials

Funding

Pfizer

Disclosure

C. Huang: Employee Pfizer Inc. M. Khoeler: Employee and shareholder Pfizer Inc. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

DEVELOPMENTAL THERAPEUTICS (ID 5676)

Lecture Time
13:15 - 13:15
Speakers
  • R. Plummer
  • U. Lassen
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15
Poster display session Poster Display session

376P - Preclinical evaluation of the anti-CLDN18.2 antibody, IMAB362, in pancreatic carcinoma (ID 2070)

Presentation Number
376P
Lecture Time
13:15 - 13:15
Speakers
  • C. Heinz
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Claudin 18.2 (CLDN18.2) is a tight junction protein restricted to the gastric mucosa cells; however, in the context of malignant transformation CLDN18.2 can be found in tumors derived from organs that do not normally express CLDN18.2, such as the pancreas. IMAB362 is a first-in-development monoclonal antibody that specifically targets CLDN18.2-expressing tumor cells. The aims of this preclinical study were to examine CLDN18.2 expression in pancreatic cancer (PC) and to evaluate IMAB362 as a single agent or in combination with standard chemotherapy for the treatment of PC.

Methods

CLDN18.2 expression in normal and neoplastic pancreatic tissues from patients with PC was assessed by a validated, semi-quantitative, IHC assay. IMAB362 pharmacodynamics, including binding characteristics, antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and IMAB362-induced antitumor activity, were assayed in vitro (human PC cell lines) and in vivo (PC xenograft mouse models).

Results

CLDN18.2 expression levels and patterns in normal and malignant pancreatic tissue were very different. Whereas CLDN18.2 was not observed in normal tissue, CLDN18.2 was expressed in about 50% of tested neoplastic pancreatic tissues and premalignant lesions. IMAB362 bound with high specificity and high affinity to CLDN18.2-positive (CLDN18.2+) PC cells. IMAB362-induced efficient lysis of CLDN18.2+ PC cells through ADCC. In vitro, pretreatment of human PC cells with chemotherapeutic agents resulted in CLDN18.2 stabilization and a higher epitope density, which led to an increase in IMAB362-induced ADCC. Repeated doses of IV IMAB362 in mice bearing CLDN18.2+ PC xenografts resulted in slowed tumor growth and survival benefit. In vitro studies and animal data showed the antitumor activity of IMAB362 to be further augmented by pretreatment with gemcitabine and gemcitabine plus oxaliplatin.

Conclusions

In these preclinical studies, IMAB362 induced cell death in CLDN18.2+ human PC cell lines and xenografted tumors, and demonstrated potential as a targeted treatment for PC, both as single agent and in combination with chemotherapy.

Legal entity responsible for the study

Ganymed Pharmaceuticals AG, A company of Astellas Pharma, Inc

Funding

Ganymed Pharmaceuticals AG, A company of Astellas Pharma, Inc

Disclosure

C. Heinz: Employee of Ganymed Pharmaceuticals AG. In addition, Dr. Heinz has patents 33PCT 34PCT, and 36PCT issued. R. Mitnacht-Kraus: Employee of Ganymed Pharmaceuticals AG, a company of Astellas Pharma, Inc. In addition, Dr. Mitnacht-Kraus has a patent P-24PCT issued, a patent P-33PCT issued, a patent P-34PCT issued, and a patent P-36PCT issued. M. Kreuzberg, S. Wöll: Employee of Ganymed Pharamceuticals AG, a company of Astellas Pharma, Inc. U. Sahin: Stock option owner, ex-shareholder and cofounder of Ganymed Pharmaceuticals AG and Founder/CEO/shareholder of Biontech Holding outside the submitted work. Dr. Sahin has several patents issued to this work that have been acquired by Astellas. Ö. Türeci: Stock option owner, ex-shareholder, cofounder & CEO of Ganymed Pharmaceuticals AG, has received consultancy fees from Astellas, and has several patents issued to this work that have been acquired by Astellas.

Collapse
Poster display session Poster Display session

377P - Preclinical characterization of IMAB362-vcMMAE, an anti-CLDN18.2 antibody–drug conjugate (ID 2067)

Presentation Number
377P
Lecture Time
13:15 - 13:15
Speakers
  • M. Kreuzberg
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Antibody–drug conjugates combine the specific targeting and antitumor activity of monoclonal antibodies with the potent cell killing activity of cytotoxic small molecule drugs. IMAB362 is a monoclonal antibody specific for the tight junction protein Claudin 18.2 (CLDN18.2). In normal tissue, CLDN18.2 is exclusively expressed in the gastric mucosa. In the context of malignant transformation, CLDN18.2 can be found in gastric tumors as well as tumors from organs that do not normally express CLDN18.2 (eg, pancreas). Preclinical characterization of IMAB362 conjugated to the antimitotic molecule, monomethyl auristatin E, with a valine–citrulline linker (IMAB362–vcMMAE) is presented here.

Methods

IMAB362–vcMMAE binding characteristics and internalization were assessed in CLDN18.2-expressing human cell lines. Cell viability and IMAB362–vcMMAE-mediated cytotoxic effects (direct and indirect [bystander]) were also assessed in CLDN18.2 in vitro models. Xenograft mouse models of pancreatic and gastric cancers were developed to assess the cytotoxic and antitumor effects of IMAB362–vcMMAE in vitro.

Results

IMAB362–vcMMAE showed a slightly decreased relative binding affinity on CLDN18.2-transfected cells and cells that endogenously express CLDN18.2 compared with unconjugated IMAB362. In cell lines that internalized IMAB362–vcMMAE, cell viability was reduced by 45–90%; EC50 negatively correlated with CLDN18.2 expression level, with the lowest EC50 being <30 ng/mL. By contrast, no reduction in cell viability occurred in cells without target expression. IMAB362–vcMMAE produced CLDN18.2-negative cell death via bystander effect in vitro (co-cultured tumor cells). In vivo, intravenous IMAB362–vcMMAE resulted in dose-dependent inhibition of tumor growth as well as prolonged survival in early and advanced tumors in both pancreatic and gastric cancer mouse models; significant antitumor activity was observed after a single 8 or 16 mg/kg IV bolus injection. No systemic or organ-specific IMAB362–vcMMAE-related toxicity was observed in the mice.

Conclusions

IMAB362–vcMMAE is a highly specific and potent antibody-drug conjugate against in vivo and in vitro models of gastric and pancreatic cancers.

Legal entity responsible for the study

Ganymed Pharamaceuticals AG, A company of Astellas Pharma, Inc

Funding

Ganymed Pharamaceuticals AG, A company of Astellas Pharma, Inc

Disclosure

M. Kreuzberg: Emplyee of Ganymed Pharamceuticals AG, a company of Astellas Pharma, Inc. R. Mitnacht-Kraus: Employee of Ganymed Pharmaceuticals AG, a company of Astellas Pharma, Inc. In addition, Dr. Mitnacht-Kraus has a patent P-24PCT issued, a patent P-33PCT issued, a patent P-34PCT issued, and a patent P-36PCT issued. U. Sahin: Stock option owner, ex-shareholder and cofounder of Ganymed Pharmaceuticals AG and Founder/CEO/shareholder of Biontech Holding outside the submitted work. Dr. Sahin has several patents issued to this work that have been acquired by Astellas. Ö. Türeci: Stock option owner, ex-shareholder, cofounder & CEO of Ganymed Pharmaceuticals AG, has received consultancy fees from Astellas, and has several patents issued to this work that have been acquired by Astellas.

Collapse
Poster display session Poster Display session

378P - Preclinical characterization of IMAB362 for the treatment of gastric carcinoma (ID 1962)

Presentation Number
378P
Lecture Time
13:15 - 13:15
Speakers
  • R. Mitnacht-Kraus
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Monoclonal antibodies (mAbs) are a key component of cancer therapy. An ideal therapeutic mAb would selectively bind to cancer cells while avoiding binding to healthy tissue. In normal tissue, Claudin 18.2 (CLDN18.2) is expressed in gastric mucosal cell tight junctions, largely inaccessible to mAbs. Upon malignant transformation, perturbations in cell polarity lead to cell surface exposure of CLDN18.2 making it targetable by mAbs. This has driven the development of an anti-CLDN18.2 mAb, IMAB362, which specifically targets CLDN18.2-positive cancer cells while sparing normal tissue. The preclinical characterization of IMAB362 as a targeted therapy for gastric cancer (GC) is presented here.

Methods

CLDN18.2 expression was characterized in primary tumors and metastases (32 of which corresponded to primary tumors) from GC patients using a validated, semi-quantitative, IHC assay. IMAB362 binding characteristics and mechanism of action were assessed in vitro using CLDN18.2-expressing cell lines and in vivo in mouse tumor xenografts. Antitumor activity of IMAB362 was assessed in human GC cell line xenografts in mice treated with chemotherapy with/without IMAB362.

Results

In patient-derived GC tissue, CLDN18.2 was frequently (∼80%) and robustly expressed in both primary and metastatic tumors. IMAB362 was highly selective for CLDN18.2 both in vivo and in vitro. IMAB362 mediated effective and target-selective antibody-dependent cellular cytotoxicity (ADCC) against GC cell lines with endogenous CLDN18.2 expression and induced complement-dependent cytotoxicity (CDC)-mediated lysis of CLDN18.2-expressing tumor cells. IMAB362-mediated ADCC and CDC were not affected by the presence of CLDN18.2-negative cancer cells. Treatment with chemotherapy sensitized tumor cell lines to IMAB362-mediated mechanisms by increasing CLDN18.2 expression; improved antitumor activity was observed in xenografted mice treated with IMAB362 + chemotherapy compared with mice treated with chemotherapy alone.

Conclusions

IMAB362 is a target-selective mAb with strong immune effector mediated antitumor activity (ADCC, CDC) that contributes to the elimination of CLDN18.2-expressing GC cells and synergizes with chemotherapy.

Legal entity responsible for the study

Ganymed Pharmaceuticals AG, A company of Astellas Pharma, Inc A company of Astellas Pharma Inc Ganymed Pharmaceuticals GmbH A company of Astellas Pharma Inc Ganymed Pharmaceuticals GmbH A company of Astellas Pharma Inc Ganymed Pharmaceuticals AG, A company of Astellas Pharma Inc

Funding

Ganymed Pharmaceuticals AG, A company of Astellas Pharma, Inc A company of Astellas Pharma Inc Ganymed Pharmaceuticals AG, A company of Astellas Pharma, Inc

Disclosure

R. Mitnacht-Kraus: Employee of Ganymed Pharmaceuticals AG, a company of Astellas Pharma, Inc. In addition, Dr. Mitnacht-Kraus has a patent P-24PCT issued, a patent P-33PCT issued, a patent P-34PCT issued, and a patent P-36PCT issued. M. Kreuzberg: Employee of Ganymed Pharamceuticals AG, a company of Astellas Pharma, Inc. M. Utsch: Employee of Ganymed Pharmaceuticals AG, a company of Astellas Pharma, Inc. In addition, Dr. Utsch has a patent PCT/EP2012/002210 issued. U. Sahin: Stock option owner, ex-shareholder and cofounder of Ganymed Pharmaceuticals AG and Founder/CEO/shareholder of Biontech Holding outside the submitted work. Dr. Sahin has several patents issued to this work that have been acquired by Astellas. Ö. Türeci: Stock option owner, ex-shareholder, cofounder & CEO of Ganymed Pharmaceuticals AG, has received consultancy fees from Astellas, and has several patents issued to this work that have been acquired by Astellas.

Collapse
Poster display session Poster Display session

379P - A novel mRNA-based patient selection strategy identifies fibroblast growth factor receptor (FGFR) inhibitor-sensitive tumors: Results from rogaratinib Phase-1 study (ID 2626)

Presentation Number
379P
Lecture Time
13:15 - 13:15
Speakers
  • M. Joerger
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Altered FGFR signaling is a potential target for anticancer therapy. Rogaratinib (BAY1163877) is an oral inhibitor of FGFRs 1-4. Screening for patients based on tumor FGFR1-3 mRNA overexpression, we reported in a phase I study that selected urothelial carcinoma patients were highly sensitive to rogaratinib treatment (NCT01976741; Joerger et al, ESMO 2016). Here we further identify patients sensitive to rogaratinib with malignancies not previously identified as being driven by FGFRs.

Methods

Subjects with treatment-refractory advanced or metastatic solid tumors were screened for high FGFR1-3 mRNA expression levels by RNA in situ hybridization (RNAscope®; Advanced Cell Diagnostics, Inc., Newark, CA) and Nanostring® assay (NanoString Technologies, Inc., Seattle, WA) from fresh or archival tumor specimens. FGFR-positive patients were treated with 800 mg BID on a continuous 21-day cycle. Responses were assessed by Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1.

Results

More than 500 patient biopsies were screened for FGFR1-3 mRNA levels. Seventy two FGFR-positive patients were treated with rogaratinib, with 63 evaluable for response. Clinical responses were observed in tumor types not previously associated with FGFR alterations, including a partial remission (PR) in a patient with a FGFR1 mRNA-positive adenoid cystic carcinoma of the tongue and a PR in a FGFR3 mRNA-positive head and neck squamous cell carcinoma patient. Long-lasting stable disease with tumor shrinkage was also seen in patients with a) FGFR3 mRNA-positive gastric cancer, b) FGFR3 mRNA-positive lung squamous cell carcinoma c) FGFR3 mRNA-positive lung adenocarcinoma, d) FGFR2 mRNA-positive breast cancer and e) FGFR1 mRNA-positive hemangioendothelioma with complete disappearance of edema over 18 months.

Conclusions

Patient selection for treatment with rogaratinib based on quantification of FGFR1-3 mRNA isoforms in all tumor types irrespective of underlying data on DNA alterations is feasible and yields clinically meaningful responses in tumor types not been previously associated with altered FGFR signaling.

Clinical trial identification

NCT01976741

Legal entity responsible for the study

Bayer AG

Funding

Bayer AG

Disclosure

S. Bender, P. Ellinghaus, M. Ocker: Employment: Bayer AG. S. Ince, P. Rajagopalan: Employment: Bayer HealthCare Pharmaceuticals. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

380P - A phase 1b study evaluating the safety and pharmacokinetics (PK) of regorafenib (REG) in combination with cetuximab (CTX) in patients with advanced solid tumors (ID 2537)

Presentation Number
380P
Lecture Time
13:15 - 13:15
Speakers
  • C. Weekes
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Combining REG with CTX may overcome intrinsic and acquired resistance in EGFR-sensitive and -resistant tumors. We evaluated the safety, PK, maximum tolerated dose (MTD), and preliminary efficacy of REG plus standard dose of CTX (initial 400 mg/m2 intravenously followed by 250 mg/m2 weekly) in a phase 1b study. Final results from the intermittent REG dosing arm (3 weeks on/1 week off) are reported, with final results from the terminated continuous REG dosing arm (n = 11) previously presented (Weekes et al. AACR 2016, abstract CT148).

Methods

This was an open-label, dose-escalation (3 + 3 design) study in patients with locally advanced or metastatic solid tumors who progressed after standard therapy. The starting dose of REG was 120 mg once daily (QD) in a 28-day cycle (3 weeks on/1 week off) plus CTX. If tolerable, REG was escalated to 160 mg QD. If not tolerable, the REG dose was reduced to 80 mg QD. Dose-limiting toxicities (DLTs) were evaluated in Cycle 1. Adverse events (AEs) were graded according to NCI-CTCAE v4.03. Antitumor activity was assessed using RECIST v1.1.

Results

As of January 31, 2017, 31 patients received REG in an intermittent schedule plus CTX: 8 patients received REG 120 mg and 23 received REG 160 mg. One DLT of grade 3 hand–foot skin reaction was reported in 6 evaluable patients at the 120 mg dose level. No DLT was confirmed at the 160 mg dose level. The MTD was declared at the standard dose of REG 160 mg QD (3 weeks on/1 week off) plus the standard dose of CTX. The most common AEs, regardless of relationship to study drug, were hypophosphatemia (42%), fatigue (39%), and nausea (39%). The most common grade ≥3 REG-related AEs were hypophosphatemia (23%) and fatigue (10%). REG AUC(0–tlast) was 29.1 mg·h/L at 160 mg and 17.4 mg·h/L at 120 mg. CTX had no effect on the PK of REG. One patient (120 mg REG) had a partial response; 6 (160 mg REG; 29%) and 2 (120 mg REG; 25%) patients had stable disease.

Conclusions

REG at 160 mg QD (3 weeks on/1 week off) plus standard dose of CTX was tolerated with no unexpected toxicities. Observed AEs were in line with known REG and CTX safety profiles.

Clinical trial identification

NCT01973868

Legal entity responsible for the study

Bayer

Funding

Bayer

Disclosure

C. Weekes: Advisory Board: Celgene, Merrimack, Bayer. A.C. Lockhart: Corporate Sponsored Research: Amgen, Bayer, Daiichi Sankyo, EMD Serono, Genentech/Roche, Eli Lilly, Millennium/Takeda, Novartis, Sanofi, Teva, Zenyaku Kogyo. H-J. Lenz: Research/Education Grant, Honoraria, Advisory Board and Consulting: Bayer, EMD. J.J. Lee: Research/Education Grant: Merck Advisory Board: Genentech/Roche. A. Cleton: Employment: Bayer. Stock Ownership: Bayer, Pfizer. F. Huang, I. Sturm: Employment and Stock Ownership: Bayer.

Collapse
Poster display session Poster Display session

381P - Clinical study of apatinib in the treatment of malignant pleural effusion and malignant celiac effusion (ID 1393)

Presentation Number
381P
Lecture Time
13:15 - 13:15
Speakers
  • M. Xin
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Malignant pleural effusion and malignant celiac effusion is a severe complication of the advanced malignant tumor, The formation of malignant pleural effusion and malignant celiac effusion relied on VEGF growth factor.Apatinib blocking vascular endothelial growth factor (VEGF) combining with the receptor and blocking signal transduction pathway, thereby inhibiting tumor and the formation of new blood vessels, inhibit the formation of the malignant pleural effusion and malignant celiac effusion. We conducted the study to exploring the single-agent oral path of Apatinib for the treatment of malignant pleural effusion and malignant celiac effusion of the clinical curative effect.

Methods

Patients were eligible for inclusion in the study if they had histologically confirmed malignancies, with malignant pleural effusion and malignant celiac effusion. Eastern Cooperative Oncology Group (ECOG) performance status of 0-2. Study Objective: Clinical curative effect. Study Design and Treatment: This study recruited 13 patients with histologically confirmed malignancies, and the patients with malignant pleural effusion and malignant celiac effusion from January 2016 to October 2016. All patients had an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2. All patients were given the oral drug Apatinib, the doses of oral given is 500 mg or 425 mg everyday until progressive disease (PD) or intolerable toxicity. Assessment and Statistical analysis: The assessmet were determined according to the World Health Organization (WHO) criteria. The ORR and DCR analyses were based on frequencies.

Results

381P Baseline patient characteristics

Characteristic Apatinib(n = 13)
Age(years)Median(range) 58(44-84) Sex Male 7(53.85) Female 6(46.15) EGOG PS 0 2(15.38) 1 6(46.15) 2 5(38.46) Carcinoma Lung cancer 6(46.15) Pancreatic cancer 4(30.77) Breast cancer 2(15.38) Thymic cancer 1(7.7)
Efficacy: Apatinib is effective for the treatment of malignant pleural effusion and malignant celiac effusion: CR 5 cases, PR 5 cases, SD 3 cases, overall-response rate (ORR)76.92%, disease-control rate (DCR)100%.

Conclusions

Apatinib is a safe, feasible way in the treatment of malignant pleural effusion and malignant celiac effusion. Relying on the simple method and favourable rencent curative effect. Apatinib provides a new way for the treatment of malignant pleural effusion and malignant celiac effusion that deserves further research.

Legal entity responsible for the study

GuoXin Ma

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

382P - Evaluation of NRP-1 TM domain targeting peptide in melanoma (ID 1660)

Presentation Number
382P
Lecture Time
13:15 - 13:15
Speakers
  • M. Pierdant
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Membrane proteins and their interactions play central roles in a variety of cellular processes including nutrient uptake, signaling and cell-cell communication. Many cancers, neurological, metabolic and immune disorders can be attributed to the abnormal function of membrane proteins. All of these membrane proteins either contain one or several transmembrane (TM) strands and/or undergo some degree of oligomerization. Thus, stabilization or disruption of these protein-protein interactions is therefore of great interest for the modulation of protein function within the cell. In this context, TM domains of neuropilin-1 receptor (NRP1) and human epidermal growth factor receptor (HER2) are particularly studied in our laboratory to develop anti-cancer therapies based on the interference of the TM domains. Antitumor effects of TM peptides targeting NRP1 or HER2 has been proved in glioblastoma (Nasarre et al., 2010) and breast cancer (Arpel et al., 2016) respectively. In addition, HER2 has been proved to reduce metastasis in a metastatic model of breast cancer (Arpel et al., 2014). Moreover, NRP1 expression has been detected by immunostaining in tumor specimens obtained from patients with prostate, lung, pancreatic, colon carcinoma and melanoma.

Methods

In order to expand the therapeutic applications of these TM peptides, we evaluated its anti-tumoral activity in melanoma in an orthotopic allograft model in nude mice (B16F10 cell line).

Results

Our results show, that a local administration of MTP-NRP1 (1µg/kg, 3x/week) significantly reduces tumor development after 11 days of treatment. These results concur with the dT/dC% value at day 13 (18,3%) indicating that the treatment is efficiently impacting the tumor volume after this period. The application of the RECIST criteria identified 7.1% of mice presenting stable disease while 78,6% exhibited partial response. However 14.3% of mice were non responders to the treatment.

Conclusions

Hence, current work is conducted to analyze by gene array profiling the differences in NRP1 and other cancer promoting receptors between responder and non-responder populations. This is a prerequisite to decide whether MTP-NRP1 could be developed in this indication, particularly by associating it to another drug in the non-responding population.

Legal entity responsible for the study

INSERM U1109, “Microenviromental Niche in Tumorigenesis and Targeted Therapy” MN3T lab, Labex Medalis, University of Strasbourg, France.

Funding

Labex Medalis, University of Strasbourg, France.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

383P - A phase I dose-escalation study of the novel peptide ALM201 in patients (pts) with advanced solid tumours (ID 4174)

Presentation Number
383P
Lecture Time
13:15 - 13:15
Speakers
  • A. El-Helali
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

ALM201 is a novel 23-amino acid peptide derived from FKPB-L, a human endogenous protein with inherent anti-angiogenic activity. Pre-clinically, ALM201 potently inhibits cell migration, invasion and neo-vessel formation without effects on cell cycle or proliferation.

Methods

We enrolled pts with solid tumours using a single patient (10 - 40mg) then 3 + 3 (80 - 300mg) dose escalation design. ALM201 was administered subcutaneously (S.C.) once daily on days 1-5, 8-12, and 15-19 every 21 days. All pts continued until disease progression (PD) or dose-limiting toxicity (DLT). Primary objectives were to determine the safety, tolerability and recommended phase II dose (RP2D) of ALM201. Secondary objectives were to determine the pharmacokinetics (PK) and anti-tumour activity. Plasma and urine samples were analysed by a validated LC-MS/MS method.

Results

We report interim data in 18 evaluable pts enrolled in 8 dose levels. Cancers included ovarian (5), colorectal (4), NSCLC (2), endometrial (1), gallbladder (1), cervical (1), urachal (1), renal (1), pancreatic (1) and mesothelioma (1). Doses of 10 - 300mg were well tolerated. No DLTs were observed. The only toxicity was grade 1 injection site skin reaction. Median treatment duration was 11.1 weeks (range 3-18 wks). Two patients had stable disease for up to 6 cycles prior to progression. Maximal plasma concentrations were typically observed 1.5h (0.75-4h) after dosing indicating fairly rapid absorption. Above 40mg, plasma concentrations were consistently seen up to 6h after dosing (assay LLOQ = 100ng/mL). Where the terminal phase could be defined, half-lives <2h were reported (0.9-1.8h, n = 4 pts). Plasma Cmax and AUC tended to increase with dose; with no evidence of dose non-proportionality between 10 - 160mg and lower than proportional increases in exposure observed above 160mg. No evidence of drug accumulation was observed over successive dose cycles. No unchanged parent drug was detectable in patient urine in the 0-6h collection phase after dosing.

Conclusions

Monotherapy ALM201 administered S.C. demonstrated a very good safety profile and acceptable PK in patients with advanced solid tumors. The RP2D analysis is currently ongoing.

Clinical trial identification

EudraCT No: 2014-001175-31

Legal entity responsible for the study

Almac Discovery

Funding

Almac Discovery and Invest Northern Ireland

Disclosure

R. Kennedy: Employee at Almac Diagnostics & Almac Group. A. Cranston: Employee at Almac Discovery. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

385P - Interim pharmacokinetic (PK) and pharmacodynamic (PD) data from the first-in-human study of NUC-3373, a pyrimidine nucleotide analogue, in patients with advanced solid tumors (ID 3432)

Presentation Number
385P
Lecture Time
13:15 - 13:15
Speakers
  • E. Ghazaly
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

NUC-3373, a phosphoramidate transformation of FUDR, is designed to bypass 5-FU and capecitabine resistance. Intracellular levels of the active 5-FU metabolite FUDR-MP in vitro were 363x higher than with 5-FU. Here, we report the interim PK and PD data from the ongoing first-in-human dose-escalation study of NUC-3373 in patients (pts) with advanced solid tumors (NuTide:301).

Methods

NUC-3373 was administered as 30- min IV infusion on days 1, 8, 15 and 22 of a 28-day cycle. The first 3 pt cohorts received NUC-3373 at 125mg/m2, 250mg/m2 and 500mg/m2. The primary objective was to determine RP2D. Secondary objectives included safety, PK and PD profiles, and anti-tumour activity. Blood samples were collected pre-dose and at 11 time-points up to 48h post-dose during cycle 1. Plasma and intracellular metabolites were measured by UPLC-MS/MS; western blotting of extracted PBMCs measured thymidylate synthase (TS) within ternary complexes (TS-T).

Results

PK/PD analyses were conducted on 16 of the 17 pts recruited to the first 3 dosing cohorts pts (94%), median age 59 yrs (range 24-71), with 7 cancer types, the majority (76%) being colorectal cancer. Mean plasma Cmax and AUC of NUC-3373 were dose proportional. Linear PK was confirmed across the studied dose range with clearance of 3.4 ± 0.6 L/hr and plasma t1/2 of 9.4h ±0.98h. Intracellular FUDR-MP was detectable at 5 minutes post-infusion with t1/2 of 14.3h ±1.7h and was still present at 48h. In the 500mg/m2 cohort the mean intracellular Cmax and AUC0-24 of FUDR-MP were 4.2pmol/106 cells and 16.5pmol/106 cells/hr. Within 1 hour of infusion, FUDR-MP was present within TS-T leading to depletion of the intracellular dTMP pool after 2-4h. Toxic metabolites FBAL and FUTP were undetectable intracellularly or in plasma. Dose escalation continues.

Conclusions

PK/PD data demonstrate NUC-3373 generates high intracellular concentrations of the active cytotoxic metabolite FUDR-MP, which efficiently sequester TS into TS-T inhibiting its activity. This and lack of toxic metabolite accumulation indicate NUC-3373 has a favorable PK profile compared to the established fluoropyrimidines.

Clinical trial identification

ClinicalTrials.gov NCT02723240 EudraCT Number: 2015-002250-13 Release date: December 8, 2015

Legal entity responsible for the study

University of Oxford

Funding

Nucana BioMed Ltd

Disclosure

D.J. Harrison: Advisory in Cytosystems, Nucana and Ryboquin. Leadership role in Ryboquin The Institute received research funding from Nucana Ltd and Ryboquin Ltd. T.R.J. Evans: Advisory role in Bristol-Myers Squibb, Vertex, GSK, Baxter and Celgene. The institute received research funding from several Pharmaceutical companies. S.P. Blagden: Advisory role in Celgene and Novartis. Holder of an IP in RNA Guardian. Travel grant from Nucana. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

386P - Efficacy of pembrolizumab in phase 2 KEYNOTE-164 and KEYNOTE-158 studies of microsatellite instability high cancers (ID 5340)

Presentation Number
386P
Lecture Time
13:15 - 13:15
Speakers
  • L. Diaz
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

A high level of microsatellite instability (MSI-H) is indicative of a tumor deficient in mismatch repair (dMMR). Prior reports suggest that anti-PD-1 antibody therapy provides durable responses in patients (pts) with MSI-H cancers. Here we assessed efficacy of PD-1 blockade using pembrolizumab in pts with dMMR/MSI-H (hereafter termed MSI-H) advanced cancer enrolled in two ongoing, global, multicenter phase 2 studies KEYNOTE (KN)164 and KN158. We report results in 61 pts with MSI-H CRC and 77 pts with MSI-H non-CRC across 20 tumor types.

Methods

KN164 enrolled pts with MSI-H colorectal cancer (CRC) and ≥2 prior therapies, whereas the multicohort KN158 study included pts with MSI-H non-CRC and ≥1 prior therapy. MSI-H status was determined locally by IHC or PCR or centrally by PCR. Eligible pts in both studies received pembrolizumab 200 mg Q3W. Tumor response was assessed every 9 wk. Primary endpoint was ORR by independent central review per RECIST v1.1. Database cut-off date was Feb 10, 2017 for KN164 (≥54 wk follow-up) and Jan 27, 2017 for KN158 (≥27 wk follow-up).

Results

KN164 enrolled 61 pts with MSI-H CRC (90% with ≥2 prior therapies) and KN158 enrolled 77 pts with MSI-H non-CRC (52% with ≥2 prior therapies), at data cutoff. Tumor types represented in KN158 in at least 2 pts included endometrial (n = 17), gastric (n = 11), small intestinal (n = 10), pancreatic (n = 9), biliary (n = 8), mesothelioma and small cell lung (n = 3 each), adrenocortical, bladder, and thyroid (n = 2 each) cancers. ORR was 27.9% (n = 17 [all confirmed]; 95% CI 17.1%-40.8%) for MSI-H CRC and 37.7% (n = 29 [23 confirmed and 6 unconfirmed]; 95% CI 26.9%-49.4%) for MSI-H non-CRC. Median DOR was not reached for MSI-H CRC (range 2.9+ to 12.5+) or MSI-H non-CRC (range 2.4+ to 9.2+). Median OS was not reached for either MSI-H CRC or MSI-H non-CRC, with 6-mo OS rates of 87% and 73%, respectively.

6-mo PFS rates were 43% for MSI-H CRC and 45% for MSI-H non-CRC. 4 (7%) pts with MSI-H CRC and 7 (9%) with MSI-H non-CRC had serious drug-related AEs. The safety profile was consistent with that previously seen for pembrolizumab.

Conclusions

Pembrolizumab provides robust antitumor activity with durable responses in heavily pretreated pts with MSI-H cancers.

Clinical trial identification

ClinicalTrials.gov NCT02460198. MK-3475 164 First received: May 29, 2015 Last updated: February 16, 2017 Last verified: February 2017 AND ClinicalTrials.gov NCT02628067. MK-3475 158 First received: December 9, 2015 Last updated: February 23, 2017 Last verified: February 2017

Legal entity responsible for the study

Merck & Co., Inc.

Funding

Merck & Co., Inc.

Disclosure

L. Diaz: Travel expenses from Merck & Co., Inc. A. Marabelle: Advisory board for Merck Serono, eTheRNA, Lytix pharma, Kyowa Kirin Pharma, Bayer, Novartis, Bristol-Myers Squibb, Symphogen, Genmab, Amgen, Biothera, Nektar, GSK, Oncovir, Pfizer, Seattle Genetics, Flexus Bio, Roche-Genentech. Speakers’ bureau for MSD. T.W. Kim: Advisory board member for Amgen. Research funding from Taiho, Bayer. R. Geva: RG: Advisory board member for Bayer, MSD & Novartis. Honoraria from Bristol-Myers Squibb, Lilly, Medison, Roche, Novartis and Janssen. E. Van Cutsem: Research funding from Amgen, Bayer, Boehringer, Celgene, Ipsen, Lilly, MSD, Merckserono, Novartis, Roche, Sanofi and Servier. T. André: Honoraria from Bohrigner Ingenheim, Merck, Serono, Baxter, Bayer and Roche. P.A. Ascierto: Advisory board member for Bristol-Myers Squibb, Roche-Genentech, MSD, Novartis, Array, Amgen, Merck Serono, and Pierre Fabre. M. Maio: Honoraria, travel expenses and consulting fees from Bristol-Myers Squibb, Merck Sharp & Dohme, Roche, GSK, & Medimmune. R. Guimbaud: Advisory board member for Novartis. Travel expenses, including accommodations from Roche, Ipsen, Lilly and Sanofi. D. Jaeger: Consulting or advisory roles and Roche, Bristol-Myers Squibb & Bayer. T. Yoshino, A. Joe, B. Lam, J. Ding, S. Pruitt, S.P. Kang: SPK is employed by Merck & Co., Inc. and may own stock options in the company. D.T. Le: Research grants and honoraria from Merck & Co., Inc. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

387P - Preliminary results from subsets of patients (pts) with advanced gastric cancer (GC) and esophageal carcinoma (EC) in a dose-escalation/expansion study of BGB-A317, an anti-PD-1 monoclonal antibody (mAb) (ID 2097)

Presentation Number
387P
Lecture Time
13:15 - 13:15
Speakers
  • J. Desai
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

BGB-A317 is a humanized IgG4 anti-PD-1 mAb that blocks PD-L1/L2 binding to PD-1 restoring T-cell-mediated tumor inhibition. The Fc-hinge region has been engineered to preclude FcγR1-mediated binding to macrophages/myeloid-derived suppressor cells (MDSCs). Upregulation of PD-1/L1 and predominance of macrophages and MDSCs have been reported in GC and EC supporting the rationale of evaluating BGB-A317 in pts with GC or EC.

Methods

This ongoing, open-label, dose-escalation/expansion study is being conducted to evaluate the safety, tolerability and anti-tumor activity of BGB-A317 in pts with advanced solid tumors. Pts with histologically confirmed advanced GC or EC were eligible and treated with BGB-A317 at 2 mg/kg or 5 mg/kg every two weeks (Q2W) or Q3W. Adverse events (AEs) were assessed per NCI-CTCAE v4.03 and tumor assessments were performed approximately every two months via RECIST v1.1.

Results

As of 6 MAR 2017, 55 pts [median age 62 yrs (22-81)] with recurrent/refractory GC (n = 28) or EC (n = 27) were treated. Most were Caucasian (n = 36) and all pts had received ≥1 prior line of anti-cancer treatment. Median treatment duration was 51 days (5–363); 19 pts remain on study. The most common treatment-emergent AEs were fatigue (n = 11), nausea (n = 9) and dysphagia (n = 8); 46% pts experienced AEs ≥Grade (Gr) 3 but none were treatment related. One serious AE (diarrhea [Gr 2]) was considered related to treatment by investigators. Of the 47 evaluable pts, the disease control rate, defined as the proportion of pts who achieved complete or partial response (CR or PR) or stable disease (SD), is 32%. PRs have been reported in 3 pts (GC = 2; EC = 1) with duration of responses being 96, 125 and 188 days respectively, 2 pts are still on treatment; 5 initial documentations of PRs awaiting confirmation (GC, n = 2; EC, n = 3) have been reported in 12 pts with SD (GC = 5; EC = 7).

Conclusions

BGB-A317 appears to be generally well tolerated in pts with recurrent/refractory GC or EC. The preliminary safety profile and anti-tumor activity appear to be consistent with other checkpoint inhibitors and support continued exploration and development of BGB-A317 in pts with advanced GC or EC.

Clinical trial identification

NCT02407990, March 26, 2015

Legal entity responsible for the study

Beigene Ltd.

Funding

Beigene Ltd.

Disclosure

J. Desai: Honoraria: Bayer, Merck Serono, Novartis. Consulting or advisory role: Amgen, Bayer, Bionomics, Circadian Technologies, Merck Serono, Norvartis. Research funding: GlaxoSmithKline (Inst), Roche-Genentech (Inst), Ventana Medical Systems (Inst). B. Markman: Personal fees from BeiGene during the conduct of the study. V. Atkinson: Honoraria: Bristol-Myers Squibb; MSD; Novartis. Consulting/advisory role: Bristol-Myers Squibb; MSD; Novartis. Travel, accommodations, expenses: Bristol-Myers Squibb; MSD; Novartis. A. Zhu: Research Funding: Lilly (Inst). J. Song: Employee of BeiGene USA, Inc. Q. Qi: Employee of Beigene (Beijing) Co. Ltd. A. Kang: BeiGene employee. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

388P - Preliminary results from a subset of patients (pts) with advanced head and neck squamous carcinoma (HNSCC) in a dose-escalation and dose-expansion study of BGB-A317, an anti-PD-1 monoclonal antibody (mAb) (ID 2098)

Presentation Number
388P
Lecture Time
13:15 - 13:15
Speakers
  • L. Horvath
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

BGB-A317 is a humanized IgG4 anti-PD-1 mAb that blocks PD-L1/PD-L2 binding to PD-1 restoring T-cell mediated tumor response. The Fc-hinge region has been engineered to preclude FcγR1 mediated binding to macrophages/myeloid-derived suppressor cells, a potential mechanism of PD-1 bound T-cell clearance. Regulatory Foxp3+ T-cells and PD-1+T-cell infiltration in the tumor microenvironment have been reported in HNSCC supporting the rationale for evaluation of BGB-A317 in pts with HNSCC. Here we present the preliminary results from a subset of pts with HNSCC treated with BGB-A317.

Methods

This ph 1, open-label, multi-center, dose-escalation/expansion study was conducted to evaluate the safety, tolerability, and anti-tumor activity of BGB-A317 in pts with advanced solid tumors. Pts with histologically confirmed advanced HNSCC who progressed following standard of care treatment were eligible to receive BGB-A317 administered at a dose of 5 mg/kg Q3W. Adverse events (AEs) were assessed per NCI-CTCAE v4.03. Tumor assessments were performed Q9W per RECIST v1.1.

Results

As of 6 Mar 2017, 18 pts with recurrent HNSCC were enrolled (median age, 63 years [25-78]). Most pts were male (89%), Caucasian (67%) and had received ≥2 prior lines of anti-cancer treatment. The median treatment duration for BGB-A316 was 104 days (30-245); 7 pts remain on study. Most treatment-emergent AEs were Grade (Gr) 1/2 in severity and the more common AEs were fatigue (n = 6), constipation (n = 3) and ear discomfort (n = 3). Eleven unique AEs ≥Gr 3 were reported in 7 pts: dysphagia, nausea salivary gland enlargement, dyspnea, pleuritic pain, aspiration pneumonia, infection-related COPD exacerbation, parotitis, ocular hyperemia, and wound hemorrhage. A partial response (PR) has been confirmed in 1 pt, and 8 pts have stable disease (SD,) including 2 unconfirmed PRs. The disease control rate, defined as the proportion of pts who have achieved CR, PR and SD, is 50%.

Conclusions

BGB-A317 appears to be well tolerated in pts with recurrent HNSCC. The preliminary safety profile and anti-tumor activity support continued investigation of BGB-A317 in this setting.

Clinical trial identification

NCT02407990, March 26, 2015

Legal entity responsible for the study

Beigene Ltd.

Funding

Beigene Ltd.

Disclosure

L. Horvath: Clinical trials agreement with budget from Beigene, during the conduct of the study; J. Desai, S. Sandhu: Honoraria: Bayer, Merck Serono, Novartis. Consulting or advisory role: Amgen, Bayer, Bionomics, Circadian Technologies, Merck Serono, Norvartis. Research funding: GlaxoSmithKline (Inst), Roche-Genentech (Inst), Ventana Medical Systems (Inst). A.G. Hill: Research funding, Stock and Other Ownership Interests: Tasman Oncology. Travel Accommodations, Expenses: Bristol-Myers Squibb. B. Markman: Personal fees from Beigene during the conduct of the study. Z. Chen, J. Hou: Employee of BeiGene Ltd. X. Tan: Employee of BeiGene (Beijing) Co. Ltd. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

389P - Preliminary results from a subset of patients (pts) with advanced ovarian cancer (OC) in a dose-escalation/expansion study of BGB-A317, an anti-PD-1 monoclonal antibody (mAb) (ID 2099)

Presentation Number
389P
Lecture Time
13:15 - 13:15
Speakers
  • T. Meniawy
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

BGB-A317 is a humanized IgG4 anti-PD-1 mAb that blocks PD-L1/L2 binding to PD-1 restoring T-cell-mediated tumor inhibition. The Fc-hinge region has been engineered to preclude FcγR1 mediated binding to macrophages/myeloid-derived suppressor cells (MDSCs). Upregulation of PD-1/L1 and predominance of macrophages and MDSCs have been reported in OC supporting the rationale of evaluating BGB-A317 in pts with OC.

Methods

An open-label, multi-center, dose-escalation/expansion study is being conducted to evaluate the safety, tolerability and anti-tumor activity of BGB-A317 in pts with advanced solid tumors. Pts with histologically confirmed advanced OC were eligible and treated at different dose levels (0.5, 2, 5, 10 mg/kg intravenously [IV] every 2 weeks [Q2W] in dose escalation, or at 2 or 5 mg/kg IV Q2W or Q3W, or 200 mg IV Q3W in dose expansion, or 5 mg/kg IV Q3W in indication expansion). Tumor assessments, including CA125, occurred approximately every 2 months and response was collected according to both RECIST 1.1 and GCIG criteria. Adverse events (AEs) were assessed per NCI-CTCAE v4.03.

Results

As of 6 Mar 2017, 51 pts [median age 62 (19–80) yrs] with recurrent/refractory OC were enrolled. Most pts were Caucasian (88%), all had received ≥1 prior line of anti-cancer treatment (median 3 [1–12]). Median duration of treatment was 68 (22–446) days; 7 pts remain on study. The most common treatment-emergent AEs were nausea (37%), fatigue (28%), and abdominal pain (28%). 49% of pts experienced an AE ≥Grade (Gr) 3; stomatitis (n = 1) and diarrhoea (n = 1) were Gr 3 AEs considered treatment-related by investigators. Mucosal inflammation, pyrexia and colitis were serious AEs considered treatment-related by investigators (n = 1, each). Among 51 evaluable pts, the disease control rate is 43%; 2 PRs have been reported including 1 pt who remains on study and to date has achieved an 89% reduction in target lesions.

Conclusions

BGB-A317 appears to be generally well tolerated in pts with recurrent/refractory OC. The preliminary safety profile and anti-tumor activity are consistent with that observed with other checkpoint inhibitors and support continued investigation of BGB-A317.

Clinical trial identification

NCT02407990, March 26, 2015

Legal entity responsible for the study

Beigene Ltd.

Funding

Beigene Ltd

Disclosure

T. Meniawy: Non-financial support and other from BeiGene during the conduct of the study; Non-financial support from Bristol-Myers Squibb outside the submitted work. J. Desai: Honoraria: Bayer, Merck Serono, Novartis. Consulting or advisory role: Amgen, Bayer, Bionomics, Circadian Technologies, Merck Serono, Norvartis. Research funding: GlaxoSmithKline (Inst), Roche-Genentech (Inst), Ventana Medical Systems (Inst). L. Horvath: Clinical trial agreement with budget from Beigene, during the conduct of the study; S. Sandhu: Honoraria: Bristol-Myers Squibb, Merck. Consulting or Advisory: Amgen. Z. Wu: Employee of Beigene USA, Inc. Z. Qin, K. Kang: Employee of BeiGene (Beijing) Co. Ltd. B. Markman: Personal fees from Beigene during the conduct of the study. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

390P - A phase I study of durvalumab (D) in combination with olaparib (O) and cediranib (C) in recurrent women’s cancers (ID 4055)

Presentation Number
390P
Lecture Time
13:15 - 13:15
Speakers
  • A. Zimmer
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Recent data showed a PARP inhibitor, O and a VEGFR1-3 inhibitor, C together are clinically superior to O alone in recurrent platinum-sensitive ovarian cancer (OvCa). We hypothesized reduced VEGF signaling by C and DNA damage by O may complement anti-tumor activity of immune checkpoint blockade, D. We previously reported the safety data and RP2D of D in combination with O or C. We now report RP2D and PK/PD data of the D+O+C therapy (NCT02484404).

Methods

Eligible patients (pts) with PS 0-1 and good end organ function received D+O+C in a 3 + 3 design. An intermittent C schedule (C; 5 days on/2 days off) at 15 or 20 mg (dose level: 1, 2) was combined with D 1500 mg IV q28 days, and O tablets 300 mg BID. The DLT period was one 28d cycle. Safety was assessed by CTCAEv4.0 and response by RECISTv1.1. Plasma samples were collected for O and C PK analysis and for pro-inflammatory cytokines (IFN-γ, IL-10, IL-12, IL-2, IL-6, IL-8 and TNF-a) pre-treatment and on therapy (cycle 1 day 15 and cycle 3 day 1).

Results

Nine women (median age 59yr [44-73], and median 3 prior therapies [2-6]) were treated. Of the 9 pts, 7 had OvCa, 1 endometrial (EnCa) and 1 triple negative breast Ca. Grade 3/4 AEs include hypertension (1/9), anemia (1/9) and lymphopenia (3/9). No pts experienced DLTs. One patient required dose reduction during cycle 5, for grade 3 anemia. Three PRs were observed in 2 OvCa and 1 EnCa (response rate of 33%, median 5 months [4+-6+]), and 4 had SD (median 5 months [3+-10+]), yielding 78% disease control rate. There were no significant changes in O and C PK parameters caused by D and the co-administration of C or O. All cytokines plasma levels were not changed significantly by the treatment. PD-L1 expression by IHC and immune subsets by flow cytometry are under analysis.

Conclusions

The RP2D for D+O+C (D 1500 mg q28d+O 300 mg tablets BID+C 20 mg 5 days on/2 days off) is tolerable and active in recurrent women’s cancers. A phase II expansion study of D+O+C is to open in OvCa.

Clinical trial identification

(NCT02484404)

Legal entity responsible for the study

National Cancer Institute Center for Cancer Research

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

391P - Phase I expansion of olaparib (PARP inhibitor) and AZD5363 (AKT inhibitor) in recurrent ovarian, endometrial and triple negative breast cancer (ID 4871)

Presentation Number
391P
Lecture Time
13:15 - 13:15
Speakers
  • S. Westin
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

We sought to confirm the recommended phase II dose (RP2D) of the combination of olaparib and AZD5363 in women’s cancers and evaluate molecular markers of response and resistance.

Methods

Olaparib tablet formulation (O) was given orally BID, AZD5363 (A) was given orally on a 4 day on/3 day off schedule. Two dose levels were planned per the ComPAKT study (Yap AACR 2015). Responses were defined using RECIST 1.1. Patients had biopsies at baseline and after 28 days of treatment for planned correlative testing.

Results

To date, 38 patients (pts) have been enrolled. Median number of prior therapies was 4 (1-8). Only 7 (18%, 5 ovarian, 2 breast) pts had known germline BRCA mutation. The first two pts on DL1 (O 300mg; A 400mg) experienced DLTs of diarrhea and vomiting. Therefore, 6 pts were treated on DL-1 (O 300mg; A 320mg). There were no DLTs on DL-1, therefore, 6 additional pts were treated on DL1. There were no DLTs on re-explored DL1. DL1 was confirmed as the RP2D. Expansion phase was performed with an additional 24 pts. Most common adverse events (≥15%) were anemia (89%, G3/4 16%), nausea (76%, G3/4 5%), diarrhea (74%, G3/4 5%), leukopenia (61%, G3/4 11%), elevated creatinine (58%, G3/4 3%), hyperglycemia (42%, G3/4 0%), fatigue (42%, G3/4 0%),vomiting (39%, G3/4 5%), anorexia (32%, G3/4 0%), mucositis (26%, G3/4 0%), hypertriglyceridemia (18%, G3/4 0%), thrombocytopenia (18%, G3/4 0%), neutropenia (18%, G3/4 10%), hypercholesterolemia (18%, G3/4 0%), hyponatremia (16%, G3/4 5%) and constipation (16%, G3/4 0%). Of 30 pts evaluable for response, overall response rate (RR) was 24%. Seven had confirmed partial response including 1 ovarian, 4 endometrial and 2 triple negative breast cancer pts. RR among endometrial cancer pts was 50% (4/8). Six additional pts had stable disease for greater than 4 months including 4 ovarian and 1 endometrial cancer pts. Assessment of molecular correlatives of response and resistance are ongoing.

Conclusions

The combination of olaparib tablet formulation and AZD5363 is well tolerated at the confirmed RP2D and demonstrates preliminary evidence of durable tumor activity in ovarian, endometrial and triple negative breast cancer. Promising response was seen in the endometrial cancer cohort.

Clinical trial identification

NCT02208375 August 4, 2014

Legal entity responsible for the study

University of Texas MD Anderson Cancer Center

Funding

AstraZeneca, NIH SPORE for Uterine Cancer, NIH SPORE for Ovarian Cancer, Sabin Family Foundation

Disclosure

S. Westin: Advisory Board/Consulting: AstraZeneca, Clovis, Roche/Genenetech, Medivation, Ovation, Casdin Capital, Vermillion. Research Support: AstraZeneca, Critical Outcomes Technologies, Inc, Novartis. J. Litton: Research Support: GSK, Medivation/Pfizer, Novartis, EMD-Serono. Advisory Board/Consulting: Novartis, Medivation/Pfizer. P. Soliman: Research: Novartis. M. Frumovitz: Research support: Novadaq, Navidea. Consulting: Novadaq. A. Jazaeri: Research Support: AstraZeneca, Lion Biotech, Pfizer. Advisory Board/Consulting: EMD-Serono, Genentech. S. Moulder: Research support: Oncothyreon, Novartis, Pfizer, EMD-Serono. P. Rugman, J. Lindemann, E. McMurtry: Employee of AstraZeneca. G. Mills: Research Support: Abbvie, Adelson Medical, Research Foundation, AstraZeneca, Nanostring, Pfizer, Tesaro. Consultant: AstraZeneca, Medimmune, Pfizer Licensed Technology HRD Assay to Myriad Genetics. R. Coleman: Research Support: AstraZeneca, Clovis, Janssen, Merck, Roche-Genentech, Millennium. Advisory Board/Consulting: AstraZeneca, Clovis, Abbvie, Janssen, Merck, Roche-Genentech, Tesaro, Novartis, Pfizer, Bayer, Array, Millennium. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

392P - Generation of a novel preclinical PK/PD model provides insights into PARP inhibitor clinical monotherapy activity (ID 2443)

Presentation Number
392P
Lecture Time
13:15 - 13:15
Speakers
  • M. O'Connor
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In Phase II trials, an olaparib capsule dose that inhibits PARP (100 mg bd) was clinically inferior to the MTD (400 mg bd). Using an in vivo model, we investigated this discrepancy and the effect of dose and schedule on tumour progression.

Methods

PK, PD and antitumour activity were assessed using olaparib (100, 50, 25, 10, 2.5 mg/kg qd) in a breast cancer patient-derived xenograft (PDX) with mutated BRCA2 and TP53. We studied the relationship between PARP1 inhibition and DNA single-strand breaks (SSBs) by creating a mathematical model. We also compared PK/PD model parameters with free minimum olaparib steady-state plasma concentration (Cmin,ss) in seven Phase I–III clinical studies.

Results

Only 100 and 50 mg/kg olaparib doses caused tumour regression, yet all but the lowest led to acute PAR inhibition. The differentiating factor was time over PAR IC95; simulations using the mathematical model at steady state predicted a threshold where >95% reduction in PAR caused >20-fold increase in DNA SSBs. Overlaying clinical data, we saw that 400 mg bd capsule and 300 mg bd tablet doses provide continuous PAR inhibition (Cmin,ss>IC95 upper confidence level); 100 mg bd capsule does not. Dosing schedule assessment showed PDX regression with continuous 100 mg/kg olaparib (0/10 tumours progressed by day 100). By contrast, continuous dosing at 50 mg/kg or intermittent dosing (1 wk on/1 wk off) at 100 mg/kg resulted in 2/10 and 10/10 progressions, respectively. Treatment withdrawal upon regression led to regrowth in all cases; re-challenge at continuous 100 mg/kg was effective (9/10 regressions), 50 mg/kg was not (0/10). In a maintenance setting, switching to lower or intermittent dosing resulted in tumour response more similar to that of continuous 100 mg/kg (1/10, 2/10 and 0/10 progressions, respectively).

Conclusions

Discrepancy between biologically and clinically effective olaparib doses is explained by a need for continuous >95% PAR inhibition. Both the 400 mg bd capsule and 300 mg bd tablet dose, used in the ongoing clinical programme, provide this. Dosing schedule data suggest continuous PAR inhibition is also needed for long-term responses. These data highlight important principles for treatment of patients with PARP inhibitor monotherapy.

Legal entity responsible for the study

AstraZeneca

Funding

AstraZeneca

Disclosure

M. O\'Connor, E. Cadogan, A. Hughes, M. Learoyd, H. Xu, J. Li, J. Yates: Employee of AstraZeneca and owns stock with AstraZeneca. E. Leo: Employee of AstraZeneca.

Collapse
Poster display session Poster Display session

393P - Pharmacodynamic (PD) biomarkers for the p70S6K/Akt inhibitor, M2698: Translation from animal to human and relevance to dose selection (ID 3523)

Presentation Number
393P
Lecture Time
13:15 - 13:15
Speakers
  • W. Xiong
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

M2698 is an oral, potent and selective inhibitor of p70S6K/Akt1/3 in the PAM pathway with the potential to block signaling from the Akt feedback loop and overcome tumor resistance. Preclinical studies suggest a steep exposure – adverse event relationship. Therefore, insights into drug-dependent target modulation (S6 phosphorylation (pS6) inhibition) and its association with efficacy could inform dose selection.

Methods

Pharmacokinetic (PK) data from the phase I, first-in-human (FIH) dose escalation trial conducted in patients with advanced cancer who received daily (d) oral M2698 (15-320mg/d) were evaluated by nonlinear, mixed effect modeling. Using a PK/PD model developed with data from a breast tumor cell line derived xenograft (CLDX) in mice, tumor pS6 time profiles in humans were simulated using a mouse PD model driven by human PK. Model predictions were calibrated by comparing simulations to clinical observations, with an assumed variation in sensitivity (IC50) to pS6 between CLDX and human tumors. Predicted pS6 time profiles and clinically observed pS6 inhibition in human tumors and peripheral blood mononuclear cells (PBMC) informed the dose escalation decision in the FIH study.

Results

M2698 PK profiles were best described by a two-compartment linear model with transit compartments for delayed absorption. Consistent exposure-dependent effects of pS6 inhibition in PBMCs were observed at 160-320 mg/d, with 70-80% pS6 inhibition observed in some tumors. Based on predicted pS6 inhibition, CLDX tumors were 2-3x more sensitive than human tumors. Applying a 2-3x higher IC50, simulations suggested that 250-350mg/d would achieve the PD threshold of continuous tumor pS6 inhibition ≥ 80% in 90% of a human population, leading to escalation to 380mg/d.

Conclusions

Understanding inter-species variation can improve the precision of preclinical-to-clinical translation. For M2698, preliminary clinical PD data showed that human tumors were 2-3x less sensitive to pS6 inhibition than CLDX tumors in mice. Collective PMBC and tumor PD outcomes suggest that 160 -320 mg/d M2698 may result in considerable pS6 inhibition; a range for selection of a phase 2 dose.

Clinical trial identification

NCT01971515

Legal entity responsible for the study

Merck KGaA

Funding

Merck KGaA

Disclosure

W. Xiong, I. Celik, P. Girard: Merck employee. H. Tian, A. Clark, J. Shaw, R. Kaleta: EMD Serono employee

Collapse
Poster display session Poster Display session

394P - Phase 1 study of ipatasertib (AKT inhibitor) for investigating safety, tolerability, pharmacokinetics (PK), efficacy, and biomarkers in Japanese patients (pts) with solid tumors including castration-resistant prostate cancer (CRPC) (ID 946)

Presentation Number
394P
Lecture Time
13:15 - 13:15
Speakers
  • S. Takahashi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Ipatasertib is a highly selective small-molecule inhibitor of AKT showing antitumor activity. Clinical efficacy and safety in non-Japanese pts with metastatic CRPC has been reported (ASCO, ESMO 2016). We investigated safety, tolerability, PK, efficacy, and biomarkers in Japanese pts.

Methods

Phase 1, open label, dose-escalation study; 3 + 3 design. Primary endpoints were safety, tolerability, and PK. In Stage 1, pts with solid tumors were administered 200, 400, or 600 mg ipatasertib PO daily for 21 days of a 28-day cycle. In Stage 2, pts with CRPC were administered ipatasertib (200 or 400 mg PO daily) in combination with abiraterone acetate (AA) (1000 mg PO daily) plus prednisolone (5 mg PO twice a day) for the whole 28-day cycle. In an exploratory analysis of biomarkers, we investigated the relationship between PI3K pathway alterations (PIK3CA mutation or amplification and PTEN loss) and clinical efficacy of ipatasertib.

Results

A total of 21 pts were enrolled. Stage 1: 3, 4, and 8 pts were enrolled in the 200, 400, and 600 mg cohorts. One pt in the 600 mg cohort experienced DLT [Gr3 nausea]. The most common AEs of any grade (≥30% of pts) were nausea, diarrhea, decreased appetite, vomiting, and fatigue. Cmax and AUC of ipatasertib were dose-proportional from 200 to 600 mg. Eight pts had stable disease (SD); 2 of those 8 pts continued study treatment beyond 4 months. Stage 2: 3 pts each were enrolled in the 200 and 400 mg cohorts. No DLTs were observed. The most common AEs of any grade were nausea, diarrhea, vomiting, diabetes mellitus, dysgeusia, and dizziness. Ipatasertib Cmax and AUC were similar to those in monotherapy. Of the 6 pts, complete response was observed in 1 pt and SD was observed in 1 pt. Three pts continued study treatment beyond 4 months; 2 of those 3 pts had previously received AA and enzalutamide. Biomarker results will be presented.

Conclusions

Ipatasertib was well tolerable for Japanese pts. Based on our results, the recommended doses of ipatasertib for further development are 600 mg for monotherapy and 400 mg for in combination with AA plus prednisolone.

Clinical trial identification

JapicCTI-152910, 22-May-2015

Legal entity responsible for the study

Chugai Pharmaceutical co., LTD.

Funding

Chugai Pharmaceutical co., LTD.

Disclosure

S. Takahashi: Corporate-sponsored Research: AstraZeneca, MSD, Taiho, Chugai, Novartis, Daiichi-Sankyo, Bayer, Parexel, Ono. Y. Fujiwara: Advisory Board: Bristol-Myers Squibb, Ono. Corporate-sponsored Research: AstraZeneca, Bristol-Myers Squibb, Chugai, Daiichi-Sankyo, Eisai, Eli Lilly, GlaxoSmithKline (GSK), MerckSerono, MSD. N. Matsubara: Advisory Board: Janssen Pharma, Sanofi. Corporate-sponsored Research: Janssen Pharma, Bayer, MSD, Chugai, Taiho, Sanofi. J. Tomomatsu: Industrial Physician: Eisai. S. Iwasa: Corporate-sponsored Research: Eli Lilly, Daiichi-Sankyo, Novartis, Bristol-Myers Squibb, Chugai, Eisai. A. Yamasaki: Employee of Chugai. C. Endo, S. Yokoyama: Employee and Stock ownership: Chugai. T. Doi: Advisory Board: Eli Lilly, MSD, Amgen, Daiichi-Sankyo. Corporate-sponsored Research: Eli Lilly, Taiho, Novartis, Merck Serono, MSD, Boehringer Ingelheim, Pfizer, Sumitomo Dainippon, Chugai, Kyowa Hakko Kirin, Daiichi-Sankyo, Celgene, Quintiles, Janssen, Astellas.

Collapse
Poster display session Poster Display session

395P - DS-1205b, a novel, selective, small-molecule inhibitor of AXL, delays the onset of resistance and overcomes acquired resistance to EGFR-TKIs in a human EGFR-mutant NSCLC (T790M-negative) xenograft model (ID 1014)

Presentation Number
395P
Lecture Time
13:15 - 13:15
Speakers
  • T. Jimbo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

AXL is a receptor tyrosine kinase that plays an important role in signal transduction in normal and malignant cells. Abnormal expression and/or activation of AXL can provide a survival advantage for certain cancer cells, and AXL up-regulation is associated with poor prognosis in several cancers. Recently, it has been reported that up-regulation of AXL expression is a mechanism of EGFR-TKI resistance in EGFR-mutant non-small cell lung cancer.

Methods

Kinase activity was measured by mobility shift assay. The inhibition of hGAS6-induced migration was measured in AXL-transfected NIH3T3 (NIH3T3-AXL) cells. The in vivo anti-tumor effects of DS-1205b mono- and combination-therapy with EGFR-TKI were evaluated in NIH3T3-AXL allograft and HCC827 xenograft models. Protein expression was analyzed by Western blot or immunohistochemistry and gene expression was analyzed by RT-PCR or RNA seq.

Results

We found that DS-1205b selectively inhibited AXL kinase activity with IC50 of 1.3 nM, and with NIH3T3-AXL cells, DS-1205b potently inhibited the hGAS6-induced migration in vitro with EC50 of 2.7 nM. DS-1205b monotherapy exerted significant antitumor activity in an NIH3T3-AXL allograft model. In an HCC827 xenograft model, combination treatment with DS-1205b and osimertinib significantly delayed on the onset of tumor resistance compared to osimertinib alone in a manner proportional to DS-1205b does. DS-1205b also showed a similar resistance delay effect with erlotinib combination in the same xenograft model. AXL up-regulation was associated with the development of resistance to erlotinib treatment in another HCC827 xenograft study, and DS-1205b restored the antitumor activity of erlotinib in erlotinib-resistant tumors in a dose-dependent manner.

Conclusions

In an HCC827 xenograft model of EGFR-mutant NSCLC, inhibition of AXL activity by DS-1205b restored sensitivity to erlotinib, and addition of DS-1205b to osimertinib delayed the onset of resistance to osimertinib. These findings support further non-clinical and clinical studies targeting inhibition of AXL in EGFRm NSCLC.

Legal entity responsible for the study

Daiichi Sankyo Co., Ltd.

Funding

Daiichi Sankyo Co., Ltd.

Disclosure

T. Jimbo, T. Taira, T. Komatsu, K. Kumazawa, N. Maeda, N. Haginoya, T. Suzuki, M. Ota, Y. Totoki, C. Wada, K. Inaki, T. Isoyama, M. Uno: All authors are employees of Daiichi Sankyo Group which is developing DS-1205c.

Collapse
Poster display session Poster Display session

397P - Immune related adverse events (irAEs) in early phase immunotherapy (IO) trials: Implications for recommended phase 2 dose (RP2D) determination (ID 1464)

Presentation Number
397P
Lecture Time
13:15 - 13:15
Speakers
  • Y. Kanjanapan
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

IO agents have a unique profile of irAEs. Due to the potential of delayed severe irAEs, we examined if conventional dose-limiting toxicity (DLT) periods may underestimate the rate of clinically significant irAE (csAE), defined as an irAE that required systemic therapy, drug delay or discontinuation.

Methods

A retrospective chart review of patients (pts) on early phase IO trials at Princess Margaret Cancer Centre examined severity (CTCAE v4.0), management, timing of onset and resolution of all grade (G) irAEs. A generalized estimating equation model assessed the association between time on treatment (Rx) and csAE, adjusted for duration of IO. Potential predictors of csAEs were assessed.

Results

From 8/2012-9/2016, 239 pts across 21 trials received ≥1 dose of IO (72% single agent; 28% IO-based combination). The most common tumors were melanoma (23%) and lung (18%) cancer. Among 890 total Rx-related irAEs, 93 (10%) were csAEs, including 22 (24%) endocrine, 15 (16%) gastrointestinal [GI], 11 (12%) respiratory, 10 (11%) skin and 9 (10%) hepatic csAEs. Median onset was ≤90 days for hepatic, GI and general (eg fatigue) csAEs, and >90 days for endocrine, respiratory, skin and musculoskeletal csAEs (P = 0.03). One pt with G3 hepatitis and G4 hypophosphatemia met protocol-defined DLT criteria and 27 irAEs fulfilled DLT criteria but occurred after the DLT period. 61 pts had csAEs, with 21 (34%) having >1 csAE. The onset of first csAE was 0-6 weeks (wks) in 30 (49%) pts; wks 7-12 in 16 (26%); wks 13-48 in 10 (16%); and ≥49 wks in 5 (8%) pts. The odds ratio (OR) for the first csAE occurrence within first 6 wks vs ≥ 6 wks was 3.3 (95% CI 2.0-5.6, P = 0.002), accounting for varying Rx duration. After adjustment for time on Rx, csAE correlated only with response on univariate analysis (OR 4.3, 95% CI 2.1-8.9, P < 0.001), but not with single or combination IO, age, ECOG status, prior IO or prior therapy lines.

Conclusions

Risk of first-onset csAE was higher during the initial 6 wks of IO, supporting use of conventional DLT period for dose escalation decision. However, as late csAEs were also seen, RP2D determination should consider the entire temporal course of csAE. Occurrence of csAEs positively correlated with response to IO, relative to time on Rx.

Legal entity responsible for the study

Princess Margaret Cancer Centre

Funding

None

Disclosure

M. Butler: Advisory board for: Merck, Bristol-Myers Squibb, Novartis, Turnstone, EMD Serono, Immunocore. D. Hogg: Advisory board for Roche, Bristol-Myers Squibb, Novartis, EMD Sereno, Merck. N.B. Leighl: Research funding from Novartis. L.L. Siu: Research funding from Bristol-Myers-Squibb, Merck, Novartis, AstraZeneca/Medimmune and Roche-Genentech. P.L. Bedard: Research funding from Novartis, Roche-Genentech. Bristol-Myers-Squibb, AstraZeneca/Medimmune. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

398P - Drug-induced electrolyte abnormalities in oncology phase I trials: Analysis of 1088 cases treated at The Royal Marsden Hospital (ID 3186)

Presentation Number
398P
Lecture Time
13:15 - 13:15
Speakers
  • A. Ingles Garces
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The incidence and clinical significance of electrolyte abnormalities (EAs) in phase I studies is not well documented. The objective of this study is to evaluate the incidence of EAs, graded according to CTCAE v4.03, and its correlation with factors influencing them.

Methods

A retrospective chart review was performed of 1088 cases in 82 phase 1 clinical trials consecutively treated from 2011 to 2015 at the Drug Development Unit, The Royal Marsden Hospital. Cox regression was used to examine the relationship between overall survival and baseline characteristics, treating the occurrence of grade 3/4 EAs as a time-varying covariate.

Results

The most common EAs in all grades during trials are: hyponatremia 62%, hypokalemia 40%, hypophosphatemia 32%, hypomagnesemia 17% and hypocalcemia 12%. Overall, grade 3/4 EAs occurred in 19% of cases. More specifically, grade 3/4 EAs were observed, as follow: hyponatremia 10%, hypophosphatemia 6%, hypokalemia 5%, hypomagnesemia 1%, hypermagnesemia 1%. Grade 3/4 EAs occurred during the dose-limiting toxicity window in 8.73% of cases. Overall, diarrhea was associated with hypomagnesemia in all grades (HR 1.78, 95% CI: 1.32-2.39, p < 0.001), with G3/G4 hypokalemia (HR 1.93, 95% CI: 1.09-3.43, p = 0.02) and hyponatremia in all grades (HR 0.79, 95% CI: 0.67-0.93, p = 0.006). Vomiting was also associated with hypomagnesemia in all grades (HR 1.45, 95% CI: 1.08-1.95, p = 0.01) and G3/4 hypokalemia (HR 2.91, 95% CI: 1.62-5.23, p < 0.001). Baseline hypoalbuminemia, hyponatremia and female gender are associated with higher risk of developing other EAs during trial in the univariate analysis. Patients who developed G3/4 EAs during follow-up had a poorer median overall survival (OS) (26 weeks vs 37 weeks, HR = 1.61; 95% CI: 1.37-1.90; p < 0.001).

Conclusions

Baseline EAs are common in patients with advanced cancers participating in phase I trials. This is the first study to demonstrate the clinical significance of baseline hypoalbuminemia and hyponatremia, which are predictors of development of other EAs in phase 1 patients. G3/4 EAs are adverse prognostic factors of OS independent of serum albumin levels.

Legal entity responsible for the study

The Royal Marsden Hospital NHS Foundation Trust

Funding

None

Disclosure

J. de Bono: Consulting or advisory role: Astex, AstraZeneca, Genentech, Genmab, GSK, Merck, Pfizer, Sanofix. Research funding: AstraZeneca, Genentech, GSK, Sanofi, Janssen. U. Banerji: Receipt of grants/research supports: AstraZeneca, Chugai, Onyx, BTG. Receipt of honoraria or consultation fees: Astex, Karus Therapeutics, Novartis, Vernalis. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

400P - LCZ 696, administered during doxorubicin, trastuzumab or pertuzumab treatment, prevents cardiotoxicity in our in vitro model (ID 4574)

Presentation Number
400P
Lecture Time
13:15 - 13:15
Speakers
  • N. Maurea
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Doxorubicin (DX), Trastuzumab (T) and Pertuzumab (P) are antineoplastic drugs used in the treatment of breast cancer. Adverse cardiovascular events related to anticancer drugs are among the leading causes of morbidity and mortality in cancer patients. Sacubitril-valsartan (LCZ 696) is a combination drug, made up of neprilysin inhibitor sacubitril and angiotensin II receptor blocker valsartan, used for the treatment of heart failure in patients with a reduced ejection fraction. Here, we aim to assess whether LCZ 696, administered during DX, T or P treatment, reduces in vitro anticancer drugs-related cardiotoxicity compared to Valsartan (V), used as a control drug.

Methods

The H9C2 rat cardiomyoblasts were seeded in 96-well plates at a density of 1 x 104 cells/well and incubated at 37 °C with 5% CO2 for 16 hours. After the addition of 200 nM of T, P or DX in the culture medium, cells were incubated for 72 hours. The cells were further treated in the absence or presence of 10 µM of LCZ 696 or V for additional 3 days. Viable cells were counted by trypan blue exclusion test and cell survival was expressed as percentage of viable cells compared to control untreated cells.

Results

LCZ 696 reduced significantly T, P and DX related toxicity in H9C2 cardiomyoblasts as evidenced by the higher percentage of viable cells treated with combinations of T, P or DX with LCZ 696 with respect to cells treated with T, P or DX alone (p < 0.001). V reduced significantly T and DX related toxicity in H9C2 cardiomyoblasts treated with combinations of T or DX and V with respect to the cells treated with T or DX, used as single agents (p < 0.001). However there was no significant reduction of toxicity when H9C2 cells were treated with P + V. Thus, both LCZ 696 and V reduced significantly DX and T related toxicity when administered to H9C2 cardiomyoblasts after the antineoplastic treatment (no significant difference between LCZ 696 and V treatment, p = 0.6). Moreover, LCZ 696 was significantly more effective than V (p < 0.001) in reducing both T and P related toxicity when administered to cultures of H9C2 cardiomyoblasts after antineoplastic treatments.

Conclusions

LCZ 696, administered during DX, T or P treatment, significantly increases the viability of treated cells, thus reducing cardiotoxic effects of these drugs, as demonstrated by our in vitro experiments. The future perspective aims to test LCZ 696 in in vivo models to assess its capability to blunt left ventricular dysfunction after antineoplastic treatments.

Legal entity responsible for the study

Nicola Maurea

Funding

None

Disclosure

M. De Laurentiis: Advisory Board: Novartis, Roche, Pfizer, AstraZeneca, Celgene, Eisai. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

401P - MEK inhibitor retinopathy (ID 5022)

Presentation Number
401P
Lecture Time
13:15 - 13:15
Speakers
  • T. Shukair
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

To evaluate the presence and characteristics of subretinal fluid (SRF) associated with the use of MEK inhibitors in the treatment of systemic cancer MEK retinopathy is described as symmetrical bilateral disease that develops in a time-dependent and dose-dependent manner.

Methods

In this prospective, observational study, collected data from 14 patients with locally advanced or metastatic cancer undergoing treatment with the MEK inhibitor as clinical trials between 2010-2013. They underwent regular ophthalmological examinations including determination of visual function, biomicroscopy, dilated fundoscopy and optical coherence tomography (OCT).

Results

Of the 14 participants, 10 (71%) were men; the mean (SD) age was 65 years (range, 41-80 years). Six (48%) study participants developed SRF during the study period. OCT revealed subfoveal neuroretinal elevation, serous retinal detachments often asymptomatic. In general it solves spontaneously without any apparent functional deficits or changes in structural integrity, and does not require the suspension of the treatment.

Conclusions

The presence of serous retinal detachment in patients undergoing treatment with the MEK inhibitor is common. Visual symptoms were mild and mainly transient and the presence of SRF did not lead to permanent ocular disorder. It is important to investigate all previous ocular disorders and pharmacologic interactions of MEK inhibitor that could associate with ocular effects.

Legal entity responsible for the study

START Madrid

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

402P - A Phase I/II study everolimus in combination with paclitaxel-carboplatin in patients with advanced adenocarcinoma of the stomach (ID 1394)

Presentation Number
402P
Lecture Time
13:15 - 13:15
Speakers
  • H. Loong
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Significant proportion of patients (pts) with adenocarcinoma of stomach (ADCS) present with advanced disease. Paclitaxel (P), either alone, or in combination with carboplatin (C) is well-tolerated, but has modest activity in ADCS. The PI3K-Akt pathway played an important role in cell proliferation and apoptosis in pre-clinical ADCS models. Everolimus (E) is a potent inhibitor of mTOR, a downstream mediator of the PI3K-Akt pathway. Combining chemotherapies with mTOR inhibition may improve outcome

Methods

A single-arm, dose-escalation study of E, in combination with P and C (E+PC) was conducted in pts with metastatic and/or loco-regionally advanced ADCS [NCT01514110]. In the phase 1 portion (P1P), the maximum-tolerated dose (MTD), recommended phase 2 dose (RP2D) and safety of E+PC, were determined using a standard 3 + 3 design. Starting dose (dose level I) was E 5mg/d, P 175mg/m2 and C AUC5 every 3 weeks. Dose-limiting toxicities (DLT) were defined as grade 4 haematological or grade 3 or 4 non-haematological toxicities. Preliminary efficacy of E+PC in pts with ADCS, defined by clinical benefit rate (CBR) (CR+PR+SD for 6wks or more as per RECIST), and survival were determined in the phase 2 portion

Results

30 pts were enrolled (P1P = 12) from Jan 2008 to Nov 2014. In the P1P, 2 DLTs (G5 GI bleeding and G3 joint pain) were experienced at dose level II, thus establishing dose level I as the MTD and RP2D. 21 pts were treated at RP2D. Baseline demographics of phase 2 portion: M/F: 9/12, Median age 54 (range 40-69), ECOG PS 0/1/2: 10/10/1. Prior lines of chemotherapy 0/1/≥2: 7/12/2. Median cycles: 6 (range 1-19). Common related ≥G3 adverse events (AEs) include (%): neutropenia (48%), anaemia (43%), thrombocytopenia (29%), mucositis (10%). Febrile neutropenia occurred in 10% (n = 2) of pts. 18 pts were evaluable for response (5 PR, 9 SD, 4 PD). CBR 77.8% (95% CI 58.6-97.0%). Median PFS and OS was 6.9 and 9.0months (95%CI 3.5 – 7.6; 3.8 – 15.1months) respectively

Conclusions

E+PC administered at RP2D was well-tolerated. Comparing with prior reported series of PC alone, E+PC showed more favorable efficacy and has promising activity in pts with advanced ADCS. Acknowledgement- Supported by Novartis Pharmaceuticals Ltd

Clinical trial identification

NCT01514110

Legal entity responsible for the study

The Chinese University of Hong Kong

Funding

Novartis Pharmaceuticals Ltd.

Disclosure

H.H. Loong: Research Funding: MSD. Advisory: Novartis, Roche. Travel Support: Abbvie, Bayer, Bristol-Myers-Squibb, Novartis, Roche. Speakers Bureau: Abbvie, Bayer. W. Yeo: Advisor: Novartis, Eli Lilly. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

403P - Phase I studies of the novel carcinoembryonic antigen T-cell bispecific (CEA-CD3 TCB) antibody as a single agent and in combination with atezolizumab: Preliminary efficacy and safety in patients (pts) with metastatic colorectal cancer (mCRC) (ID 4001)

Presentation Number
403P
Lecture Time
13:15 - 13:15
Speakers
  • N. Segal
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

CEA-CD3 TCB (RG7802, RO6958688) is a novel T-cell bispecific antibody targeting CEA on tumor cells and CD3 on T cells. Preclinically, CEA-CD3 TCB had potent antitumor activity, leading to increased intratumoral T-cell infiltration and activation, T-cell–mediated tumor cell killing and PD-L1/PD-1 upregulation.

Methods

In 2 ongoing dose-escalation phase I studies, CEA-CD3 TCB is given as monotherapy IV QW (S1) or in combination (QW) with atezolizumab 1200 mg Q3W (S2) in pts with advanced CEA positive (> 20% of tumor cells expressing moderate or high) solid tumors. In S1, 80 pts (70 CRC) were treated at dose levels of 0.05-600 mg; in S2, 45 pts (35 CRC) at 5-160 mg.

Results

At doses ≥ 60 mg (31 evaluable pts with CRC in S1; 14 in S2), CT scans revealed signs of tumor inflammation within 48 h of the first dose, consistent with CEA-CD3 TCB mode of action. 2 (6%) CRC pts in S1 (both microsatellite stable [MSS]) and 3 (21.5%) in S2 (2 MSS CRC, 1 MSI high [out of 2]) had confirmed partial response (PR; RECIST v1.1). Additionally, tumor reduction of − 10% to − 30% (stable disease) was seen in MSS CRC pts (4 [13%] in S1 and 5 [36%] in S2). At weeks 4-6, 9 (29%) CRC pts in S1 and 7 (50%) in S2 had metabolic PR (FDG PET; EORTC criteria). At all doses in S1, the most common related AEs were pyrexia (56%), infusion-related reactions (IRRs; 50%) and diarrhea (40%). In S1 (out of 59 pts > 40 mg), the most common grade ≥ 3 (G3) related AEs were IRRs (24%) and diarrhea (7%). Five pts experienced DLTs: G3 dyspnea, G3 diarrhea, G3 hypoxia, G4 colitis and G5 respiratory failure (G4 and G5 at 600 mg [exceeding MTD]). DLT events were likely associated with tumor lesion inflammation, per investigators. In S2, there was no evidence of new or additive toxicities, with 2 DLTs at 160 mg (1 G3 ALT increase in a pt with liver metastases and 1 G3 maculopapular rash). Updated data will be presented.

Conclusions

Evidence of antitumor activity in advanced CRC and other CEA-expressing tumors was observed during dose escalation with CEA-CD3 TCB monotherapy. Enhanced activity and a manageable safety profile was seen in combination with atezolizumab.

Clinical trial identification

NCT02324257

Legal entity responsible for the study

F Hoffmann-La Roche Ltd.

Funding

F Hoffmann-La Roche Ltd.

Disclosure

N.H. Segal: Advisory board and research funding from Genentech/Roche, MedImmune/AstraZeneca, Bristol-Myers Squibb, Merck and Pfizer. J. Saro: Employee of Roche and stock holder of Roche. I. Melero: Advisory board: Bristol-Myers, Roche-genentech, AstraZeneca, Lilly, Merck Serono, Bayer, Genmab, Alligator, Bioncotech, Tusk Grants from: Roche-Genetech, Bristol-Myers, Bioncotech. A. Marabelle: PI: Roche, Bristol-Myers Squibb, Merck, Pfizer, Lytix, Eisai, Astra Zen Kyowa Kirin Pharma, Novartis, Bristol-Myers Squibb, Symphogen, Genmab, Amgen, Biothera, Nektar, GSK, Oncovir, Seattle Genetics, Flexus Bio, Pierre Fabre, Onxeo, Bayer, Daichii Sankyo, Imaxio, Sanofi, BioNTech. J.M. Cleary: Research funding to his institution from Merrimack Pharmaceuticals, Taiho Oncology, Merck, Roche, Abbvie, Precision Biologics, and Bristol-Myers Squibb. V. Karanikas, S. Bouseida, F. Sandoval, D. Sabanes: Roche employee. S. Sreckovic: Roche employee and in a possession of Roche stock. H.I. Hurwitz: Honoraria: Roche and Lilly Consultant: Roche, Bristol-Myers Squibb, Lilly, Novartis, Incyte, TRACON Pharma, Acceleron Pharma, GlaxoSmithKline, OncoMed Institutional Funding: Roche, GlaxoSmithKline, Novartis, TRACON Pharma, Bristol-Myers Squibb, Regeneron, Lilly, Macrogeneics, NCI. L. Paz-Ares: Scientific Advise to Roche, Bristol-Myers Squibb, MSD, AstraZeneca, Novartis, Lilly, Pfizer and Merck AG. J. Tabernero: Advisory boards for Amgen, Bayer, Boehringer Ingelheim, Celgene, Chugai, Genentech, Lilly, MSD, Merck Serono, Novartis, Pfizer, Roche, Sanofi, Symphogen, Taiho, and Takeda. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

404P - Dose escalation study of vemurafenib with crizotinib or sorafenib in patient with BRAF-mutated advance cancers (ID 3880)

Presentation Number
404P
Lecture Time
13:15 - 13:15
Speakers
  • D. Sakamuri
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

BRAF inhibitors are effective in melanoma and other cancers with BRAF mutations; however, patients ultimately develop therapeutic resistance through activation of alternative signaling pathways such as MET, PDGFR and CRAF. We hypothesized that combining the BRAF inhibitor vemurafenib and MET inhibitor crizotinib or PDGFR/CRAF inhibitor sorafenib can overcome resistance.

Methods

We designed a phase I study (3 + 3 design) to determine the safety of vemurafenib (240-960 mg PO BID q 28 days) with crizotinib (250 mg PO daily or BID q 28 days) in arm A or sorafenib (200 mg PO daily to 400mg PO BID q 28 days) in Arm B in patients with BRAF-mutant advanced cancers. Endpoints included maximum tolerated dose (MTD), dose limiting toxicities (DLT), safety, response (RECIST 1.1) and plasma cell-free DNA mutation analysis.

Results

Thirty-six patients (arm A, 13; arm B, 23), median number of 3 prior therapies (29 [81%] had prior BRAF/MEK inhibitors) were treated. Patients (melanoma 17/36, 47%; papillary thyroid cancer 5/36, 14%; colorectal cancer 3/36, 8%; lung adenocarcinoma 2/36, 6%; other 9/36, 25%) had BRAF V600E (30), V600K (3) or other BRAF mutations (3). Vemurafenib 720mg BID with crizotinib 250mg daily and vemurafenib 720 mg BID with sorafenib 400mg/200mg were identified as MTDs. DLTs included grade (G) 3 rash (2) in arm A and G3 rash and G3 hypertension in arm B. Other G3 treatment related toxicities were G3 fatigue (2), G3 anemia (1), G3 thrombocytopenia (1), G3 neutropenia (1), G3 thromboembolic event (1) in arm A and G3 hypertension (1), G3 headache (1), G3 diarrhea (2), G3 intraocular inflammation (1) in arm B. In Arm A, 3 of 13 (23%) patients (melanoma refractory to BRAF monotherapy [2] and lung adenocarcinoma) attained a partial response (PR). In Arm B, 4 of 23 (17%) patients (ovarian cancer refractory to MEK inhibitor, melanoma, lung adenocarcinoma, papillary thyroid cancer) attained a PR. Optional longitudinal collection of plasma cfDNA to assess clonal evolution was performed and will be presented at the meeting.

Conclusions

Vemurafenib in combination with crizotinib or sorafenib is well tolerated with encouraging activity including patients previously treated with BRAF/MEK inhibitors.

Clinical trial identification

NCT01531361

Legal entity responsible for the study

MD Anderson Cancer Center

Funding

National Center for Advancing Translational Sciences (grant no. UL1 TR000371); National Institutes of Health through MD Anderson’s Cancer Center Support Grant (P30 CA016672)

Disclosure

F. Janku: Research funding: Novartis, Genentech, BioMed Valley Discoveries. Scientific Advisory Board: Novartis. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

405P - Hepatic functional imaging and genomics to predict irinotecan pharmacokinetics and pharmacodynamics: The PREDICT IR study (ID 2161)

Presentation Number
405P
Lecture Time
13:15 - 13:15
Speakers
  • M. MICHAEL
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Body surface area-based dosing of Irinotecan (IR), has not accounted for its significant pharmacokinetic (PK) and pharmacodynamic (PD) variability. Given IR’s unique metabolism, hepatic functional nuclear imaging (HNI) with probes for hepatic transporters correlated with its PK. This study further evaluated the utility of HNI combined with extensive excretory/metabolic/PD pharmacogenomics (PG) to predict IR PK and PD in patients (pts) treated with FOLFIRI to enable dose individualization.

Methods

Eligible pts had advanced colorectal cancer, suitable for 1st/2nd-line FOLFIRI± Bevacizumab. Pts had blood analyzed by Affymetrix DMET™ Plus Array and additional SNPs were genotyped. For HNI, pts were given IV 250MBq 99mTc-IDA and imaging data analyzed for hepatic extraction/excretion parameters (clearance [CL], 1hour retention [1hRET], deconvulutional CL [DeCL], hepatic extraction fraction [HEF]). Pts treated with chemotherapy, q2-weekly, and restaged after 4 cycles. Blood taken for IR and metabolite (SN38, SN38G) analysis on day 1 cycle 1, PK parameters derived by non-compartmental analysis. Statistical correlations were evaluated between (i) IDA HNI and (2) PGs, with IR PK, toxicity, objective response (ORR) and progression-free survival (PFS).

Results

32 pts analysed, 31 pts completed 4 cycles. (1) PK correlates: (a) HNI CL and 1hRET with SN38 Metabolic CL, (P = 0.04) and (b) HNI DeCL with IR AUC(0-∞) (P = 0.04). (2) Grade 3+ diarrhea (N = 4, 13%) predicted by SN38 AUC(0-∞) andMetabolic CL (P = 0.04), and gene variants for SCL22A2 and -28A3, ABCC2, UGT2B17, CYP2C18 and DPYD (P < 0.05) (3) Grade 3+ neutropenia (N = 9, 28%) predicted by SN38 PK exposure (P < 0.02), HNI CL and 1hRET (P < 0.0001) and variants for SLC7A7-, SLC22A2-, CHST1-, UGT1A1-, -2B7, ABCB1. (4) ORR (N = 6, 20%) predicted by Methylene tetrahydrofolate reductase (MTHFR) 677C > T (P = 0.002), SN38 exposure (P < 0.003), and variants in metabolic/transporter genes (P < 0.05). (5) PFS by SN38 PK exposure, MTHFR 677C > T, HNI CL, HNI HEF and variants in PK genes (P < 0.05).

Conclusions

Hepatic functional imaging with extensive pharmacogenomics correlated with Irinotecan PK and PD enabling the future development of nomograms to individualize its dosing.

Clinical trial identification

Australian Clinical Trials Registry: ACTRN12610000898055

Legal entity responsible for the study

Peter MacCallum Cancer Centre

Funding

Australian Federal Government: National Health and Medical Research Council Project Grant

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

406P - Ongoing phase 1 trial of SL-801, a novel XPO-1 inhibitor, in patients with advanced solid tumors; Interim results (ID 5010)

Presentation Number
406P
Lecture Time
13:15 - 13:15
Speakers
  • J. Wang
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Exportin-1 (XPO1) is a critical nuclear export protein involved in nuclear-cytoplasmic transport and is overexpressed in multiple malignancies. SL-801 is a novel oral small molecule inhibitor of XPO1 and has shown potent in vitro and in vivo anti-tumor activity against a broad range of hematologic and solid malignancies. SL-801 has been shown to reversibly inhibit XPO1, which may translate to selective activity and potential safety benefits. Study SL-801-0115 is a first-in-human, dose-escalation study in adult patients with locally advanced, unresectable or metastatic solid tumors that are resistant to or relapsed following available standard systemic therapy. Preliminary interim results in the ongoing dose-escalation Phase 1 clinical study are reported.

Methods

The objective of this multicenter, dose-escalation phase 1 trial is to evaluate the safety and tolerability of SL-801, identify the maximum tolerated dose (MTD), and assess pharmacokinetics (PK) with increasing doses. SL-801 is administered daily on Day 1 through Day 4 and Day 8 through Day 11 every 21 days. The starting dose cohort was 5 mg and has reached 35 mg to date.

Results

As of 3/31/17, 19 heavily pretreated (range: 1-11 prior therapies) adult patients with advanced solid tumors have received SL-801 (6 Females, 13 Males). Median age is 62 years (range: 39-75). MTD has not been reached. Median follow-up is 1.4 months (range: 0.1-4.9). Pharmacokinetic analyses are ongoing. The most frequent treatment-related Grade 1-2 AEs were fatigue and nausea (26%), diarrhea (16%), and myalgia, vomiting and decreased appetite (11% each). Grade 3 AEs, possibly treatment-related, included diarrhea (n = 1; 5 mg) and acute renal injury (n = 1; 30 mg). There were no grade 4 or 5 treatment-related events. Three patients achieved stable disease, for 3-7 cycles, by investigator assessment.

Conclusions

The initial dose-escalation of SL-801 appears to be well tolerated to date in patients with advanced solid malignancies. Enrollment continues in this ongoing Phase 1 trial and updated safety and efficacy data will be presented. Clinical trial information: NCT02667873.

Clinical trial identification

NCT02667873

Legal entity responsible for the study

Stemline Therapeutics

Funding

Stemline Therapeutics

Disclosure

A. Olguin, J. Bullington, S. Shemesh, J. Chen: Stemline Therapeutics: employment, stock options. C. Brooks: Stemline Therapeutics: employment, stock options, patents. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

407P - Antitumor efficacy of triple monoclonal antibody inhibition of epidermal growth factor receptor (EGFR) with MM-151 in EGFR-dependent and in cetuximab-resistant human colorectal cancer cells (ID 2236)

Presentation Number
407P
Lecture Time
13:15 - 13:15
Speakers
  • S. Napolitano
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Novel and more efficient anti-EGFR drugs capable to overcome acquired resistance to first generation of anti-EGFR inhibitors needs to be investigated.

Methods

MM151 is a mixture of three different monoclonal IgG1 antibodies directed toward three different, non-overlapping, epitopes of the EGFR. We performed an in vivo study by using human CRC cell lines (SW48, LIM 1215 and CACO2) which are sensitive to EGFR inhibitors, in order to evaluate the activity of MM-151 as compared to standard anti-EGFR mAbs, such as cetuximab, as single agent or in a sequential strategy of combination MM-151 with irinotecan (induction therapy) followed by MM-151 with a selective MEK1/2 inhibitor (MEKi) (maintenance therapy). Furthermore, the ability of MM-151 to overcome acquired resistance to cetuximab has been also evaluated in cetuximab-refractory CRC models.

Results

MM151 shown stronger antitumor activity as compared to cetuximab. The maintenance treatment with MM-151 plus MEKi resulted the most effective therapeutic modality. In fact, this combination caused an almost complete suppression of tumor growth in SW48, LIM 1215 and CACO2 xenografts with a mean tumor volume of 13 mm3, 13 mm3 and 75 mm3, respectively at 30 week. Moreover, in this treatment group, mice with no evidence of tumor were more than double as compared to single agent treated mice. Its superior activity has also been demonstrated, in cetuximab-refractory CRC models.

Conclusions

These results provide experimental evidence that more efficient and complete EGFR blockade may determine better antitumor activity and could contribute to prevent and/or overcome acquired resistance to EGFR inhibitors.

Legal entity responsible for the study

Università degli studi della Campania, “Luigi Vanvitelli”

Funding

Associazione Italiana per la Ricerca sul Cancro (AIRC)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

408P - Validation of the Royal Marsden Hospital (RMH) prognostic score on an enriched early treatment line cohort for phase I trial patients (ID 3794)

Presentation Number
408P
Lecture Time
13:15 - 13:15
Speakers
  • M. Blanco Codesido
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The RMH score has been validated to predict survival in different populations of patients starting phase I clinical trials. Most of the populations where it has been validated are of heavily treated patients that lack other treatment options. The type of phase I trials is changing and we aimed to validate the score in a new cohort with more patients treated on an early line, rahter than the usual classic phase I trials heavily pretreated patient population.

Methods

We analyzed the RMH score in the patients treated in our center in a phase I trial between 2012 and 2017. We collected demographics data, overall survival after starting the trial, the RMH score (albumin, LDH, and number of metastatic sites) for all patients and the treatment line. We considered a late line anything over two treatment lines and in any case if the patient did not have any other treatment available depending on the tumor type. An early line was the first or second treatment line when the patient did have further lines available.

Results

We treated 77 patients on a phase I trial in our institution, 23 males and 54 females. Mean age was 55 years (26-77). RMH score was (0/1/2/3) in (31/23/20/3) patients. Thirty-three patients were treated on an early line. Median survival for low score (0/1) was 639 days and for high score (2/3) was 327 days p = 0.0834. The mean survival for patients with low RMH score was higher than those with a high RMH score in every treatment line, although due to the low number of patients in some of those categories the difference was not significant.

Conclusions

The RMH score did predict well the overall survival in our patients. The survival times in our institution are higher than those previously published, probably due to the inclusion of patients on earlier treatment lines than those used before to calculate and validate the score. Our findings support the use of the RMH score for the selection of patients entering phase I trials irrespectively of the design of the trial (early vs. late line).

Legal entity responsible for the study

Medical Oncology Department, Hospital General Universitario Gregorio Marañón

Funding

Instituto de Investigación Sanitaria Hospital Gregorio Marañón

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

409P - Updated results of phase 1 study of DS-8201a in patients with HER2 expressing non-breast, non-gastric malignancies (ID 4868)

Presentation Number
409P
Lecture Time
13:15 - 13:15
Speakers
  • J. Tsurutani
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

DS-8201a is a HER2 targeting antibody-drug conjugate of high drug to antibody ratio (7 to 8) with a novel linker and topoisomerase I inhibitor. In preclinical studies, DS-8201a showed a broad antitumor spectrum, including efficacy against low HER2 expressing breast cancer (BC) and HER2 expressing non-gastric and non-breast cancers. The current phase 1 trial includes dose escalation (Part 1) and expansion (Part 2) including BC, gastric cancer (GC) and other HER2 expressing solid tumors.

Methods

Part 1 used a mCRM to identify the recommended dose in patients (pts) with BC or GC. Part 2 was designed to evaluate the safety and efficacy in 4 expansion cohorts: HER2 positive BC, HER2 positive GC, low HER2 expressing BC, and other solid tumors expressing HER2. HER2 expression was determined by IHC, FISH, NGS or other platforms. Adverse events (AEs), objective response rate (ORR), and disease control rate (DCR: CR + PR + SD) were assessed.

Results

Twenty four pts in Part 1 and 113 pts in Part 2 were enrolled. 24 of 113 pts were HER2 expressing solid tumors other than BC and GC. DS-8201a was administered up to 8.0 mg/kg in Part 1, and dose level of 6.4 mg/kg IV every 3 weeks was chosen. DLTs were not observed in the study. In the updated Part 1 results, confirmed ORR was 35%, DCR was 91% (BC: 88%, GC: 100%), and the median duration of treatment was ≥32 weeks in heavily pretreated BC and GC pts. Non-BC and non-GC cohort consists of 11 CRC, 5 NSCLC, 4 salivary gland, 2 Paget’s disease, 1 cholangiocarcinoma and 1 esophageal cancer. ORR including under confirmation and DCR were 33% and 91%, respectively in evaluable 12 pts. Two out of 5 evaluable pts with CRC and 2 out of 4 evaluable pts with salivary gland achieved PRs. Of all pts in this phase 1 study, the most common AEs of any grades were nausea (≥Gr1 60%; ≥Gr3 2%), decreased appetite (≥Gr1 55%; ≥Gr3 4%), vomiting (≥Gr1 30%; ≥Gr3 0%) and platelet count decreased (≥Gr1 30%; ≥Gr3 9%). Updated phase 1 results will be presented.

Conclusions

DS-8201a was well tolerated and is remarkably active in pts with heavily pretreated HER2 expressing BC and GC with durable disease control. Promising efficacy in HER2 expressing other tumors was observed and warrants further investigation.

Clinical trial identification

NCT02564900

Legal entity responsible for the study

Daiichi Sankyo CO., LTD.

Funding

Daiichi Sankyo CO., LTD.

Disclosure

J. Tsurutani: Paid honoraria directly by Eisai, Kyowa Hakko Kirin, Chugai, AstraZeneca, Novartis, Taiho. Paid for any consulting or advisory role by Novartis, Daiichi Sankyo, MSD, Lilly, Roche. Conducted research project funded by Daiich Sankyo, Roche, Lilly, MSD. T. Doi: Paid for consulting by Lilly, Chugai, Kyowa, Behringer, Novartis, MSD, Daiichi Sankyo, Amgen. Conducted study funded by Taiho, Novartis, Merck, Astellas, MSD, Janssen, Boehringer, Takeda, Pfizer, Lilly, Sumitomo, Chugai, Bayer, Kyowa, Daiichi Sankyo. H. Iwata: Paid honoraria directly by Daiichi Sankyo, Chugai, AstraZeneca. Paid for consulting or advisory role by Daiich Sankyo and Pfizer. Conducted research project funded by Daiich Sankyo, Chugai, AstraZeneca, Pfizer, Novartis, Eisai, MSD, Lilly. S. Takahashi: Paid honoraria directly by Eisai, Astellas, Taiho, Bayer, Daiichi Sankyo, Pfizer, AstraZeneca. Paid for consulting or advisory role by Chugai Astellas. Conducted study funded by Eisai, Chugai, MSD, Lilly, AstraZeneca, Novartis, Taiho, Daiichi Sankyo. K. Tamura: Conducted study funded by Daiichisankyo, MSD, pfizer, AstraZeneca. K. Shitara: Paid for consulting or advisory role by Chugai Pharma, Takeda, Bayer, Lilly, Bristol-Myers, Squibb. Conducted study funded by Dainippon Sumitomo Pharma, Lilly, MSD, Sanofi, Daiichi Sankyo, Taiho Pharmaceutical, Bayer, Chugai. H. Taniguchi: Honoraria for speaking or writing engagements from Takeda, Chugai, Merck Serono, Taiho, Bayer, Lilly, Yakult. Financial support for clinical trials or research found by Otsuka, Boehringer Ingelheim, MSD Oncology, Takeda. B. Li: Consulting and/or Advisory Board for Genentech, ThermoFisher Scientific, Biosceptre Australia. Principle Investigator of clinical trials supported by Genentech, AstraZeneca, BioMedValley. A. Shimomura: Conducted study funded by AstraZeneca. Y. Sato, K. Akiyama, Y. Fujisaki, C. Lee, A. Yver: Employee of Daiichi Sankyo. K. Nakagawa: Paid for any consulting or advisory role by Astellas. Ono, AstraZeneca, Boehringer Ingelheim, Daiichi Sankyo. Conducted study funded by GlaxoSmithKline, Yakult, AstraZeneca, PAREXEL, Kyowa Hakko, Otsuka, Pfizer, Astellas, Daiichi Sankyo. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

410P - Phase I study of the investigational, oral pan-RAF kinase inhibitor TAK-580 (MLN2480) in patients with advanced solid tumors (ST) or melanoma (MEL): Final analysis (ID 4583)

Presentation Number
410P
Lecture Time
13:15 - 13:15
Speakers
  • A. Olszanski
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

MAPK pathway mutations leading to signaling hyperactivation are common in many ST; as RAF kinases play a key role in MAPK signaling, they represent a valid target for therapy. As a pan-RAF inhibitor, TAK-580 is differentiated from approved BRAF-specific RAF inhibitors. Here we report the expanded cohort data from a single-agent, first-in-human study of TAK-580 (NCT01425008).

Methods

Patients with advanced ST or inoperable stage III/IV MEL received TAK-580 Q2D or QW in 28-d cycles. Primary objectives were safety and maximum tolerated doses [MTD] of TAK-580; secondary objectives included preliminary antitumor activity, PK and PD effects. Safety and PK were compared between 2 MTD regimens (200 mg Q2D vs 600 mg QW). Preliminary antitumor activity of TAK-580 Q2D vs QW was evaluated in NRAS-mutation positive (mut) MEL. Activity and efficacy (PFS) were assessed in BRAF-mut MEL. Plasma PK were assessed pre- and post-dose between d1 and d28 of cycle 1 (C1). Tumor biopsies were taken at screening and post-dose on d21 or 22 of C1. Disease assessments were performed at baseline and every 2 cycles thereafter.

Results

80 patients received TAK-580 200 mg Q2D (60 MEL + 20 PK-evaluable ST) and 19 MEL patients received 600 mg QW. DLTs observed were: periorbital edema and maculopapular rash (280 mg Q2D); rash and hyperbilirubinemia (800 mg QW). For Q2D and QW, 41% and 32% of patients, respectively, had Gr ≥ 3 drug-related adverse events (AE); 19% and 11% discontinued due to AEs. Total weekly exposure (AUC168h) after TAK-580 600 mg QW was comparable to 3-fold AUC48h after 200 mg Q2D. Of 14 NRAS-mut MEL Q2D patients, 1 achieved PR (1.5 mos). In comparison, none of 17 NRAS-mut MEL QW patients had an objective response. 50% of the 16 BRAF-mut MEL Q2D patients achieved a PR with a median PFS of 4.6 mos (range 1.0–40.8).

Conclusions

The safety and PK profiles of TAK-580 Q2D and QW at MTD were acceptable. QW dosing improved safety but not efficacy over Q2D dosing. PD results were consistent with the proposed mechanism of action of TAK-580 with observed RAF pathway inhibition. These data support the use of QW dosing in the assessment TAK-580 given in combination.

Clinical trial identification

NCT01425008

Legal entity responsible for the study

Millennium Pharmaceuticals, Inc., Cambridge, MA, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited

Funding

Millennium Pharmaceuticals, Inc., Cambridge, MA, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited

Disclosure

A.J. Olszanski: Consulting or advisory role: Merck, Takeda, BMS, Kyowa Hakko Kirin, G1 Therapuetics; Research funding: Takeda, Immunocore, EMD Serono, Amgen, Incyte, Kyowa Hakko Kirin, Lilly, Advaxis, Mirati Therapuetics, Ignyta, Novartis, Pfizer, BMS, Kura; Travel/accommodation/expenses: Takeda, Churchill Pharmaceuticals, Kyowa Hakko Kirin, G1 Therapuetics. R. Gonzalez: Consultant: Bristol-Myers Squibb, Novartis, Genentech. Research support: Merck, Novartis, Genentech, Bristol-Myers Squibb, Incyte, Syndax, Takeda. P. Corrie: Advisory boards; Novartis, Pierre Fabre, Bristol-Myers Squibb, MSD, Celgene. Research funding: Celgene. Speaker honoraria: MSD, Novartis. M. Middleton: Consulting or advisory role: GSK, BMS, Amgen, Merck, Roche (all compensated); Clovis, Immunocore (both uncompensated); Travel/accommodation/expenses: Roche, Merck; Corporate-sponsored research: GSK, AZ, Eisai, Clovis, BMS, Amgen, Roche, Merck, Vertex, Immunocore, Pfizer, Medimmune. P. Lorigan: Advisory board: GSK, Novartis, Roche, Bristol-Myers Squibb, Merck, Amgen. Travel/accommodation/expenses: Bristol-Myers Squibb, MSD. A. Daud: Stock ownership: OncoSec, Inc.; Advisory board or board of directors: Novartis, Merck, Pfizer, Genentech; Corporate-sponsored research: Merck, Pfizer, Genentech, BMS. S. Zhang, E. Hoberman: Employment: Millennium Pharmaceuticals. Inc. B. Bahamon, L. Rangachari, M. Kneissl: Employment: Millennium Pharmaceuticals, Inc. D. Rasco: Corporate-sponsored research: Takeda Oncology. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

411P - Pharmacological activity of CB-103: An oral pan-NOTCH inhibitor with a novel mode of action (ID 5472)

Presentation Number
411P
Lecture Time
13:15 - 13:15
Speakers
  • D. Weber
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

NOTCH signalling is a developmental pathway known to play critical roles during embryonic development as well as for regulation of self-renewing tissues. Aberrant activation of NOTCH signalling leads to deregulation of the self-renewal process resulting in sustained proliferation, invasion and metastasis, all of which are hallmarks of cancer. When the NOTCH pathway is inappropriately activated by genetic lesions (over expression of NOTCH ligands/receptors, GOF mutations in NOTCH receptors as well as chromosomal translocations), it becomes a major driver for NOTCH-dependent cancers and resistance to standard of care treatment. Several therapeutic approaches have been utilized to block NOTCH signalling, e.g. a) the use of monoclonal blocking antibodies (mAbs) against NOTCH ligands and receptors and b) the use of small molecule gamma-secretase inhibitors (GSIs). Here we report the pharmacological characterization of CB-103, a first-in-class orally-active small molecule, protein-protein interaction inhibitor of the NOTCH transcriptional activation complex.

Methods

Primary pharmacodynamic (PD) studies were conducted to investigate CB-103 in relation to its desired therapeutic effect for treating advanced or metastatic haematological and solid tumour malignancies as NOTCH pathway inhibitor. Regarding the PD effect, in vitro studies demonstrated for CB-103 a dose-dependent decrease in NOTCH signalling activation with a unique mechanism compared to GSIs and mAbs. In a panel of > 120 cell lines of various malignancies CB-103 was active on a subset of 24 cancer cell lines, including different solid tumours (breast, lung, sarcomas), lymphomas and leukaemias.

Results

Moreover, CB-103 demonstrated anti-NOTCH activity in the Triple-Negative Breast Cancer HCC1187 cell line, being resistant to GSIs due to a NOTCH2 chromosomal translocation. In addition, CB-103 exhibited anti-tumour efficacy in multiple in vivo models and patients derived xenograft models.

Conclusions

Safety pharmacology and toxicology studies have been completed and revealed an excellent non-clinical safety profile of CB-103. A first-in-human Phase I/IIA clinical study in advanced solid tumours and haematological malignancies is under preparation.

Legal entity responsible for the study

Cellestia Biotech AG

Funding

Cellestia Biotech AG

Disclosure

D. Weber: Chief Medical Officer of Cellestia, co-founder, stock ownership. R. Lehal: Chief Scientific Officer, co-founder, stock ownership. J-P. Bourquin: Medical advisory board Cellestia. M. Bauer: Chief Executive Officer, co-founder, stock ownership. M. Murone: Chief Operating Officer, co-founder, stock ownership. F. Radtke: Chairman of the Board, co-founder, stock ownership. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

412P - Design and development of potent E1 ubiquitin activating enzyme inhibitor, CPL-410-005, as novel anticancer therapy (ID 2927)

Presentation Number
412P
Lecture Time
13:15 - 13:15
Speakers
  • A. Stanczak
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The ubiquitin-proteasome system is crucial in tumorigenesis. The division rate of cancer cells, thus protein synthesis, is increased in comparison to normal ones, what sensitizes tumors for any protein changes. Proteasome inhibitor - bortezomib was the first inhibitor registered in treatment of blood cancers. However, drugs beneficial for solid tumors are still missing. Therefore targeting of E1 enzyme as a start of UPS pathway may serve as a promising anticancer therapy.

Methods

We have designed a novel E1 small molecule inhibitor, CPL-410-005. The inhibitory potency of compound was assessed on purified E1 enzyme, using biochemical assay. Assays to measure cellular poliubiquitylation or ubiquitin-like modifications level were developed. The compound’s biological activity and selectivity was evaluated in a number of cancer cells using cell viability assays, Western Blot and flow cytometry, analyzing programmed cell death, unfolded protein response or cell cycle inhibition.

Results

CPL-410-005 inhibits E1 enzyme with greater potency than MLN7243. This results in cellular polyubiquitylation inhibition, while the impact on other ubiquitin-like modifications (neddylation, sumoilation) is minor. Tumor proliferation rate inhibition was pronounced in reference to the non-malignant cells [IC50 values of 20 nM for HCT-116 cells vs IC50 values of 400 nM for HEK293 cells]. Moreover, a higher level of unfolded protein response or programmed cell death was observed in cells treated with CPL-410-005 in reference to MLN7243 (5% apoptotic cells vs 30% for MLN7243 vs CPL-410-005, respectively). In case of CPL-410-005, apoptosis rate was higher in tumor than in normal cells (80% apoptotic cells vs 20%, respectively). A high throughput study was performed, to determine the activity of CPL-410-005 on 120 human tumor cell lines, showing that >85% tested cell lines responded with IC50 value below 100nM. The initial in vivo studies on tumor human xenografts are ongoing.

Conclusions

We have designed and evaluated in vitro a potent E1 inhibitor - CPL-410-005, which shows promising in vitro activity. Further preclinical studies are necessary to develop this compound as a novel anticancer therapy.

Legal entity responsible for the study

Celon Pharma

Funding

Celon Pharma

Disclosure

A. Stanczak, A. Górnicka, B. Stypik, M. Mroczkiewicz, J. Pieczykolan, K. Dubiel: Full-time employee of Celon Pharma S.A., Lomianki, Poland. M. Wieczorek: Chief Executive Officer of Celon Pharma S.A., Lomianki, Poland.

Collapse
Poster display session Poster Display session

413P - RX-3117, a novel hypomethylating agent, shows promising therapeutic activity in combination with nab-paclitaxel and check-point inhibitors in preclinical models (ID 5044)

Presentation Number
413P
Lecture Time
13:15 - 13:15
Speakers
  • J. Frank
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

A novel nucleoside analogue, RX-3117, is being evaluated in a Phase IIa study in patients with advanced pancreatic and bladder cancer. RX-3117 shows promising antitumor activity in xenografts including patient-derived xenografts resistant to gemcitabine. Here we demonstrate the preclinical effects of combination therapy with RX-3117 + Abraxane or anti-PD1 immunotherapy.

Methods

One colorectal (MC38), pancreatic (Pan02) syngeneic xenograft and patient-derived pancreatic (CTG-0723) xenograft model were exposed to 60 mg/kg RX-3117 po, 5 days on, 2 days off for three weeks. Pan02 and MC38 received RX-3117 alone or in combination with 100ug anti-PD1, ip. PDx CTG-0723 received one cycle of RX-3117, followed by a second cycle of RX-3117 + 10 mg/kg Abraxane, iv. MC38 tumor-infiltrating lymphocytes were measured at days 5 and 12 with RX-3117.

Results

In MC38 at day 28, RX-3117 or anti-PD1 showed TGIs of 90% and 93%, whereas the combination showed 99% TGI. Differences were also observed in TILs. Relative to vehicle (CD4+:10.6+/-1.6, CD8+: 8.6+/-1.1), %CD4 + (17.4+/-1.4) and CD8+ cells (12.3+/-1) increased. %MDSCs decreased on Day 5 in blood (42+/-7.7 vs 29+/-6). %CD8+ increased (9.6+/-3.3 vs 12.3+/-3.2) and %MDSC decreased (15.4 +/- 3.7 vs 10.6 +/- 3.3) in tumor on Day 12. In Pan02, RX-3117 + anti-PD1 resulted in a day 32 TGI of 60%. Anti-PD1 alone had a day 32 28% TGI. In CTG-0723, the first cycle of RX-3117 at 10, 30 and 60 mg/kg produced TGIs of 33%, 46% and 77%. The second cycle, RX-3117 + Abraxane, day 46 TV showed TGIs of 55%, 58% and 83%.

Conclusions

We demonstrate the antitumor effect of RX-3117 as a single agent and in combination with Abraxane or anti-PD-1. The combination of RX-3117/anti-PD1 in MC38 produced 7 tumor-free survivors out of 10 compared to 2 of 10 by anti-PD1 alone, indicating RX-3117 may mobilize the right population of lymphocytes to enable anti-PD-1 to work more effectively. In Pan02, RX-3117 exhibited better TGI than anti-PD-1. In CTG-0723, the combination of RX-3117 and Abraxane showed additive TGI. These studies demonstrate the therapeutic potential of RX-3117 in multiple cancers and validate the combination of RX-3117 with anti-PD1 in several cancer types.

Legal entity responsible for the study

Rexahn Pharmaceuticals, Inc.

Funding

Rexahn Pharmaceuticals, Inc

Disclosure

J. Frank, Y.B. Lee, D.J. Kim: Employee of Rexahn Pharmaceuticals. E. Benaim: Officer at Rexahn Pharmaceuticals, employee, stock-holder.

Collapse
Poster display session Poster Display session

414P - A phase Ib trial of JX-594 (Pexa-Vec), a targeted multimechanistic oncolytic vaccinia virus, in combination with low-dose cyclophosphamide in patients with advanced solid tumors (ID 5332)

Presentation Number
414P
Lecture Time
13:15 - 13:15
Speakers
  • M. Toulmonde
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

JX-594 (Pexa-Vec) is a targeted oncolytic vaccinia virus designed to selectively replicate in and destroy cancer cells with epidermal growth factor receptor (EGFR)/ras pathway activation. Direct oncolysis plus GM-CSF expression is accompanied by tumor vascular disruption and anti-tumoral immunity. JX-594 was well-tolerated intravenously (IV) and intratumorally (IT). Given the immunomodulatory effects of low-dose cyclophosphamide (CP), anti-tumor synergy is predicted with JX-594.

Methods

CP was delivered orally at the dose of 50 mg BID one week on one week off. JX-594 was delivered IV at day 8 of each day 28-cycle. 2 dose levels of JX-594 were explored: 3.108 and 1.109 plaque forming units (pfu). The primary objective of the study was to determine the safety of JX-594 in combination with low-dose CP in patients with advanced solid tumors. Secondary objectives include response rates, PFS, pharmacokinetics and pharmacodynamics.

Results

Ten patients entered the study. 9 were evaluable for safety. No dose limiting toxicity was observed. The combination regimen was well-tolerated. The most frequent adverse events were grade 1-2 fever/transient flu-like symptoms (n = 10), grade 1-2 nausea (n = 5), grade 1-2 anemia (n = 4) and grade 1-2 fatigue (n = 4). 2 patients (breast cancer, ovarian cancer) had stable disease as best overall response.

Conclusions

IV JX-594 was well-tolerated in combination with low-dose CP. PK and PD (immunological profiling) will be presented at the meeting. Two phase 2 studies are ongoing in patients with advanced HER2 negative breast cancer and advanced soft-tissue sarcomas, respectively.

Clinical trial identification

NCT02630368

Legal entity responsible for the study

Institut Bergonié

Funding

INCA

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

415P - Population pharmacokinetic analysis of OT-101 (trabedersen) in patients with advanced tumors (ID 5455)

Presentation Number
415P
Lecture Time
13:15 - 13:15
Speakers
  • W. Wang
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

OT-101 (Trabedersen) is a phosphorothioate antisense oligodeoxynucleotide specifically inhibiting the expression of transforming growth factor-beta 2 (TGF-b2), whose overexpression is a pivotal factor for malignant progression in solid tumors. In the clinical Phase I/II study, plasma pharmacokinetic (PK) profile of OT-101 administered intravenously was evaluated in patients with advanced tumors. A population PK model was built to further understand the factors contributing to the variability in PK of OT-101.

Methods

A total of 61 patients with pancreatic cancer (n = 37), malignant melanoma (n = 19), or colorectal carcinoma (n = 5) were treated with OT-101 with escalating doses in 2 treatment schedules (1st schedule: 7-days-on/7-days-off; 2nd schedule: 4-days-on/10-days-off; up to 10 cycles). The plasma concentration data of OT-101 were analyzed using nonlinear mixed-effect modeling (Phoenix NLME 7.0). The influence of age, gender, body mass index (BMI), body weight (BW), cancer type, treatment schedule, creatinine clearance (CrCl) and estimated glomerular filtration rate (eGFR) as covariates on PK was evaluated.

Results

With exclusion of protocol deviations, the final analysis dataset contained 92 patient cycles and 1188 plasma samples. Twenty-six patient cycles were from 7-days-on/7-days-off schedule and 66 were from 4-days-on/10-days-off schedule. The concentration time course of OT-101 was best described by a two-compartment model with combination of additive and multiplicative error. The estimates of PK parameters were as follows: total body clearance, 41.79 mL/hr; distribution volume of the central compartment, 6.30 L; inter-compartmental clearance, 4.02 L/hr; distribution volume of the peripheral compartment, 5965.83 L. eGFR were identified as the covariates on OT-101 central compartmental clearance, with KeGFR as 11.45.

Conclusions

The PK profile of OT-101 was best described by a two-compartment model. The model will be used with the sparse PK samples collected from the planned phase 3 clinical trial to calculate exposure measures for use in subsequent PK-PD analysis of efficacy.

Legal entity responsible for the study

Oncotelic

Funding

None

Disclosure

W. Wang, V. Trieu, L. Hwang: Employee of Oncotelic Inc. K. Ng, D. Nam: Employee of Autotelic Inc.

Collapse
Poster display session Poster Display session

416P - An open-label, multicenter phase 1b study of radium-223 + paclitaxel in cancer patients with bone metastases (ID 2988)

Presentation Number
416P
Lecture Time
13:15 - 13:15
Speakers
  • R. Geva
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Concomitant radium-223 (Ra-223) and chemotherapy is a possible option for cancer patients (pts) with bone metastases (mets). Both treatments impact hematologic parameters, and myelosuppression risk during coadministration is unknown. This phase 1b study (NCT02442063) in cancer pts with bone mets evaluated the safety of Ra-223 + paclitaxel (PTX) and the mode of interaction between treatments regarding myelosuppression.

Methods

Eligible pts had a confirmed malignant solid tumor with ≥ 2 bone mets and were candidates for PTX treatment. Treatment included 7 PTX cycles (90 mg/m2 IV per wk as per local standard of care; 3 wk on/1wk off) combined with 6 Ra‑223 cycles (55 kBq/kg IV; 1 injection every 4 wk, starting at PTX cycle 2). The primary end point was percentage of pts with neutropenia and thrombocytopenia during treatment with Ra‑223 + PTX (cycles 2 and 3) vs PTX alone (cycle 1). A previously developed dose-exposure-response model describing the time course of PTX and Ra-223–induced suppression of absolute neutrophil counts was used to evaluate the mode of interaction (additive or synergistic) between Ra-223 and PTX.

Results

Of 22 enrolled pts, 15 were treated; 13 completed cycles 1-3 and were included in the pharmacodynamics analysis. Tumors in treated pts were breast (7 pts), prostate (4 pts), bladder (1 pt), non‒small cell lung (1 pt), myxofibrosarcoma (1 pt), and neuroendocrine (1 pt). 7 pts had received ≥ 3 prior chemotherapy regimens. In the 13 pts who completed cycle 3, grade 3 neutropenia rates in cycles 2 and 3 were 31% and 8%, respectively, vs 23% in cycle 1; there were no cases of grade 4 neutropenia or grade 3/4 thrombocytopenia. No pts discontinued treatment due to toxicity from the treatment combination. Safety data for the breast cancer pt subset will be presented. The myelosuppression model showed an additive effect of Ra-223 to PTX-induced neutropenia, with an additional 10% average decrease in absolute neutrophil count vs PTX alone.

Conclusions

In pts with solid tumors and bone mets, Ra-223 was well tolerated when combined with PTX, with an additional 10% average decrease in neutrophil levels compared with PTX monotherapy. The combination should be explored further in pts with bone mets.

Clinical trial identification

NCT02442063

Collapse
Poster display session Poster Display session

417TiP - A phase 1 study of oral LOXO 292 in adult patients with advanced solid tumors, including RET-fusion non-small cell lung cancer, medullary thyroid cancer and other tumors with increased RET activity (ID 3126)

Presentation Number
417TiP
Lecture Time
13:15 - 13:15
Speakers
  • A. Drilon
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

RET is a receptor tyrosine kinase with critical roles in normal physiology. Fusions of the RET kinase with a partner protein have been identified in ∼2% of nonsmall cell lung cancers (NSCLC) and a subset of papillary thyroid cancers and other tumors. RET mutations occur in the majority of medullary thyroid cancers (MTC). Although multikinase inhibitors with anti-RET activity are in the clinic, their activity is limited by incomplete RET inhibition in patients, toxicity from off-target effects (e.g.VEGFR2) and poor pharmacokinetics (PK). LOXO-292 is a potent and specific inhibitor of RET, including fusions, activating mutations and potential acquired resistance mutations, with minimal inhibition of off targets, including > 100-fold selectivity for VEGFR2.

Trial design

This is an open label, multi-center, dose escalation and expansion Phase 1 study in adult patients with advanced solid tumors. Major eligibility criteria for dose escalation include prior cancer treatment (prior treatment with anti-RET TKIs allowed) and normal hematopoietic and major organ function. During dose escalation, when a dose level is achieved that is safe and consistent with RET target engagement, enrollment will be limited to patients with RET-fusion NSCLC, MTC and other tumors with RET alterations or increased RET activity, as identified in tumor or blood by prior molecular assays performed locally. Once the Maximum Tolerated Dose (MTD) or recommended dose for further study is identified, patients will be enrolled to one of five dose expansion cohorts, depending on tumor type (i.e. NSCLC, MTC, other cancer), prior TKI therapy and type of RET alteration. The starting dose of LOXO-292 is 20 mg orally once per day, and dose escalation is proceeding using a 3 + 3 design. The primary endpoint is establishment of the MTD/recommended dose for further study. Key secondary endpoints include: safety and tolerability, PK parameters and preliminary assessment of anti-tumor activity. Patients undergo safety, clinical and PK assessments and radiographic evaluation for their disease at regular intervals.

Clinical trial identification

Treatment of patients has begun. The study has been submitted to the NIH (https://clinicaltrials.gov) and the ClinicalTrials.gov Identifier is pending and will be provided as soon as available.

Legal entity responsible for the study

Loxo Oncology

Funding

Loxo Oncology

Disclosure

S. Smith, T. Eary, S. Cruickshank, M. Nguyen, S. Rothenberg: Ownership interest in Loxo Oncology. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

418TiP - Phase 1/2 study of the selective TRK inhibitor larotrectinib, in pediatric patients with cancer (ID 1385)

Presentation Number
418TiP
Lecture Time
13:15 - 13:15
Speakers
  • S. Gallego Mélcon
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Neurotrophin ligands and their receptors TRKA, TRKB, and TRKC (encoded by NTRK1, NTRK2, and NTRK3) are important for growth regulation, differentiation and survival of neurons. Translocations involving the NTRK1/2/3 kinase domain have been described in a broad range of adult and pediatric tumors, including infantile fibrosarcoma (IFS), spindle-cell sarcoma, congenital mesoblastic nephroma, pediatric papillary thyroid cancer, high- and low-grade gliomas and Ph-like acute lymphoblastic leukemia. Larotrectinib is the first small-molecule selective inhibitor of TRKA, -B, and -C in clinical development and has demonstrated tumor growth inhibition in preclinical models and clinically meaningful and durable responses in patients with NTRK-translocated cancers in an adult phase 1 trial.

Trial design

We have initiated an open-label, multi-center, international Phase 1/2 study with larotrectinib in pediatric patients with solid tumors and primary CNS tumors. A pediatric recommended phase 2 dose of 100mg/m2 (caped at 100mg BID) has been established. Enrollment to phase 2 began in April 2017 and is ongoing. For the phase 2 component, patients from 1-month of age with IFS or an NTRK-fusion positive tumor, including those who have not undergone definitive surgery are eligible. Patients who have not undergone definitive surgery are eligible as well. Larotrectinib is administered as an oral liquid formulation or capsules twice daily on a continuous 28-day schedule. Dosing is based on body surface area. The phase 2 portion enrolls patients with NTRK-translocated tumors and measurable disease into three cohorts: 1) infantile fibrosarcoma; 2) other extracranial solid tumors; and 3) primary CNS tumors. The primary endpoint is objective response rate, with duration of response and progression free survival as secondary efficacy endpoints. Quality of life measures and ctDNA are exploratory endpoints. Each phase 2 cohort will enroll in a single stage of up to 10 patients. Molecular abnormalities will be characterized through the analysis of archival tissue.

Clinical trial identification

NIH: NCT02637687; EudraCT #: 2016-003498-16

Legal entity responsible for the study

Loxo Oncology, Inc.

Funding

Loxo Oncology, Inc

Disclosure

M.C. Cox: Employee and stockholder of Loxo Oncology, Inc. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

419TiP - Chemosensitization of carboplatin by NOX66: Pharmacokinetics and safety (ID 1896)

Presentation Number
419TiP
Lecture Time
13:15 - 13:15
Speakers
  • I. Minns
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The experimental anti-cancer drug, idronoxil, is a selective inhibitor of PI3K/AKT in tumor cells, with studies showing it to be a potent chemosensitizing agent of carboplatin in vitro and in animal studies across a wide range of cancer cell types. These results, however, have not translated to clinical efficacy, with a Phase 3 study of combined oral idronoxil and intravenous carboplatin in patients with late-stage platinum-refractory ovarian cancer discontinued with no efficacy seen. This lack of efficacy is now thought to be due to complete conversion of idronoxil to bio-inactive Phase 2 metabolites. NOX66 is a suppository formulation of idronoxil designed to protect the drug from Phase 2 metabolism. This first-in-human study will look at the ability of NOX66 to deliver relatively high levels of idronoxil in a bio-active form, investigating (a) PK and (b) safety of NOX66 administration both as a monotherapy and in combination with carboplatin.

Trial design

This is an open label, Phase 1 PK and safety study of NOX66 as a monotherapy and in combination with carboplatin. Patients included have end stage, refactory solid tumours, and no further therapy options available. A total of 16 patients will be recruited into the study in two cohorts of 8 patients. NOX66 suppositories are formulated using 400mg of idronoxil per 2.2g suppository. Patients are allocated to receive either one or two suppositories per day. Study Part 1: NOX66 PK: Patients receive NOX66 for 14 consecutive days as monotherapy, with a follow up period of 7 days post-dosing. Blood samples will be collected throughout the monotherapy arm to measure levels of idronoxil. If no significant adverse events are noted in this 21 day period, a patient will continue in the study. Study Part 2: NOX66 plus Carboplatin: Patients receive NOX66 at the same dose as in Part 1, for 7 days. Carboplatin is administered on Day 2 of treatment. Up to 6 cycles of chemotherapy are administered, at intervals of 28 days. For Cycles 1-3, low dose (AUC4) carboplatin is administered. Subject to safety review, standard dose (AUC6) carboplatin is administered for cycles 4-6. Safety assessment is continued throughout the study, with measures to identify efficacy signals (CT scan, ECOG) performed at baseline and after Cycles 3 and 6.

Clinical trial identification

Trial Protocol Number: NOX66-001A Clinicaltrials.gov NCT02941523

Legal entity responsible for the study

Noxopharm Limited

Funding

Noxopharm Limited

Disclosure

I. Minns: Employee of Noxopharm Limited. G. Kelly: Member of the board of Directors, employee and a shareholder of Noxopharm Limited.

Collapse
Poster display session Poster Display session

420TiP - Phase 1b/2 study to assess the safety, tolerability, and clinical activity of BGB-290 in combination with temozolomide in patients with locally advanced or metastatic solid tumors (ID 2089)

Presentation Number
420TiP
Lecture Time
13:15 - 13:15
Speakers
  • M. Johnson
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Poly (ADP-ribose) polymerase (PARP) proteins are a family of DNA binding and repair proteins and are thought to play a key role in the base excision repair of DNA damage generated by temozolomide (TMZ), a DNA-alkylating agent. PARP inhibitors (PARPis) represent a class of antitumor agents that exert their cytotoxic effects by inhibiting PARP activity. Some PARPis are capable of trapping PARP proteins on DNA, further augmenting cell death. BGB-290 is a potent and selective inhibitor of PARP1/2 and has demonstrated PARP trapping capacity. Synergistic cytotoxicity has been observed in vitro and in vivo when BGB-290 is combined with low dose TMZ.

Background

Poly (ADP-ribose) polymerase (PARP) proteins are a family of DNA binding and repair proteins and are thought to play a key role in the base excision repair of DNA damage generated by temozolomide (TMZ), a DNA-alkylating agent. PARP inhibitors (PARPis) represent a class of antitumor agents that exert their cytotoxic effects by inhibiting PARP activity. Some PARPis are capable of trapping PARP proteins on DNA, further augmenting cell death. BGB-290 is a potent and selective inhibitor of PARP1/2 and has demonstrated PARP trapping capacity. Synergistic cytotoxicity has been observed in vitro and in vivo when BGB-290 is combined with low dose TMZ.

Trial design

This open-label, Phase 1b/2 dose-escalation/dose-expansion study is designed to evaluate BGB-290 at the recommended Phase 2 dose (60 mg administered orally twice daily [PO BID]) in combination with TMZ in patients with locally advanced and metastatic solid tumors. The phase 1b dose-escalation component will follow a 3 + 3 design to establish the maximum tolerated dose (MTD) of TMZ in combination with BGB-290 in ∼50 patients with solid tumors. Dose escalation will evaluate the safety, tolerability and pharmacokinetics of BGB-290 (60 mg BID) plus escalating doses of TMZ administered once daily (QD) either on Days 1-7 (Arm A) or continuously (Arm B) of each 28-day cycle. The phase 2 component will further evaluate the safety, tolerability and antitumor activity of the recommended combination dose and schedule in ∼20 patients with one of five different tumor types (Table). Enrollment into these expansion cohorts will occur simultaneously and independent of each other. Subjects will continue to receive treatment in 28-day cycles until confirmed disease progression, intolerable toxicity, or discontinuation/withdrawal.

420TiP

Treatment armTumor typeEstimated sample size
Cohort 1Platinum-sensitive high grade epithelial, non-mucinous, ovarian cancer, fallopian cancer or primary peritoneal cancer with either known deleterious or suspected deleterious germline or somatic BRCA1/2 mutation or with DNA HRD20
Cohort 2Triple negative breast cancer with either known deleterious or suspected deleterious germline or somatic BRCA1/2 mutation or with DNA HRD20
Cohort 3Metastatic castration-resistant prostate cancer with either known deleterious or suspected deleterious germline or somatic BRCA1/2 mutation or with documented HRD20
Cohort 4Extended stage small cell lung cancer who have been treated with ≤2 prior regimens20
Cohort 5Gastric or gastroesophageal junction cancer who have been treated with ≤ 2 prior regimens20

Legal entity responsible for the study

Beigene Ltd.

Funding

Beigene Ltd

Disclosure

M. Johnson: Research funding compensation for consulting to the institution from OncoMed, BerGenBio, Lilly, and various Pharm/Biotech companies. Spouse is a contract lobbyist for Astellas and Otsuka Pharmaceuticals. B. Benson, R. Wei: Employee and stock holder of BeiGene USA. R. Brachmann: Employee of BeiGene USA. M.D. Galsky: Consulting fees from Genentech, Merck, Novartis, Astellas, Astra Zeneca, and Bristol-Myers Squibb outside the submitted work; stock options in Dual Therapeutics outside the submitted work.

Collapse
Poster display session Poster Display session

421TiP - Phase 1b/2 study to assess the clinical effects of BGB-290 in combination with radiation therapy (RT) and/or temozolomide (TMZ) in patients with first-line or recurrent/refractory glioblastoma (ID 2090)

Presentation Number
421TiP
Lecture Time
13:15 - 13:15
Speakers
  • P. Wen
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Poly (ADP-ribose) polymerase (PARP) proteins are a family of DNA binding and repair proteins and are thought to play a key role in the base excision repair of DNA damage generated by TMZ. In glioblastoma (GB) cells, pharmacological modulation of PARP activity increased growth inhibition induced by TMZ in both p53-wild type and-mutant GB cells lowering the TMZ IC50. RT used in the clinical treatment of GB generates mostly single-strand breaks (SSBs). In non-replicating cells PARP inhibition only delays the repair of SSBs induced by radiation with a minimal impact on cell survival. On the contrary, PARP inhibition markedly enhances radiosensitivity of proliferating cells generating double-strand breaks. Thus, PARP inhibitors have the potential to increase the therapeutic index of RT by increasing DNA damage mainly in highly replicating tumor cells, but sparing non-cycling normal tissues. BGB-290, a potent and selective inhibitor of PARP1/2, has demonstrated potent PARP trapping, brain penetrance and antitumor activity in preclinical intracranial xenograft models.

Background

Poly (ADP-ribose) polymerase (PARP) proteins are a family of DNA binding and repair proteins and are thought to play a key role in the base excision repair of DNA damage generated by TMZ. In glioblastoma (GB) cells, pharmacological modulation of PARP activity increased growth inhibition induced by TMZ in both p53-wild type and-mutant GB cells lowering the TMZ IC50. RT used in the clinical treatment of GB generates mostly single-strand breaks (SSBs). In non-replicating cells PARP inhibition only delays the repair of SSBs induced by radiation with a minimal impact on cell survival. On the contrary, PARP inhibition markedly enhances radiosensitivity of proliferating cells generating double-strand breaks. Thus, PARP inhibitors have the potential to increase the therapeutic index of RT by increasing DNA damage mainly in highly replicating tumor cells, but sparing non-cycling normal tissues. BGB-290, a potent and selective inhibitor of PARP1/2, has demonstrated potent PARP trapping, brain penetrance and antitumor activity in preclinical intracranial xenograft models.

Trial design

This open-label, dose-escalation/dose-expansion Phase 1b/2 study was designed to determine the safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor effects of BGB-290 at the recommended Phase 2 dose (60 mg PO BID) in combination with RT and/or TMZ. The Phase 1b component will consist of 3 dose-escalation arms. Arm A: BGB-290 will be combined with RT in patients with first-line GB with unmethylated MGMT promoter (‘unmethylated GB’); Arm B: BGB-290 will be combined with both TMZ and RT in patients with first-line unmethylated GB; Arm C: BGB-290 will be combined with increasing doses of TMZ in patients with recurrent/refractory methylated or unmethylated GB. Once a recommended Phase 2 regimen has been established, up to 60 patients may be enrolled in the dose-expansion (Phase 2) cohort for that arm. In Arm C, 2 expansion cohorts with up to 60 patients each may be opened: 1 for unmethylated GB and 1 for methylated GB.

Legal entity responsible for the study

Beigene Ltd.

Funding

Beigene Ltd.

Disclosure

P. Wen: Grants, personal fees, and/or non-financial support from Agios, Angiochem, AstraZeneca, Genentech/Roche, GlaxoSmithKline, Immunocellular Therapeutics, Karyopharm, Merck, Novartis, and other biotech/pharma companies outside submitted work. D. Schiff: Grants from Cavion & Celldex, personal fees from VBI, Orbus, Monteris, Genentech-Roche, Heron Pharmaceuticals, Midatech, and Oxigene, outside the submitted work. R. Brachmann: Employee of Beigene USA, Inc. R. Weitzman: Consultant to BeiGene. T. Cloughesy: Personal fees from Pfizer, Tocagen, Roche, Novocure, Nektar, VBL, ABBVIE, Upshire Smith, Notable Labs, Oxigene, NewGen, Agios, Cortice, MedQia, PRoNai,and other pharma/biotech companies, outside the submitted work. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

422TiP - PROCLAIM-CX-2009: A first-in-human trial to evaluate CX-2009 in adults with metastatic or locally advanced unresectable solid tumors (ID 2496)

Presentation Number
422TiP
Lecture Time
13:15 - 13:15
Speakers
  • J. Garcia-Corbacho
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

CX-2009 is a novel recombinant Probody™ drug conjugate (PDC) derived from a humanized monoclonal antibody (mAb) against CD166 and conjugated to N-succinimidyl 4-(2-pyridyldithio) butanoate-N2′-deacetyl-N2′-(4-mercapto-4-methyl-1-oxopentyl)-maytansine (SPDB-DM4, licensed from Immunogen), a potent microtubule inhibitor. PDCs are fully recombinant mAb prodrugs designed to remain inactive until they are cleaved into an active mAb by tumor-associated proteases. This tumor-specific activation allows PDCs to target highly and homogeneously expressed tumor antigens while avoiding binding to these same targets on healthy tissue. An example is CD166 (also referred to as activated leukocyte cell adhesion molecule [ALCAM]), which is highly expressed in multiple cancers but also in healthy tissue. In preclinical studies, CX-2009 exhibited antitumor activity and reduced peripheral binding compared to the corresponding anti-CD166 ADC.

Trial design

PROCLAIM-CX-2009 (PRObody CLinical Assessment In Man) is an open-label, multicenter, dose-escalation study to determine the maximum tolerated dose (MTD) and the recommended phase 2 dose (RP2D) of CX-2009 in 7 selected tumor types with high CD166 expression (breast, lung, prostate, ovarian, endometrial, head and neck, and biliary carcinomas). Part A (n ≤ 50) will initiate with accelerated dose titration, followed by a standard 3 + 3 design to determine the MTD and ending in a modified toxicity probability interval 2-design cohort treated at the MTD to determine the RP2D. Part B of the study will be a dose expansion phase testing CX-2009 administered at the RP2D in the same 7 tumor types (up to 14 patients each, n ≤ 98). Eligibility is based on confirmed refractory metastatic or locally advanced unresectable tumor. Outcome measures include assessment of safety, tolerability, pharmacokinetics, and efficacy based on RECIST 1.1. Exploratory biomarkers will characterize tumor CD166 expression and mitotic markers as well as CX-2009 activation in tumor versus peripheral blood.

Clinical trial identification

NCT03149549

Legal entity responsible for the study

CytomX Therapeutics, South San Francisco, CA, USA

Funding

CytomX Therapeutics, South San Francisco, CA, USA

Disclosure

M. Middleton: Grants: Roche, AstraZeneca, GSK. Advisory board: Amgen, Novartis, Rigontec, CytomX. Personal fees: Amgen, Roche, GSK, Novartis, Bristol-Myers Squibb, Eisai, Merck, CytomX. A. Yang Weaver, M. Will: Employee of CytomX Therapeutics. J. Harding: Consultant to Bristol-Myers Squibb. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

423TiP - The first-in-human, dose-finding PROCLAIM-CX-072 trial to assess the antitumor activity and tolerability of the probody therapeutic CX-072 as monotherapy and in combination with ipilimumab or vemurafenib in solid advanced tumors and lymphomas (ID 5399)

Presentation Number
423TiP
Lecture Time
13:15 - 13:15
Speakers
  • V. Boni
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

CX-072 is a novel protease activatable prodrug (Probody™ therapeutic) derived from a human monoclonal antibody against programmed cell death ligand 1 (PD-L1). CX-072 was designed to restrict its activity to the tumor microenvironment and remain largely inactive in nonmalignant tissue. A surrogate molecule for CX-072 displayed potent antitumor activity with reduced systemic immune activation and immune-related toxicities in preclinical tumor models, potentially enabling new and more effective combination therapies. The phase 1/2 PROCLAIM-CX-072 (PRObody CLinical Assessment In Man) study will assess the safety, tolerability, and antitumor activity of CX-072, as monotherapy and in combination, in adults with advanced or recurrent solid tumors and lymphomas.

Trial design

In an open-label, multicenter, dose-escalation, 3 + 3 design, CX-072 will be administered as monotherapy (Part A) in 2 combination schedules with ipilimumab 3 mg/kg 3 weekly × 4 (Parts B1 and B2) and in combination with vemurafenib 960 mg/kg twice daily (Part C). The expansion cohort (Part D) will include CX-072 monotherapy in PD-L1–responsive tumor types. Patient recruitment was initiated on January 11, 2017. Key inclusion criteria are: Parts A and B1—advanced, refractory solid tumor or lymphoma in checkpoint inhibitor-naive patients for whom approved PD agents are not available; Part B2—advanced, refractory solid tumors or lymphomas with measurable disease that progressed on previous treatment with a PD-1/PD-L1 inhibitor but patients did not discontinue due to toxicity; Part C—checkpoint inhibitor, BRAF-inhibitor, and MEK-inhibitor-naive metastatic V600E BRAF-mutated melanoma. Efficacy will be determined according to irRECIST v1.1 criteria, and safety and tolerability will be assessed based on the incidence and nature of dose-limiting toxicities, adverse events (AEs), and serious AEs. Exploratory biomarkers will be used to characterize tumor protease activity, immune response pattern within the tumor, and CX-072 activation in tumor vs peripheral blood.

Clinical trial identification

NCT03013491

Legal entity responsible for the study

CytomX Therapeutics, South San Francisco, CA, USA

Funding

CytomX Therapeutics, South San Francisco, CA, USA

Disclosure

J. Wydmanski: Consultant to Bristol-Myers Squibb. B. Irving, M. Will: Employee of CytomX Therapeutics. F. Thistlethwaite: Personal fees from Novartis, Bristol-Myers Squibb, Pfizer, and Ipsen. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

424TiP - Phase 1b multi-indication study of the antibody drug conjugate anetumab ravtansine in patients with mesothelin-expressing advanced or recurrent malignancies (ID 3308)

Presentation Number
424TiP
Lecture Time
13:15 - 13:15
Speakers
  • A. Adjei
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Mesothelin is expressed in a wide variety of tumours, including mesothelioma, ovarian, pancreatic, gastric/GEJ, NSCLC, triple-negative breast cancer, cholangiocarcinoma, and thymic carcinomas. Anetumab ravtansine (BAY 94-9343), is a novel fully human anti-mesothelin IgG1 antibody conjugated to the maytansinoid tubulin inhibitor DM4 and has shown encouraging anti-tumor activity in mesothelioma and ovarian cancer patients in a phase I study. We will therefore conduct a signal generating study with anetumab ravtansine in six additional high unmet medical need malignancies with mesothelin expression (NCT03102320).

Trial design

Eligibility criteria include: ≥18 years, unresectable locally advanced or metastatic recurrent or relapsing disease, one or more prior lines of therapy, and availability of tumour tissue for mesothelin expression testing. Mesothelin-positive patients with selected adenocarcinomas (NSCLC, triple negative breast, gastric including gastroesophageal junction) and thymic carcinoma will receive anetumab ravtansine as monotherapy at 6.5 mg/kg IV on a 21-day cycle. Patients with cholangiocarcinoma will receive anetumab ravtansine in combination with cisplatin (25 mg/m2 IV day 1 and 8 on a 21-day cycle for up to 6 cycles) and patients with pancreatic adenocarcinoma will receive anetumab ravtansine in combination with gemcitabine (1000 mg/m2 IV day 1 and 8 on a 21-day cycle). A safety run-in phase (18-24 patients each) will be conducted for the combination regimens prior to enrolling patients in the main study phase. The primary objective of the main phase of the study is objective response rate (ORR) of anetumab ravtansine as monotherapy or combination therapy in patients with either of two mesothelin expression levels: high (≥30% positive tumor cells with moderate and stronger membrane staining intensity) and low-mid (≥5% all intensities and <30% positive tumour cells with moderate and stronger membrane staining intensity). Secondary objectives include safety, disease control rate, duration of response, durable response rate, and progression-free survival. Approximately 348 patients will be enrolled.

Clinical trial identification

NCT03102320

Legal entity responsible for the study

Bayer AG

Funding

Bayer AG

Disclosure

A. Walter: Employee of Bayer AG. L. Cupit, J. Siegel, A. Holynskyj, B.H. Childs, C. Elbi: Employee of Bayer HealthCare Pharmaceuticals Inc. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

425TiP - A phase 1 study of SY-1365, a selective CDK7 inhibitor, in adult patients with advanced solid tumors (ID 4152)

Presentation Number
425TiP
Lecture Time
13:15 - 13:15
Speakers
  • A. Tolcher
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

SY-1365 is a selective and potent covalent CDK7 inhibitor. CDK7 activity has been implicated in malignancies with transcriptional dependencies such as SCLC, TNBC, ovarian cancer, MYCN-amplified neuroblastoma, and various hematologic malignancies including AML and T-ALL. Preclinical studies in solid tumor and hematologic malignancies show treatment with SY-1365 leads to antitumor activity, showing apoptosis in vitro and complete regressions in xenograft models. This first clinical study is focused on patients with advanced solid tumors. The primary objectives are to assess the safety and tolerability of SY-1365 administered intravenously as a single agent, and to determine dose-limiting toxicities, maximum tolerated dose, and the recommended phase 2 dose. Secondary objectives include evaluation of pharmacokinetic (PK) properties and pharmacodynamic (PD) effects of SY-1365 in tumor and surrogate tissues, as well as assessment of preliminary anti-tumor activity.

Trial design

This is a multi-center, open-label Phase 1 trial that is expected to enroll approximately 70 patients with advanced solid tumors. The dose escalation phase of the trial is open to solid tumor patients for whom standard curative or palliative measures do not exist or are no longer effective. Initially, SY-1365 will be administered intravenously twice weekly for 3 weeks of each 4-week cycle. Regimen optimization will be based upon PK, PD, and safety data prior to an expansion phase to evaluate preliminary antitumor activity of SY-1365 in 25 patients with SCLC, TNBC or ovarian cancer. A second expansion cohort will enroll 10 patients with tumors of any histology to evaluate PD endpoints in paired tumor biopsies. SY-1365 target engagement in peripheral blood mononuclear cells and available tumor biopsies will be assessed by measuring CDK7 occupancy over the course of treatment. Downstream biological pathway impact of SY-1365 will be measured by quantifying changes in gene expression as a result of transcriptional inhibition. Induction of tumor cell apoptosis will also be investigated. This trial opened in May 2017. ClinicalTrials.gov identifier: NCT03134638.

Clinical trial identification

Study Protocol Number: SY-1365-101. ClinicalTrials.gov NCT03134638

Legal entity responsible for the study

Syros Pharmaceuticals, Inc

Funding

Syros Pharmaceuticals, Inc

Disclosure

A. Tolcher: Co-owner of South Texas Acelerated Research Therapeutics which receives fees for consulting and board memberships from companies (17); and research funding from companies (34) for his role as principle investigator. E. di Tomaso: Employee and stock owner of Syros Pharamceuticals. N. Waters, D.A. Roth, K. Stephens: An employee and stock holder of Syros Pharmaceuticals. G. Shapiro: Advisory boards for Pfizer, Lilly, G1 Therapeutics, Roche and Vertex Pharmaceuticals. Research funding from Pfizer and Lilly for CDK inhibitor-based projects. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

426TiP - First-in-human study of AMC303 as monotherapy in patients with advanced solid tumor of epithelial origin (ID 4454)

Presentation Number
426TiP
Lecture Time
13:15 - 13:15
Speakers
  • E. Calvo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

CD44v6 is an isoform of the CD44 family of transmembrane glycoproteins for hyaluronan. High CD44v6 expression was correlated with tumor invasion, metastasis, recurrence and chemoresistance. CD44v6 is a co-receptor of the receptor tyrosine kinases c-Met, RON and VEGFR-2 that play a critical role in the development and progression of many types of cancer. Inhibition of CD44v6 efficiently blocks activation of c-Met, RON and VEGFR-2 and intracellular downstream signaling processes. AMC303 is a highly specific and selective inhibitor of CD44v6 for which strong anti-tumor activity was demonstrated in vitro and in vivo. In xenotransplantation animal models, intermittent application of AMC303 resulted in a marked reduction of the primary tumor, prevention of metastatic spread and regression of existing metastases. Good safety and tolerability were demonstrated in pre-clinical studies. The starting dose of 0.1 mg/kg and dose escalation steps are based on the safety profile and are supported by modelling of human pharmacokinetic (PK) profiles from animal exposure studies. Blocking of CD44v6 by AMC303 represents a novel and promising approach to block cancer related RTK pathways by an extracellular acting drug.

Trial design

A First-in-Human Phase I/Ib study in cancer patients was initiated (NCT03009214). The study was designed as a two part open-label, non-randomized, multicentre, dose escalation study with a 3 + 3 design (Part 1) and an expansion cohort at the Maximum Tolerated Dose (MTD)/Recommended Phase 2 Dose (RP2D) (Part 2). Inclusion and exclusion criteria are: Type of cancer (e.g. epithelial cancer for which CD44v6 is known to be highly expressed), ECOG status 0-2, and adequate hematological, renal and hepatic function. Cancer patients are enrolled after failure of conventional therapy or for whom no standard treatment is available. The primary endpoints in Part 1 are safety and tolerability and PK properties. The effects of AMC303 on RTK pathways are analysed in plasma samples (ELISA and Luminex) and mandatory tumor biopsies (immunohistochemistry, protein profiling). Part 2 will focus on selected tumor types as evaluated from the pharmacological effects of AMC303 in part 1.

Clinical trial identification

EudraCT number: 2016-001358-16

Legal entity responsible for the study

Amcure GmbH

Funding

None

Disclosure

H. Bender, K. Dembowsky: Employee by amcure GmbH and stock holder. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

NEW DIAGNOSTIC TOOLS (ID 5677)

Lecture Time
13:15 - 13:15
Speakers
  • J. Seoane
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15
Poster display session Poster Display session

1261P - mRNA capture sequencing enabled liquid biopsy screening (ID 5601)

Presentation Number
1261P
Lecture Time
13:15 - 13:15
Speakers
  • J. Vandesompele
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In contrast to general belief, a substantial part of the human protein coding transcriptome is abundantly present in the blood as extracellular mRNA, ready to exploited. It is well known that cancer cells actively and passively release RNA cargo into circulation and their detection may inform on the patient disease status.

Methods

We developed and applied a probe based RNA capture sequencing method as a sensitive RNA sequencing workflow to study thousands of transcripts in cell-free RNA from cancer patients’ plasma. The method is based on exome-style enrichment of a randomly primed cDNA library with preservation of strandedness information. More than one million capture probes target 21,000 human messenger RNA and 60,000 human long non-coding RNA genes. Apart from RNA abundance profiling, this type of data can also be used to detect structural RNA variants, such as somatic mutations, fusion genes, and RNA editing events, all known to play an important role in cancer.

Results

On average, between 6000-10,000 RNA genes are reproducibly detected in 0.2 ml of plasma. Detection and coverage sensitivity is greatly increased by using larger plasma volume and improved adaptor ligation strategies. We also observed a positive correlation between number of platelets in plasma and detected genes and variants, in line with their tumor-educated nature. Our benchmarked RNA variant pipeline identifies thousands of germline and somatic variants in circulating mRNA. A dedicated titration experiment in which plasma from cancer and healthy individuals were mixed in known ratios demonstrates excellent quantitative performance. Pronounced RNA abundance differences and enriched pathways are observed between cancer types and during treatment. The RNA capture sequencing also works on other body fluids, such as urine and serum, and simultaneous targeting of mRNA and lncRNA provides substantial enrichment of otherwise low-abundant lncRNAs.

Conclusions

RNA capture sequencing of liquid biopsies is a promising new application to support precision oncology and is expected to enhance therapy stratification, treatment response monitoring and early detection of relapse.

Legal entity responsible for the study

Biogazelle and Ghent University

Funding

Biogazelle, Illumina

Disclosure

J. Vandesompele: Apart from professorship at Ghent University, co-founder and part-time CSO at Biogazelle, a Ghent University spin-off company.

Collapse
Poster display session Poster Display session

1262P - Use of droplet digital PCR for quantitative and automatic analysis of the HER2 status in breast cancer patients (ID 1762)

Presentation Number
1262P
Lecture Time
13:15 - 13:15
Speakers
  • K. Otsuji
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Digital polymerase chain reaction (dPCR) has been used to yield an absolute measure of nucleic acid concentrations. Recently, a new method referred to as droplet digital PCR (ddPCR) has gained attention as a more precise and less subjective assay to quantify DNA amplification. We demonstrated the usefulness of ddPCR to determine HER2 gene amplification of breast cancer.

Methods

In this study, we used ddPCR to measure the HER2 gene copy number in clinical formalin-fixed paraffin-embedded samples of 41 primary breast cancer patients. To improve the accuracy of ddPCR analysis, we also estimated the tumour content ratio (TCR), the ratio of tumour cell count per section, for each sample.

Results

Our determination method for HER2 gene amplification using the ddPCR ratio (ERBB2:ch17cent copy number ratio) combined with the TCR showed high consistency with the conventionally defined HER2 gene status according to ASCO-CAP (American Society of Clinical Oncology/College of American Pathologists) guidelines (P < 0.0001, Fisher’s exact test). The equivocal area was established by adopting 99% confidence intervals obtained by cell line assays, which made it possible to identify all conventionally HER2-positive cases with our method. In addition, we succeeded in automating a major part of the process from DNA extraction to determination of HER2 gene status.

Conclusions

The introduction of ddPCR to determine the HER2 gene status in breast cancer is feasible for use in clinical practice and might complement or even replace conventional methods of examination in the future.

Legal entity responsible for the study

The University of Tokyo

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1263P - BRCA1 large gene rearrangements (LGRs) in Russian breast cancer patients: the development of the droplet digital PCR assay for LGR detection and the identification of recurrent exon 8 deletions (ID 2698)

Presentation Number
1263P
Lecture Time
13:15 - 13:15
Speakers
  • E. Imyanitov
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Some pathogenic BRCA1 mutations are represented by large gene rearrangements (LGRs). LGRs cannot be detected by conventional Sanger sequencing. Multiplex ligation-dependent probe amplification (MLPA) is a leading method for LGR detection, however it is entirely based on the use of commercial kits, includes relatively lengthy hybridization step and is poorly suitable for the large-scale screening for recurrent deletions.

Methods

We developed and validated the droplet digital PCR (ddPCR) assay, which covers the entire coding region of BRCA1 gene and is capable to precisely quantitate the copy number for each exon.

Results

141 breast cancer (BC) patients, who demonstrated evident clinical features of hereditary BC but turned out to be BRCA1/2 mutation-negative upon Sanger sequencing, were subjected to the LGR analysis. 4 patients with LGR were identified, with 3 cases of exon 8 deletion and 1 women carrying the deletion of exons 3-7. Excellent concordance with MLPA was observed. Exon 8 copy number was tested in additional 720 high-risk BC, and another 3 cases with the deletion were revealed; MLPA re-analysis demonstrated that exon 8 loss was a part of a larger genetic alteration in 2 cases, while the remaining patient had isolated defect of exon 8. Long-range PCR and next generation sequencing revealed, that 3 out of 4 samples with isolated exon 8 deletion had an identical rearrangement.

Conclusions

Droplet digital PCR is a reliable tool for detection of large gene rearrangements. BRCA1 LGRs are rare in Russian hereditary BC patients, with exon 8 deletion being a recurrent allele in this population.

Legal entity responsible for the study

Laboratory of Molecular Oncology, N.N. Petrov Institute of Oncology, St.-Petersburg

Funding

Russian Science Foundation (grant 14-25-00111)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1264P - Clinical evaluation of low density array based EGFR mutation detecting kit using tissue samples and liquid biopsies (ID 2973)

Presentation Number
1264P
Lecture Time
13:15 - 13:15
Speakers
  • J. Hernández-Losa
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The information on EGFR mutations status improves the benefit of targeted therapies for the non-small-cell lung cancer patients. These determinations are already part of standard protocols for cancer treatment. The study of the mutations in EGFR in liquid biopsy also permits a follow-up throughout the treatment and detection of acquired resistance.

Methods

CLART® CMA EGFR kit (GENOMICA, Spain) detects the 40 high-prevalence mutations associated with sensitivity or resistance to the treatment. These mutations are located in the exons 18, 19, 20 and 21. The kit is based on the multiplex ARMS-PCR followed by detection on a low-density microarray platform: CLART® (Clinical Arrays Technology). The samples were processed in a bench-top semi-automated system, Autoclart. A new version of the kit, CLART® CMA EGFR LB, allows using liquid biopsy as a sample with only an additional pre-PCR step.

Results

A 107 tissue biopsies from metastatic NSCLC patients were obtained and analyzed in two University Hospitals in Spain: Vall d’Hebron (Barcelona) and 12 de Octubre (Madrid). The EGFR mutations were detected with CLART® CMA EGFR kit with 96.3% concordance with the routine methods used in the hospital practice (Cobas/Therascreen/Sanger sequencing). The discrepant results were analyzed by Sanger sequencing. The sensitivity of the CLART® CMA EGFR kit is 100% and specificity is 96.5%. The same CLART platform, only with the addition of one pre-PCR amplification step was used for processing the plasma samples (liquid biopsy) from the NSCLC metastatic patients. A total of 8 samples were tested. In six samples the results obtained from tissue samples and liquid biopsies were concordant. In the two discordant samples the exon 19 deletions detected in tissue were not detected in plasma samples. The wt results obtained for these two liquid biopsies were confirmed by Next Generation Sequencing.

Conclusions

Given the high specificity and sensitivity and excellent concordance with the other platforms in hospital practice, CLART® CMA EGFR kit is valid for the use in the clinical routine. Furthermore, using the same semi-automated platform for tissue samples and liquid biopsy facilitates laboratory work and reduces turnover time per sample.

Clinical trial identification

N/A

Legal entity responsible for the study

GENOMICA

Funding

GENOMICA

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1265P - Correlation of somatic genomic alterations between tissue genomics and circulating tumor DNA (ctDNA) employing next generation sequencing (NGS) analysis in lung and gastrointestinal cancers (ID 3989)

Presentation Number
1265P
Lecture Time
13:15 - 13:15
Speakers
  • M. Toor
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Peripheral blood circulating tumor DNA (ctDNA) and tumor tissue next-generation sequencing (NGS) is routinely performed to guide therapy in cancer patients. However, little is known about the concordance or discordance between commercially available tissue genomics testing panels and ctDNA. The aim of our study was to assess concordance between matched cancer tissue genomics and blood based ctDNA in lung and gastrointestinal (GI) cancers.

Methods

Tissue genomic analysis was performed with Paradigm® (n = 17)/Caris® (n = 11) and ctDNA was analyzed with Guardant360® (n = 28). Samples included, non-small cell lung cancer (n = 10), small cell lung cancer (n = 4), colorectal cancer (n = 5), hepatocellular carcinoma (n = 2), intrahepatic cholangiocarcinoma (n = 1), pancreatobiliary adenocarcinoma (n = 3), esophageal adenocarcinoma (n = 2) and gastric adenocarcinoma (n = 1).

Results

We identified 6 (21%) patients with at least one gene mutation that was detected by both tissue genomic and ctDNA analysis. Total number of gene mutations identified in 28 patients were 106, but only 8 (7.5%) were detected by both tissue and ctDNA panels. When this testing was done within 90 days the concordance increased to 10.20%. Table.

1265P

Patients with at least one mutation detected by both platforms21% (n = 6/28)
Mutations detectable by both platforms7.54% (n = 8/106)
Mutations detectable by both platforms if interval between tissue and blood collection <90 days.10.20% (n = 5/49)

Conclusions

We identified significant discordance between tissue and ctDNA mutational profiles in lung and GI cancers. Therefore, the results from NGS platforms should be interpreted with extreme caution. Our analysis reveals that these platforms should not be used interchangeably. The discordance rate may be due to tissue heterogeneity and/or spatial and temporal clonal evolution. Standardization of the sequencing techniques and education of practicing oncologists about the limitations of liquid biopsies needs to be highlighted.

Legal entity responsible for the study

Saint Luke’s Health System

Funding

None

Disclosure

J. Subramanian: Paradigm Advisory board. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1266P - Analytical and clinical validation of a next-generation sequencing-based circulating tumor DNA (ctDNA) assay assures its clinical application (ID 5021)

Presentation Number
1266P
Lecture Time
13:15 - 13:15
Speakers
  • W. Liu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

To date, ctDNA has been exhibiting its unprecedented translational potential in cancer care. However, accurate identification of comprehensive genomic alternations is rigorously needed for its clinical utility.

Methods

Three biologically relevant reference materials with allele frequencies expected at 0.1%, 1% and 5% were used to analytically evaluate the accuracy and reproducibility. Clinically, 43 ctDNA samples from lung, liver, colorectal, breast and gastric cancers were genomically profiled and compared to the known alternations in their matched solid tumors, in terms of single base substitution, insertions/deletions, copy number variations and rearrangement.

Results

The analytical validation demonstrated unprecedented accuracy: near 100% specificity (99.6%, 99.9% and 100%) and 95.8%, 100% and 100% sensitivity for 3 reference materials, respectively. The actual detection limit was as low as 0.05%. The reproducibility was assessed as 0.998 (jaccard index) by sequencing 2 replicates of each reference. In clinical validation, compared to matched FFPE results, this ctDNA assay showed overall 99.9% specificity and 89% sensitivity, with > 90% sensitivity when only drug-gable hotspots were concerned. Eight events of gene rearrangements involving known targeted genes of ALK (n = 4), ROS1 (n = 3) and MET (n = 1) were detected from seven patients with 100% sensitivity and 100% specificity, confirmed either by IHC or panel sequencing (depth > 1,000X) over their matched FFPE biopsies. Due to its typically low abundance, CNV from ctDNA was detectable only for those highly amplified genes ( > =8 copies) with > =4 exons, demonstrating 72.2% sensitivity and 99.5% specificity.

Conclusions

Stringent criteria for both analytical and clinical validations are required for clinical utility of ctDNA. Our ctDNA assay has demonstrated high accuracy and reliability in comprehensively genomic profiling of ctDNA, especially in regard to druggable targets, which assures its translational utility in optimizing and monitoring targeted therapies in cancer management.

Legal entity responsible for the study

OrigiMed Inc

Funding

OrigiMed Inc

Disclosure

W. Liu, S. Mu, J. Yao, H. Chen, Z. Hu, J. Hu, G. Chirn, H. Kang, K. Wang, M. Yao: Employee of OrigiMed Inc.

Collapse
Poster display session Poster Display session

1267P - Combination of solid and liquid biopsy genomic profiling for tumor heterogeneity characterization (ID 5232)

Presentation Number
1267P
Lecture Time
13:15 - 13:15
Speakers
  • J. Garcia Foncillas
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Characterization of inter- and intra-tumoral heterogeneity has become a central issue in the implantation of personalized medicine. In this sense, although, the liquid biopsy has been recognized as a promising tool for prognostic, molecular profiling and monitoring of cancer disease, we are still at the beginning of its incorporation alone into the routine oncology practice. In the present study, we evaluate the usefulness of an integrated approach that combines the analysis of both solid (FFPE block) and liquid (blood sample) biopsy, into the clinical routine.

Methods

We analyzed 163 samples of metastatic patients, with different cancers types, using the OncoSTRAT&GO® solution (Biosequence SL, Valencia, Spain through OncoDNA SA, Gosselies, Belgium); that allows i) sequencing of more than 200 genes, identification of 350 genes fusion and evaluation of the expression level of tens of proteins in solid biopsy and ii) sequencing of hotspot mutations of a 27-gene panel in liquid biopsy.

Results

We focus the analysis on those actionable variants that could be detected in both solid and liquid biopsies. A complete concordance of 62.6% was observed between both types of biopsies variants. The minimum variant allele frequencies (VAFs) was found to be 0.1% and 1% in liquid and solid biopsy, respectively. The concordant and discordant VAFs were compare showing similar distributions, no significant statistical differences were found: mean values of 14.5/8.9% (P = 0.79; Mann-Whitney test) and 39.4/29.9% (P= .08; Mann-Whitney test) in liquid and solid biopsy, respectively.

Conclusions

Our findings indicate that the combination of solid and liquid biopsies analysis in clinical practice provides additional information in 37.4% of the cases. Discordant variants cannot be put down to the sensibility of the analysis and consequently should be associated to tumor heterogeneity, low tumor burden and/or treatment response. Our results show the usefulness of an integrated approach, resulting in a broad characterization of the tumor for a better disease management.

Legal entity responsible for the study

Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1268P - Retrospective analysis of a SHIVA01 trial cohort using functional mutational analysis successfully predicted treatment outcome (ID 3035)

Presentation Number
1268P
Lecture Time
13:15 - 13:15
Speakers
  • G. Tarcic
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The SHIVA01 trial was a randomized proof-of-concept phase II trial, testing the hypothesis of treating patients (pts) based on molecular profiles with molecularly targeted agents (MTA) in a histology agnostic manner. The trial results showed no statistically significant difference in PFS between MTA and control arm. The addition of functional information on identified mutations and their response to MTA’s may improve treatment outcomes.

Methods

The molecular profile of 20 pts treated with a MTA in the trial was analyzed in the NovellusDx Functional Annotation for Cancer Treatment (FACT), a functional mutational analysis platform, to reveal activated signaling pathways and measure the activity of these mutations in the presence of the MTA’s administered in the trial. The results of FACT were used to stratify the pts into responders and non-responders.

Results

We uncovered the functional landscape in 12 of the 20 pts in the analyzed group. In the remaining 8 pts, no relevant mutational alterations were identified. This analysis provided experimental evidence to the oncogenic activity of the mutations and of the combination of mutations identified in the pts. Furthermore, the response of these pts' mutations to the MTA’s used was measured in-vitro, blinded to the actual clinical results. Each patient was then assigned as either a responsive or non-responsive. When the results were used to stratify the pts’ PFS data, positive patients had a median PFS of 5.8 months vs. 1.7 months in the negative group (P = 0.03 in a non-parametric test).

Conclusions

This analysis shows the predictive power of a new and innovative method for characterization of pts molecular profiles and their in-vitro response to MTA’s. The abundance of mutations classified as VUS and multiple mutations in different genes reveals the complexity in assigning the optimal MTA and the necessity of a functional assay. The FACT analysis accurately provided prognostic predictions of the pts treated into responsive and pts with no molecular basis for a benefit in MTA treatment. Importantly, the hypothesis driving the SHIVA01 trial proved positive by the addition of the functional interpretation of the mutational data.

Legal entity responsible for the study

Institute Curie, Paris, France

Funding

None

Disclosure

G. Tarcic, O. Edelheit, Z. Barbash, M. Vidne, B. Miron: A full time employee of NovellusDx. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1269P - Integrative multi-platform meta-analysis of gene expression profiles in pancreatic ductal adenocarcinoma patients for identifying novel diagnostic biomarkers (ID 3482)

Presentation Number
1269P
Lecture Time
13:15 - 13:15
Speakers
  • O. Caba
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Applying differentially expressed genes (DEGs) to identify feasible biomarkers in diseases can be a hard task when working with heterogeneous datasets. Expression data are strongly influenced by technology, sample preparation processes, and/or labeling methods. The proliferation of different microarray platforms for measuring gene expression increases the need to develop models able to compare their results, especially when different technologies can lead to signal values that vary greatly. Integrative meta-analysis can significantly improve the reliability and robustness of DEG detection. The objective of this work was to develop an integrative approach for identifying potential cancer biomarkers by integrating gene expression data from two different platforms. Pancreatic ductal adenocarcinoma (PDAC), where there is an urgent need to find new biomarkers due its late diagnosis, is an ideal candidate for testing this technology.

Methods

Expression data from two different datasets, namely Affymetrix and Illumina (18 and 36 PDAC patients, respectively), as well as from 18 healthy controls, was used for this study. A meta-analysis based on an empirical Bayesian methodology (ComBat) was then proposed to integrate these datasets. DEGs were finally identified from the integrated data by using the statistical programming language R.

Results

After our integrative meta-analysis, 5 genes were commonly identified within the individual analyses of the independent datasets. Also, 28 novel genes that were not reported by the individual analyses ('gained' genes) were also discovered. Several of these gained genes have been already related to other gastroenterological tumors.

Conclusions

The proposed integrative meta-analysis has revealed novel DEGs that may play an important role in PDAC and could be potential biomarkers for diagnosing the disease.

Legal entity responsible for the study

Grupo de Tratamiento de Tumores Digestivos

Funding

Grupo de Tratamiento de Tumores Digestivos

Disclosure

M. Benavides: Received honoraria from Roche and honoraria for advisory role from Roche. C. Guillen-Ponce: Received honoraria from Roche; honoraria for advisory role from Roche Pharma, Bayer, Merck Serono, Sanofi Aventis, Celgene y Novocure; other remuneration from Celgene, Roche Pharma, Sanofi Aventis, Merck Serono. E. Aranda Aguilar: Received honoraria for advisory role from Amgen, Bayer, Celgene, Merck, Roche, Sanofi. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1270P - 3D Cultured Tumour from Patients to Predict Treatment Response (ID 5395)

Presentation Number
1270P
Lecture Time
13:15 - 13:15
Speakers
  • W. Vader
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Treatment selection for cancer patients is still a challenge. Average response rates for standard chemotherapy are low due to a lack of predictive markers. Genetic approaches to improve treatment efficacy have not yet delivered solutions for the day-to-day clinic. Functional testing of 3D cultures of patient tumour biopsies has the potential to identify tumours that are sensitive to standard drugs, search for alterative drugs when treatment options appear exhausted, and prevent overtreatment. Our technology based on image analysis of 3D tumour cultures accommodates accurate evaluation of drug sensitivity with small amounts of heterogeneous tumour material. We aim to validate our methods, and develop diagnostics to predict drug response for cancer patients.

Methods

3D cultures embedded in a protein-rich hydrogel are generated from tumour biopsies, and exposed to standard-of-care therapies, targeted therapies and drug combinations. An automated high content screening platform measures cell and tissue morphology, and reports responses such as tumour cell killing, growth arrest and local invasion. Per tumour type and drug, morphological features are selected as standard read-outs for the response. Proof of Concept (PoC) trials have been initiated to compare drug sensitivity of tumour cultures with treatment response in the clinic.

Results

We present results of PoC experiments showing drug sensitivity in 3D cultures of fresh and cryopreserved tumour material of gastric, endometrial, cervical, and ovarian cancer patients. Standard-of-care therapies were tested and results were compared per drug (combination). Differentiated drug responses are identified for treatment schedules including platinum-based drugs, taxanes, anthracyclines, 5-FU. In addition, responses to drugs that are not (yet) considered standard of care (PARP inhibitors) were measured.

Conclusions

Our technology enables drug sensitivity testing in 3D cultures of tumour tissues. This allows patient-specific treatment responses to developmental and standard-of-care drugs to be determined. Ongoing PoC trials will reveal the correlation of our in vitro test with treatment responses in the clinic.

Legal entity responsible for the study

VitroScan B.V.

Funding

VitroScan B.V.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1271P - 5-ALA administration for photodynamic diagnosis of peritoneal metastases due to advanced gastric cancer: A randomised, double-blind, multicentre phase I/II study (ID 2240)

Presentation Number
1271P
Lecture Time
13:15 - 13:15
Speakers
  • T. Takahashi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

For advanced gastric cancer, diagnosis of peritoneal dissemination is mandatory prior to the decision of therapy; therefore, staging laparoscopy (SL) has gained wider clinical acceptance. We have reported the efficacy of SL with photodynamic diagnosis (PDD) using 5-aminolevulinic acid (5-ALA). In this study, the safety and effectiveness of oral administration of 5-ALA PDD compared with that of conventional white-light laparoscopic diagnosis is assessed in a phase I/II study. This research also used a randomised double-blind comparison to explore the optimum dose of 5-ALA to be followed by a confirmatory study.

Methods

Subjects were patients with type 3 or 4 gastric cancer. A total of 20 or 40 mg/kg 5-ALA was administered orally 180–300 minutes before SL. The primary endpoint was safety; the secondary endpoints were sensitivity, specificity, positive predictive value, negative predictive value, and the proportion of patients with peritoneal dissemination.

Results

Thirty-one patients, comprising 19 men and 12 women, were enrolled. Fourteen patients were allocated to the 20 mg/kg group and 17 to the 40 mg/kg group. The median age was 70 years. The proportions of adverse events were 53.8% and 41.2% in the 20 and 40 mg/kg groups, respectively. Hypotension was noted as serious adverse event in 1 patient. The sensitivities of PDD in the 20 and 40 mg/kg groups were higher (95.7% and 100%, respectively) than those of conventional diagnosis (73.9% and 67.8%) (p = 0.0625 and p = 0.0313). In terms of precision, the evaluation values of diagnosis through conventional imaging compared with PDD were as follows: sensitivity, 83.5% vs. 98.6%; specificity, 75.5% vs. 38.8%; positive predictive value, 82.2% vs. 69.6%; and negative predictive value, 75.5% vs. 95.0%, respectively. In addition, one more patient was found positive for dissemination via PDD.

Conclusions

This investigator-initiated clinical trial confirmed the safety and effectiveness of 5-ALA administration in PDD for peritoneal metastases in gastric cancer. Results indicated that both doses of 5-ALA may be clinically applicable. Thus, we are now conducting a confirmatory study to apply for pharmaceutical approval of 5-ALA.

Clinical trial identification

JMA-IIA00225

Legal entity responsible for the study

Prof. Yuichiro Doki

Funding

This work was supported by the clinical trial promotion pro-gram subsidized by the Japan Medical Association with theMinistry of Health, Labour and Welfare Research Grant

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1272P - Diagnostic performance of dedicated breast PET scanner with a ring detector (ID 1183)

Presentation Number
1272P
Lecture Time
13:15 - 13:15
Speakers
  • R. HASHIMOTO
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Dedicated breast positron emission tomography scanners (dbPET) have been developed to make possible higher spatial resolution and sensitivity. The purpose of this study was to investigate the diagnostic performance of a dbPET scanner with an O-shaped detector in patients with known breast cancer.

Methods

A total of 82 female patients diagnosed with breast cancer consented to participate in this study (84 lesions: 10 ductal carcinomas in situ (DCIS), 74 invasive cancers). All patients underwent a WB-PET/MRI using Biograph mMR® (Siemens Healthcare) approximately 80 minutes after fluorine-18 fluorodeoxyglucose ((18)F-FDG;3.0MBq/kg) injection, followed by dbPET using Dedicated Breast PET System Elmammo®(Shimadzu, Kyoto, Japan) which required 5 minutes per breast.

Results

The overall imaging sensitivities of dbPET and WB-PET/MRI were 89% (75/84) and 87% (74/84) respectively. The sensitivities excluding 5 lesions which were outside the field of view of dbPET, were 95% (75/79) and 87% (69/79) respectively. The standardized uptake values (SUV) of dbPET (SUVmax mean13, range 1.64-41.6) and WB-PET/MRI (SUVmax mean 5.71, range 1.15-13.3) showed correlation with nuclear grade (NG) (Spearman's rank-correlation coefficient r = 0.507, 0.455, p < 0.001), Ki67 (r = 0.56, 0.62, p < 0.001) and estrogen receptor (r=-0.524, -0.475, p < 0.001). The sensitivities for DCIS were 90% (9/10), 60% (6/10) respectively. The sensitivities for DCIS and invasive cancers with 1cm or less maximal diameter were 86% (18/21) and 57% (12/21) respectively. We observed six lesions (three high grade DICS (1.7 cm -5cm), three high grade invasive cancers with 0.3-0.9cm) that were only detected by dbPET. On the contrary, four lesions could not be detected by either modality (low grade DCIS, invasive lobular carcinoma;2cm, low grade invasive ductal carcinoma with 0.6cm).

Conclusions

Both dbPET and WB-PET/MRI showed high imaging sensitivity, but dbPET had significantly higher sensitivity to high grade DCIS and invasive lesions up to 1cm. This level of diagnostic performance could help to detect early stage cancers as part of breast screening.

Clinical trial identification

We have registerd this trial at UMIN (Univresity Hospital Medical Information network) -CTR Serch Clinical Trials This trial protocol number of UMIN; UMIN000027227 (https://upload.umin.ac.jp/cgi-open-bin/ctr_e/index.cgi) Release date; 02.May.2017

Legal entity responsible for the study

Japan

Funding

Shimadzu corporation

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

SARCOMA (ID 5678)

Lecture Time
13:15 - 13:15
Speakers
  • H. Gelderblom
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15
Poster display session Poster Display session

1485P - Localized undifferentiated endometrial sarcomas (LUES): Results of a French Sarcoma Group (FSG) retrospective series of 39 patients (pts) (ID 4737)

Presentation Number
1485P
Lecture Time
13:15 - 13:15
Speakers
  • M. Meurer
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

UES are tumors of very bad prognosis. Although large surgical resection is the cornerstone of curative intent treatment, the optimal post-operative strategy remains unclear.

Methods

We conducted a retrospective analysis of LUES pts (Stade I-III) over the last 8 years in 10 FSG centers, from Netsarc and RRePs databases.

Results

Thirty-nine pts with primary LUES treated from 2008 to 2016 were included, with median age of 61 years (range, 43-80), and median ECOG of 0 (range, 0-3). Metrorrhagia, abdominal pain, and pelvic mass bleeding were the most common symptoms. The locoregional extension report usually included: gynecological examination (71%), and/or pelvic MRI (83%) and/or abdomino-pelvic CT scan (43%). The remote extension report was performed preoperatively for only 17/39 (50%) pts. Among 39 LUES, 24 (67%) were stage I, 3 (8%) stage II and 9 (25%) stage III. All the patients were operated on and surgical procedures were radical hysterectomy and bilateral oophorectomy for 26/39 (70%).Tumor resections were mostly R0, 23/39 (74%) and R1, 6/39 (19%). Tumor rupture occurred in 6 pts. For 7/39 LUES there was a positivity of hormonal receptors and/or cyclin D1. Twenty-two (56%) received post-operative radiotherapy (11 of them with complementary brachytherapy) and 11 (31%) adjuvant chemotherapy. With a median follow-up of 33 months (0.3-112), 17/39 pts are alive, 21/39 (54%) relapsed (7 local relapse and 13 metastases). The 3- and 5-year Overall Survival (OS) rates are 49.8% and 31.1% respectively. Median OS and Relapse-Free Survival (RFS) are 32.7 (16.3-49.1) and 23 (4.4-41.6) months, respectively. In univariate analysis; early FIGO stage (p < 0.0001), complete resection (R0-R1 vs R2; p = 0.027), ECOG (p = 0.02) and adjuvant radiotherapy (p < 0.0001) were associated with a better OS; vascular invasion (p = 0.024) and adjuvant radiotherapy (p = 0.021) were associated with a better RFS.

Conclusions

Treatment of primary LUES is radical hysterectomy and bilateral oophorectomy. Adjuvant radiotherapy appears beneficial for RFS and OS. The FIGO stage seems to have an impact on OS. A prospective study could be carried out to validate this therapeutic management.

Legal entity responsible for the study

invalid

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1486P - An epidemiological insight into epithelioid sarcoma (ES): The open issue of distal-type (DES) versus proximal-type (PES) (ID 2589)

Presentation Number
1486P
Lecture Time
13:15 - 13:15
Speakers
  • A. Frezza
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

ES is a rare sarcoma with distinctive pathologic and clinical features as well as the potential to respond to new targeted agents. We used the RARECAREnet and the SEER18 cancer registry (CR) databases (DB) to highlight the epidemiological hallamrks of ES in the EU and in the US.

Methods

The ICD-O3 code 8804 applies both to the DES and PES. Thus we broke down the primary sites using the ICD-O3 topography codes, to separate the anatomically proximal and distal forms, as a partial surrogate for the two entities of DES and PES. Incidence rate (IR), world-age adjusted IR and relative survival (RS) were calculated for ES patients diagnosed in the period 2000-07 and followed-up at least up to 31st Dec 2008. RS was estimated by the Ederer II method. 497 new cases of ES were identified from available registries in the EU, 301 in the US.

Results

Crude IR was 0.03/100,000 in the EU and 0.05/100,000 in the US. Age-adjusted IR were 0.02/100,000 and 0.05/100,000 respectively. M:F ratio was 1.6 in both sets of registries. Mean age at diagnosis was 46 in the EU and 44 yrs in the US. IR increased with age (higher in > 75-yr patients). IR in the childhood population (<14 yrs) was 0.01, i.e., 5% of all new ES cases. IR was similar for anatomically distal and proximal groups, but the latter was more incident in older patients. 5-yr RS was 50% and 52% in the EU and US, respectively. In both registries, 5-yr RS was higher in anatomically distal than in anatomically proximal (EU: 65% vs 38%; US: 69% vs 34%), evenly across all age groups (Table).

1486P Incidence and 5-years overall survival rates according to patient age and primary tumour site. * NE= not estimable for limited number of cases. DES: distal-type epithelioid sarcoma; PES: proximal-type epithelioid sarcoma.

Age (yrs)Incidence (/100.000)5-yrs OS (95% CI)DES 5-yrs OS (95% CI)PES 5-yrs OS (95% CI)
0 - 14EU: 0.01 US: 0.01EU: 53% (27-74) US: 90% (47-99)EU: 67% (27-88) US: 89% (42-98)EU: NE* US: NE*
15 - 39EU: 0.02 US: 0.05EU: 68% (59-75) US: 66% (56-74)EU: 79% (68-86) US: 71% (60-80)EU: 51% (37-63) US: 54% (36-69)
> 40EU: 0.03 US: 0.06EU: 39% (32-46) US: 39% (30-47)EU: 49% (37-61) US: 62% (45-75)EU: 33% (25-41) US: 26% (17-35)

Conclusions

ES is a very rare cancer, exceedingly low in childhood. Prognosis is serious, making the identification of new treatments a priority. New targeted agents are currently under study. However, the distinction between DES and PES is likely to be of major prognostic significance. In fact, using anatomical location as a proxy, though inappropriate, we could see clear-cut differences in the epidemiological outcome. An effort to stratify ES patients by their pathologic subtype is warranted in all studies, all the more on new agents.

Legal entity responsible for the study

Fondazione IRCCS Istituto Nazionale Tumori Milano

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1487P - Natural history of alveolar soft part sarcoma (ASPS): Impact of brain metastases and role of anti-angiogenic therapies (AAT) (ID 5025)

Presentation Number
1487P
Lecture Time
13:15 - 13:15
Speakers
  • G. Malouf
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

ASPS is a rare sarcoma subtype with clinical specificities, such as an indolent behavior, brain metastasis and resistance to doxorubicin. AAT have shown clinical activity in this setting, but little is known on the optimal therapeutic strategy, and the management of brain metastasis (BM).

Methods

We retrospectively analyzed patients (pts) treated in 3 referral centers of the French Sarcoma Group. Factors associated with BM development and overall survival (OS) were analyzed. In addition, progression-free survivals (PFS) under AAT in patients with and without BM were reported.

Results

We identified 75 pts [median age at diagnosis: 23 (5-96 years), 61% females]. Among those, 31 (41%) pts had documented synchronous lung metastasis (LM), and none had BM. Median OS in pts with localized and metastatic disease were 279 months, (95% CI, 279-NR) and 74 months (95% CI, 62-144) (Log-rank, p = 0.002), respectively. Only surgical complete resection (R0) was associated with better OS in localized disease (HR = 4.3; (95% CI, 1-19.3), p = 0.056). Fifty-two (69%) pts had documented LM in the course of the disease; among those, 13 (17%) pts developed BM within a median interval of 35 months (95% CI, 17-48) from LM. Initial tumor size was associated with BM-free-survival (≥5cm vs <5 cm): HR = 6.7 (95% CI, 1.7-26). Twelve pts were treated with AAT (sunitinib, n = 10) including 5 (42%) with documented BM; median PFS under AAT were 2 months (95% CI, 1.3-2.7) for pts with BM and 11 months (95% CI, 9-18) for pts without BM, respectively (p=0.004). BM was the cause of death or major contributing factor to it in 36% of pts.

Conclusions

These data highlight the indolent course of the disease leading to BM, which turned a shift in the course of the disease, along with limited efficacy of AAT in this setting. Furthermore, they suggest that the appropriate timing for AAT introduction has to be discussed in an individual basis considering the PFS/OS ratio in pts with metastatic disease.

Legal entity responsible for the study

Institut Gustave Roussy

Funding

None

Disclosure

G. Malouf: Consulting: Pfizer, BMS, Novartis Research grant: Pfizer, Novartis. O. Mir: Speaker: Lilly, Roche Consulting: Amgen, Astra-Zeneca, Bayer, Blueprint, BMS, Lilly, Novartis, Pfizer, Roche, Servier. J-P. Spano: conflict of interest with the following companies: Pfizer, BMS, MSD, Roche. J-Y. Blay: Research support: Novartis, Roche, GSK, Bayer, Pfizer. A. Lecesne: Honoraria: Amgen, Novartis, Pharmamar, Pfizer, Lilly. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1488P - Treatment patterns, clinical outcomes and prognostic factors of visceral angiosarcoma (V-AS): A report from the Asian Sarcoma Consortium (ASC) (ID 2829)

Presentation Number
1488P
Lecture Time
13:15 - 13:15
Speakers
  • A. Pang
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

We previously reported on the real world treatment and outcomes of 423 angiosarcoma pts in Asia. In this current report, we focus on the 173 pts with V-AS and evaluate the treatment patterns and prognostic factors associated with this disease subset.

Methods

This is a retrospective chart review of V-AS pts seen at 8 Asian academic study sites. Survival analysis is measured from date of presentation to the study site.

Results

Median study follow-up was 8.7 mths. Median age 52 yrs; 86% of pts presented with primary disease to study site. 42% (n = 73) had localized disease and 56% (n = 97) had locally advanced/unresectable or metastatic disease; disease status was unknown for 3 pts. Distribution of primary site as follows, liver (n = 38, 22%), cardiovascular system (n = 30, 17%), breast (n = 25, 14%), spleen (n = 18, 10%) and others (n = 62, 36%) from a myriad of organ systems each representing <10% of cases. More pts with breast AS presented with localized disease. For pts with localized disease, primary treatment was surgery in 74% (n = 54) of pts. Of 38 pts with known margins, R0 and R1 were achieved in 71% and 21% respectively. In pts who had surgery, 43% developed subsequent disease relapse/progression with a median PFS of 9.5 mths. Of 97 pts with advanced/unresectable disease, 59% of pts received chemotherapy as part of treatment while 22% and 20% had surgery/radiation only or supportive care only respectively. 46% of chemotherapy-treated pts had 1 line of treatment while 35% and 19% had 2 or > 3 lines of treatment respectively. The most common first line chemotherapy used was paclitaxel (53%) followed by liposomal doxorubicin (18%). Median OS was 11.9 mths in the overall V-AS cohort, with median OS 29.2 mths and 6.3 mths in the localized and advanced/unresectable cohort respectively. In the univariate analysis of pts with unresectable/metastatic disease, only ECOG was associated with OS.

Conclusions

This study highlights the heterogeneity and treatment challenges of visceral angiosarcoma. Overall prognosis is poor, in particular in pts with advanced/unresectable disease.

Clinical trial identification

NA

Legal entity responsible for the study

National Cancer Centre Singapore

Funding

None

Disclosure

R. Quek: Grants/research support: Novartis, Pfizer, Janssen, Bayer and Eisai. Honoraria/consultation fees: Novartis, Bayer, BMS, Merck, Roche and Eisai. Participation in a company sponsored speaker\'s bureau: Novartis, Bayer, Merck and Eisai. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1489P - High dose loco-regional chemotherapy for locally advanced angiosarcoma: A multicenter study (ID 3082)

Presentation Number
1489P
Lecture Time
13:15 - 13:15
Speakers
  • E. Huis in 't Veld
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Angiosarcomas are rare aggressive sarcomas that count for 2% of all soft tissue sarcomas. The only potential curative treatment is complete surgical excision, but a large subset of patients present with locally advanced disease. The aim of this study was to evaluate the effectiveness of Isolated Limb Perfusion (ILP) as an alternative treatment option for locally advanced angiosarcoma in the extremities.

Methods

All patients who underwent an ILP for angiosarcomas between October 1991 and October 2016 in three tertiary referral centres were identified from 3 prospectively maintained databases. Demographics, tumour, treatment and response characteristics and the disease course were all obtained from either the database or patient files. Statistical analysis was performed using SPSS statistics 24.

Results

A total of 39 patients were included. The median age was 66 (range, 24-95) years. Of these patients, 23 (59%) patients had a complete resonse after ILP, 10 (25.6%) patients had a partial response, 4 (10.3%) had stable disease and 2 (5.1%) patients had local progression immediately after ILP. Of all patients, a total of 22 patients developed local progression (56,4%) while 10 (25.6%) developed distant metastases. Median time to progression; 7,4 months (IQR 3-14,9) and median time to distant metastasis of 6,4 months (IQR 1,5-44,9). The 10 (25,6%) patients with a complete response (compared to PR/SD/PD) had a non-significant trend towards better median overall survival (81,2 vs 14,5 months) (p = 0,054), and had a significantly prolonged median progression free survival (15,4 vs. 7,3 months) (p = 0,015). A total of 5 patients underwent multiple ILP’s whereby the complete response rate of the first, second and third ILP were 60% (n = 4/6), 80% (n = 4/5) and 67% (n = 2/3) respectively. Only 13 (33,3%) patients needed further surgical intervention, consisting of wide local excision in 8 patients (20,5%) and amputation in 5 patients (12,8%).

Conclusions

ILP should be considered as treatment option in the multidisciplinary management of patients with locally advanced limb angiosarcomas, resulting in a high limb salvage rate, high number of complete responses and prolonged progression-free survival.

Clinical trial identification

Not applicable

Legal entity responsible for the study

not applicable

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1490P - Primary cardiac sarcoma (PCS): A multi-national retrospective review of clinical experience (ID 1470)

Presentation Number
1490P
Lecture Time
13:15 - 13:15
Speakers
  • T. Chen
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Primary cardiac sarcoma (PCS) is a rare but often fatal disease. The current study aimed to analyze the impact of baseline demographics, local and systemic therapies in a contemporary cohort.

Methods

Clinical records of PCS across five institutions in three continents were reviewed and collected. Kaplan-Meier method was used to estimate survival. Cox proportional hazard model was used to associate variables to progression-free survival (PFS) or overall survival (OS).

Results

47 pts with PCS (1996-2016) with a median follow-up time of 12.9 months (ms) were identified. The median age at diagnosis was 41 (range 18-79); 43% (n = 20) presented with metastatic disease. Tumor equally originated from right- (n = 23) and left-sided heart (n = 23). The common histologies were angiosarcoma (n = 18, 38%), intimal sarcoma (n = 8, 17%), and sarcoma NOS (n = 10, 21%). 66% (n = 31) had surgical (S) treatment for PCS, and only 4 (13%) pts had R0 resection. The median primary lesion size was 49 mm (20-84 mm). 70% (n = 33) of pts received at least one line of chemotherapy (C), and 51% (n = 24) received multi-modality treatment (45% S + C, 4% S + XRT, 2% S + C + XRT). The median OS was 17.7 ms (95% CI 12.4-21.8 ms). For all pts, age ≥ 65 was the only significant negative prognostic factor (HR 7.43, p < 0.01, Table 1). In localized PCS, angiosarcoma histology was a significant factor for worse PFS (HR 3.05, p = 0.04) and OS (HR 2.56, p = 0.11). 29 metastatic or relapsed pts received C. For first-line palliative C (52% combination, 48% single agent), the median PFS was 3.8 ms (95% CI 1.5-6.9 ms). The best response for first-line C was PR 10 (35%), SD 4 (14%), PD 12 (41%). 60% (6/10) of pts with PR were treated with anthracycline (angiosarcoma n = 5; intimal sarcoma n = 1). No significant improvement in OS was identified in pts presenting throughout the 20 year period of this review (pre- vs post 2012; HR 1.1, p = 0.81).

1490P Prognostic factors for all pts

VariableHR (95% CI)P Value
Age ≥ 657.43 (2.54-21.72)0.0002
Metastatic disease at diagnosis1.87 (0.90-3.88)0.09
Multi-modality treatment0.64 (0.32-1.27)0.20
Angiosarcoma histology1.58 (0.72-3.47)0.26

Conclusions

The prognosis of PCS remains poor without significant improvement in OS compared to historical levels. Further research is required for this rare entity.

Clinical trial identification

Not applicable

Legal entity responsible for the study

Not applicable

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1491P - Primary cardiac sarcoma: A retrospective study in two Korean tertiary centers (ID 5079)

Presentation Number
1491P
Lecture Time
13:15 - 13:15
Speakers
  • H. Ahn
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Primary cardiac sarcoma(PCP) is a rare diasease with dismal prognosis. Optimal treatment strategy for PCP is not clear.

Methods

We retrospectively reviewed patients diagnosed with PCS in two institutions in Korea, between 1996 and 2013. A total of 41 patients were identified. Clinical characteristics and treatment outcomes were investigated. Survival was estimated and compared by Kaplan-Meier method and Log-rank test.

Results

Median age was 44 years. Eighteen patients (43.9%) were male. The most common type was angiosarcoma (N = 20, 48.7%), followed by poorly-differentiated sarcoma (N = 7, 17.1%). Most tumors were located in atriums (N = 27, 65.9%). At diagnosis, 31 patients (75.6%) had localized disease. Surgical resection of primary cardiac tumor was performed in 28 cases (68.3%) and microscopic complete resection were achieved in 12 localized cases. Median overall survial was 13.2 months for entire patients, 17.0 months after complete resection, 17.9 months after incomplete resection and 5.7 months in non-resected cases. Patients who received adjuvant chemotherapy and/or radiotherapy after surgery had significantly better median OS of 21.6 months than 11.0 months of those received surgical resection alone without adjuvant treatment (p = 0.007).

Conclusions

Surgical resection and multidisciplinary treatment were associated with better survival in patients with primary cardiac sarcomas. Aggressive multidisciplinary approaches may have a role in subset of patients with primary cardiac sarcomas.

Legal entity responsible for the study

Hee Kyung Ahn

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1492P - The differences in recurrence and survival of extremity liposarcoma subtypes (ID 3465)

Presentation Number
1492P
Lecture Time
13:15 - 13:15
Speakers
  • M. Vos
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Liposarcomas (LPS) are most frequently located in the extremity and recur often, locally as well as on distant sites. There are currently no studies comparing the differences between the different LPS subtypes specifically on this primary site. The aim of this study is to map the differences in recurrence and survival of extremity LPS.

Methods

Two retrospective databases (Rotterdam-R, Warsaw-W) of patients treated for primary LPS located in the extremities from 1985-2015 in two tertiary referral centers were used to analyze recurrence patterns (local/distant) and survival.

Results

In total, 456 patients were identified: 192 with well differentiated LPS (WDLPS), 172 myxoid LPS (MLPS), 54 pleomorphic LPS (PLPS), 23 dedifferentiated LPS(DDLPS) and 15 other subtypes (excluded from further analysis). A difference between the two datasets was the frequency of (neo)adjuvant radiotherapy (R: 34.9% vs. W: 81.5%). In the Rotterdam cohort, local recurrence (LR) was observed most frequent in DDLPS (4/13, 5-year LR free survival 56.6%), followed by WDLPS (26/113, 64.9%), PLPS (3/20, 78.6%) and MLPS (10/77, 84.0%, p = 0.136). In the Warsaw cohort, 5-year LR free survival was the lowest in PLPS (9/34, 67.7%), followed by DDLPS (1/10, 83.3%), MLPS (12/95, 84.9%) and WDLPS (3/79, 94.1%, p = 0.001). Distant metastases (DM) were most commonly observed in PLPS in both datasets (5-year DM free survival R: 9/20, 50.3% and W: 16/34, 44.5%), but in the Rotterdam cohort MLPS (16/77, 76.3%) was the second most common subtype with DM, compared to DDLPS (2/10, 62.5%) in Warsaw. DM in WDLPS was rare in both datasets (R:3/113, 96.3%, W: 1/79, 98.5%). 5-year overall survival (OS) did not significantly differ between the two datasets (R: 38/223, 78.5% vs. W: 32/218, 80.9%, p = 0.561), but did significantly differ between the subtypes (R: p = 0.005, W: p < 0.001). In both datasets, 5-year overall survival was poorest in PLPS (R: 8/20, 57.5%, W: 15/34, 38.8%) and DDLPS (R: 4/13, 59.9%, W: 3/10, 44.4%), followed by MLPS (R: 16/77, 75.6%, W: 13/95, 83.1%) and WDLPS (R: 10/113, 88.2%, W: 1/79, 98.5%).

Conclusions

Patients with the four LPS subtypes show distinct patterns of LR, DM and OS. Despite the differences in recurrence, treatment and follow up, OS did not differ significantly between the two expertise centers.

Legal entity responsible for the study

Erasmuc MC Rotterdam

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1493P - The sarcoma policy checklist: Focusing policy efforts on sarcoma (ID 4555)

Presentation Number
1493P
Lecture Time
13:15 - 13:15
Speakers
  • B. Kasper
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The Sarcoma Policy Checklist was developed by a multi-stakeholder group of experts to help policymakers prioritise actions to close the gap in access to high-quality information and care for sarcoma patients in Europe.

Methods

Experts defined five key areas where policy efforts are most needed to improve the care of sarcoma patients across Europe. A pragmatic review of the published literature was then conducted to determine to what extent recommendations were implemented in practice. Research focused on six countries (France, Germany, Italy, Spain, Sweden and the United Kingdom) and was complemented by local expert interviews.

Results

Five key priority areas were identified by experts: Each country should have designated, accredited centres of reference for sarcoma; specialised professional training should be provided to all health care professionals involved in sarcoma care; a multidisciplinary approach to care should be offered to every patient; greater incentives for research and innovation, and more rapid access to effective treatments are needed. Most countries have specialist sarcoma centres, however, there is often a lack of defined criteria to designate specialist centres and evaluate the quality of care. Professional training is a gap in all countries, as training on rare cancers is most often not included in the general medical curriculum or in oncologists’ training. More basic research is needed to understand the underlying epidemiology of sarcomas, and help focus research on effective treatments. Greater alignment between regulatory frameworks and access frameworks such as Health Technology Assessment is needed, particularly in terms of evidentiary requirements for new treatments.

Conclusions

The heterogeneity of sarcomas poses particular challenges to research, professional training and patient access to quality treatment and care. The creation of the European Reference Network (ERN) on sarcoma, and recent advances in defining essential requirements and patient-driven principles for sarcoma care will help improve the situation of sarcoma patients across Europe. This policy paper hopes to contribute to those efforts and help drive meaningful policy change to improve patient care.

Legal entity responsible for the study

The Health Policy Partnership Ltd

Funding

Eli Lilly and Company Europe

Disclosure

N. Drove: Employment with Eli Lilly and Company, the pharmaceutical company that is funding this project. S. Boldon: Consultancy fees from Eli Lilly and Company as an employee of the Health Policy Partnership, which has led the coordination and policy writing for this project. S. Wait: Consultancy fees from Eli Lilly and Company as managing director of the Health Policy Partnership, which has led the coordination and policy writing for this project. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1494P - Dermatofibrosarcoma protuberans might not benefit from postoperative radiotherapy after local resection with negative margin (ID 3825)

Presentation Number
1494P
Lecture Time
13:15 - 13:15
Speakers
  • J. Chen
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

To evaluate the role of postoperative radiotherapy (RT) in the treatment of dermatofibrosarcoma protuberans (DFSP) after local resection with negative margin.

Methods

We retrospectively analyzed 84 consecutively treated DFSP patients who received local resection (≤2cm) with negative margin from 2000 to 2016 in our institution. Statistical analysis was performed with a commercially available statistical software package.

Results

The median follow-up was 60 months (range, 10-201). For patients (28/84) with postoperative RT, four (4/28) patients were found to had local relapse, while three of them had ≤ 1 cm surgical margin. For patients without postoperative RT, four (4/56) patients had local failure. Postoperative RT failed to improve the local recurrence-free survival (LRFS) of DFSP after local resection with negative margin (Fisher=0.431). Patients with fibrosarcomatous DFSP (FS-DFSP) were found to have higher local recurrent rate than DFSP (66.7% vs. 7.4%, Fisher=0.023). Twenty-six patients were examined for ki-67, and positive range is 1-30%. Patients with high ki-67 expression (>15%) were found to have higher local recurrence rate than the others (80.0%vs0%, Fisher=0.000).

Conclusions

Postoperative RT did not improve the LRFS of DFSP after local resection with negative margin. FS-DFSP was more likely to have a local relapse than the other types. Ki-67 might become a good predictor for local control of DFSP.

Legal entity responsible for the study

Kai xin Kaixin Du

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1495P - Low-dose chemotherapy with methotrexate and vinblastine for desmoid tumors: A single institution experience (ID 4976)

Presentation Number
1495P
Lecture Time
13:15 - 13:15
Speakers
  • J. Souza
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Desmoid tumor (DT) is a rare and locally invasive proliferative disease. Although, the absence of metastatic potential, it has the propensity for locally invasive growth and recur. Chemotherapy may be considered in inoperable and/or recurrent disease.

Methods

Patients with histological diagnosis of DT and treated with weekly low-dose chemotherapy with vinblastine and methotrexate between January 1998 and December 2015 were identified and their medical records were analyzed.

Results

Of 23 patients analyzed, most of them were women (female-to-male ratio 2.8:1). The median age at presentation was 29 years (range, 18-59 years). Tumors location was: thoracoabdominal wall (n: 11, 47.8%), extremities (n: 7, 30.4%), abdominal cavity (n: 1; 4%), and head and neck (n: 4, 17.3%). Tumor sizes were documented in 18 cases and ranged from 3 to 20 cm in largest linear dimension (median, 10 cm). Eight (34.7%) female had pregnancy history and 2 (8%) had familial adenomatous polyposis history. Eleven (47,8%) underwent surgery as first-line treatment. Five (21.7%) patients received first-line treatment with vinblastine and methotrexate, four (17.3%) patients as second-line, and 14 (60.8%) patients as third and fourth-line. Fourteen (60.8%) patients had stable disease, four (17.3%) had partial response, and five (21.7%) patients had progressive disease during chemotherapy treatment. After a median follow-up of 58 months, 12 patients had progression disease and 2 patients died. The median PFS was 29 months, without any progression after 32 months.

Conclusions

Discussion: Weekly low-dose chemotherapy with vinblastine and methotrexate appears to have significant activity. Chemotherapy could be an acceptable alternative to radical surgery in selected patients with desmoid tumors.

Legal entity responsible for the study

INCA

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1496P - EORTC experience with advanced/metastatic epithelioid sarcoma patients treated in prospective trials: Clinical profile and response to systemic therapy (ID 2110)

Presentation Number
1496P
Lecture Time
13:15 - 13:15
Speakers
  • N. Touati
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Epithelioid sarcoma (ES) is a soft-tissue sarcoma (STS) associated with a high local recurrence rate after primary resection and high incidence of distant metastasis. Little is known about clinical course and response to systemic treatments. This retrospective analysis aims to provide a reference for future ES-specific studies.

Methods

Data from patients with locally advanced/metastatic ES entered in prospective multi-sarcoma phase II/III trials were pooled: EORTC trial 62012 (doxorubicin vs. doxorubicin/ifosfamide), 62043 (pazopanib), 62072 (pazopanib vs. placebo) and 62091 (doxorubicin vs. trabectedin). Patients had either a local or centrally confirmed diagnosis of ES, inoperable/metastatic disease at study entry and were eligible for the respective trial. Response was assessed using RECIST 1.1. Progression-free survival (PFS) and overall survival (OS) were calculated from date of study entry.

Results

Among 1099 patients with advanced STS, 27 ES patients (2.5%) were eligible (17 male (63%), median age at diagnosis 50 yrs, range 19-72). 18 (66.7%) received chemotherapy as 1st line treatment (5 doxorubicin, 8 doxorubicin/ifosfamide, 2 pazopanib, 3 trabectedin) and 9 (33.3%) received pazopanib in 2nd line or later. Primary tumor was located in lower extremity (N = 8; 29.6%), upper extremity (N = 5; 18.5%), retro/intra-abdominal (N = 4; 14.8%), other locations (N = 10; 37.0%). At study entry, metastases were mainly found in lung (N = 17; 63%), lymph nodes (N = 9; 33.3%), bone (N = 8; 29.6%) and soft tissue (N = 7; 25.9%). Best response for 1st line patients was 4 partial responses (PR, 22.2%), 10 stable disease (SD, 55.6%) and 4 progressive disease (PD, 22.2%). In subsequent lines, pazopanib achieved 1 PR (11.1%), 4 SD (44.4%) and 4 PD (44.4%). All patients but one progressed. Median PFS and OS were 3.8 (95% CI: 2.2-4.8) and 10.8 months (95% CI: 8.1-21.3), respectively. 5 patients were still alive at time of the according trial analysis.

Conclusions

With all limitations of a retrospective analysis of such small dataset, objective response and survival outcomes of this locally advanced/metastatic ES population are relatively poor. The clinical testing of novel agents remains a high priority.

Legal entity responsible for the study

EORTC

Funding

Epizyme

Disclosure

P. Schoffski: Institutional support from Epizyme for advisory functions. T. Brodowicz: Personal fees from Roche (lecture fee), personal lecture and advisory fees from Amgen, Bayer, Novartis, PharmaMar, Eisai and Eli Lilly outside the submitted work. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1497P - Efficacy and safety of patients treated long-term with trabectedin (t) on the expanded access program: A retrospective analysis (ID 1371)

Presentation Number
1497P
Lecture Time
13:15 - 13:15
Speakers
  • E. Davis
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Treatment of soft tissue sarcoma (STS) with long-term systemic therapy can be limited by cumulative toxicity. Treatment with T for prolonged courses without the cumulative toxicity has been previously described from clinical trials. Here we report the efficacy and safety for patients (pts) treated long term (≥ 6 months) in a real world setting in the T Expanded Access Program from 2005-2010.

Methods

In this retrospective analysis of pts with pre-treated, relapsed/refractory STS of multiple histologies treated ≥6 mo with T (1.5 mg/m2 iv q3wk), we compared pts treated 6-12 mo and >12 mo.

Results

Of 1803 pts, 401 (21.6%) remained on treatment ≥6 mos; 268 (14.5%) for 6-12 mo and 133 (7.2%) >12 mo. Demographics did not differ. Leiomyosarcoma or liposarcoma were the most common histologies. The mOS (mo) was 18.1 and 47.0, ORR was 7.8% and 6.8%, and clinical benefit rate (CR+PR+SD) (95%CI) was 47.4% (41.3;53.6) and 38.3% (30.1;47.2) in the 6-12 mo and >12 mo groups, respectively. The incidence of adverse events (AE)s and serious adverse events (SAE)s were similar in both groups (Table). The most common grade 3/4 AEs occurring in ≥ 5% were neutropenia, thrombocytopenia, anemia, ALT/AST increase, fatigue and nausea. A majority received dose reduction or delay; the primary reason for treatment discontinuation was disease progression. The longest observed duration of treatment was 55 mo (64 cycles; synovial sarcoma) and 54 mo (73 cycles; uterine leiomyosarcoma).

1497P Safety and Efficacy

6-12 Months>12 Months
(N = 268)(N = 133)
Median Treatment Duration (mo), range8.4(6; 12)16.3 (12; 55)
Treatment Response,
Complete response, n (%)1 (0.4)3 (2.3)
Partial response, n (%)20 (7.5)6 (4.5)
Stable disease, n (%)106 (39.6)42 (31.6)
Progressive disease, n (%)20 (7.5)12 (9.0)
Not available, n (%)121 (45.1)70 (52.6)
Treatment-emergent adverse events (TEAEs)225 (84.0)119 (89.5)
Serious TEAEs88 (32.9)47 (35.3)
Treatment discontinued255(95.1)103 (77.4)
Due to disease progression192(71.6)72 (54.1)
Due to adverse event10 (3.7)2 (1.5)
Patients with cycle delay154 (57.5)82 (61.7)
Patients with dose reduction172 (64.2)104 (78.2)

Conclusions

T can be safely administered and well tolerated in pts who receive a prolonged duration (≥6 mo) of therapy. Improved mOS may be achieved in pts who experience prolonged disease stabilization following T but adjustments in dose or schedule is frequently required.

Clinical trial identification

NCT00210665

Legal entity responsible for the study

Janssen Research & Development, LLC

Funding

Janssen Research & Development, LLC

Disclosure

S. Schuetze: Honorarium for Janssen ad hoc scientific advisory board to me. Research funding to institution (University of Michigan) from Janssen. S. Patel: Consultant to: Janssen, Eisai, Novartis, CytRx, Epizyme, Bayer, Eli Lilly Grants for clinical trial from: Janssen, Eisai, Morphotek. R. Maki: Consulting or advisory role: SARC, ASCO, AADX, ARCUS, Bayer, GSAI, GEM, Novartis, GSK, Immune Design, Janssen, Kyropharm, Lilly Tracon and Presage Research Funding and Travel, Accommodations, Expenses: Tracon, Immune Design, Lilly, and SARC. R.L. Jones: Consultant for: Adaptimmune, Blueprint, Eisai, Epizyme, Daichii, Deciphera, Janssen Scientific Affairs, LLC, Immunedesign, Lilly, Merck, Pharmamar. C.R. Shin, R. Knoblauch, G. Wang, M. Smith: Employee of Janssen and own stock in Johnson & Johnson. G.D.S. Demetri: Consulting: Novartis, Janssen, PharmaMar, Daiichi-Sankyo, Adaptimmune, Eisai Patent licensed to Novartis from Dana-Farber with royalty paid to Dana-Farber Research support to Dana-Farber: Novartis, Janssen. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1498P - Efficacy and safety of trabectedin in an elderly patient subgroup (≥65 years) with advanced leiomyosarcoma (LMS) or liposarcoma (LPS) from the Expanded Access Program (EAP) (ID 2079)

Presentation Number
1498P
Lecture Time
13:15 - 13:15
Speakers
  • R. Jones
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Elderly patients (pts) (≥65 yrs) with soft tissue sarcoma may have limited treatment options due to increased comorbidities and toxicities from available therapeutic agents. Previous retrospective analyses have suggested that trabectedin (T) has similar safety and efficacy outcomes irrespective of pt age.

Methods

In this multicenter, open-label study, pts received IV T (1.5 mg/m2) every 3 wks. We retrospectively analyzed the efficacy and safety of T in pts ≥65 yrs treated from 2005-2010 on this EAP.

Results

Mean age was 71 and 49 in the ≥65 (n = 350) and <65 (=1453) groups respectively. Pt demographics and disease characteristics were similar in ECOG score, race, gender, and histology. Median duration of therapy was 3 cycles in both groups. Pts receiving prolonged therapy (>12 mo) was 26 (7.4%) and 107 (7.4%) in the ≥65 and <65 groups, respectively. Elderly patients treated with T experienced similar median OS, ORR, and CBR (Clinical Benefit Rate, CR+PR+SD) compared to the <65 group with a median OS of 11.5 mo and 12.3 mo, ORR of 7 (3.9%) and 41 (5.4%), and CBR of 78 (43.1%) and 313 (40.1%) in the in the ≥65 and <65 groups, respectively. Toxicities in the elderly group were consistent with previously reported safety profiles, and incidence in the elderly group were comparable to those of the <65 group. Treatment-emergent adverse events (TEAEs), and serious TEAEs were similar in both groups. The percentage of pts requiring dose reduction and dose delay in pts who received ≥2 cycles was also similar with 96 (33.6%) and 444 (36.2%) of pts requiring dose delay and 135 (47.2%) and 563 (46.0%) requiring dose reduction in the ≥65 and <65 groups, respectively. In both groups, the majority of pts discontinued treatment due to disease progression with only 32 (9.1%) and 118 (8.1%) of pts discontinuing treatment due to an AE in the >65 and <65 groups, respectively.

Conclusions

The efficacy and safety profile of T in pts ≥65 was similar to that observed in pts <65 in this EAP. Based upon this real world experience, T should be considered as a treatment option for elderly pts with soft tissue sarcoma and good performance status irrespective of age.

Clinical trial identification

NCT00210665

Legal entity responsible for the study

Janssen Research & Development, LLC

Funding

Janssen Research & Development, LLC

Disclosure

R.L. Jones: Consultant for: Adaptimmune, Blueprint, Eisai, Epizyme, Daichii, Deciphera, Janssen Scientific Affairs, LLC, Immunedesign, Lilly, Merck, Pharmamar. R. Maki: Consulting or advisory role: SARC, ASCO, AADX, ARCUS, Bayer, GSAI, GEM, Novartis, GSK, Immune Design, Janssen, Kyropharm, Lilly Tracon and Presage. Research funding & travel, accommodations, expenses: Tracon, Immune Design, Lilly and SARC. S. Patel: Consultant to: Janssen, Eisai, Novartis, CytRx, Epizyme, Bayer, Eli Lilly. Grants for clinical trial from: Janssen, Eisai, Morphotek. G. Wang, C.R. Shin, R. Knoblauch: Employment with Janssen and Johnson & Johnson stock ownership. G.D.S. Demetri: Consulting: Novartis, Janssen, PharmaMar, Daiichi-Sankyo, Adaptimmune, Eisai Patent licensed to Novartis from Dana-Farber with royalty paid to Dana-Farber Research support to Dana-Farber: Novartis, Janssen.

Collapse
Poster display session Poster Display session

1499P - The routine real-life use of trabectedin (T) in patients with advanced soft tissue sarcoma (STS) across Europe: An analysis of overall vs. per country results from Y-IMAGE study (ID 1540)

Presentation Number
1499P
Lecture Time
13:15 - 13:15
Speakers
  • N. Penel
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The prospective, non-interventional, phase IV Y-IMAGE study evaluated the use of T in real-life clinical practice across Europe in patients with advanced STS.

Methods

Data from adult STS patients treated with T 1.5 mg/m2 given as 24-h i.v. infusion q3w were collected. Patients must have received at least 1 cycle of T and currently be on T treatment. The primary endpoint was progression-free survival (PFS) as defined by investigators. The analyses were conducted in the overall population (OP) and separately in countries with the highest recruiting rate to cover inter-country variations: France (F), Germany (G), Italy (I) and the UK.

Results

A total of 218 patients from 41 centers and 9 European countries were evaluated. Demographics and baseline characteristics of patients recruited in the 4 countries of interest were well-balanced and comparable to those observed in OP. Patients received a median of 6 cycles of T (range: 1-44), mostly on an outpatient basis (n = 132; 60.6%). Across all centers the median cycle duration, and median dose and dose intensity were similar to those observed in OP. Analysis of PFS data showed a similar outcome in G (median PFS: 5.9 months) to that observed in OP (5.9 months), and a rather higher PFS in the UK (8.3 months), F (7.6 months) and I (6.8 months). The patients from the UK received the highest median number of cycles (10.5) and cumulative dose of T (26.2 mg) as compared to F, G and I. This was associated with favorable efficacy outcomes in those patients, particularly in terms of improved PFS (8.3 months), responses (ORR: 38.5%; DCR: 84.6%) and a high growth modulation index of 2.3. T treatment resulted in a comparable median overall survival in all patients (21.3 months), being somewhat larger among patients treated in sites across G (27.3 months). Febrile neutropenia (2.3% of patients), neutropenia, nausea, and pneumonia (1.4% each) were the most common T-related grade 3/4 adverse drug reactions.

1499P

Full analysis set, n (%)
France (n = 26)Germany (n = 29)Italy (n = 69)UK (n = 26)Overall population (n = 218)
Age at study entry (years); Median (range)58.5 (22-77)58 (23-79)59 (26-79)56.6 (25-73)58.0 (21.0-79.0)
Female15(57.7)15 (51.7)44 (63.8)13 (50.0)123 (56.4)
Histology (≥10% of patients)
Leiomyosarcoma11 (42.3)11 (37.9)29 (42.0)16 (61.5)92 (42.2%)
Liposarcoma5 (19.2)23 (33.3)7 (26.9)51 (23.4)
Synovial sarcoma4 (15.4)5 (17.2)23 (10.6%)
Cycles per patient Median (range)5.5 (2-29)6.0 (2-18)6.0 (1-30)10.5 (1-44)6.0 (1-44)
Cumulative dose received mg/patient12.1 (3.7-48.2)20.8 (5.6-51.0)14.3 (1.9-60)26.2 (3-116.4)14.7 (1.8-116.4)
Cycle duration (days)24.9 (21-41)26.8 (21-44.4)23.7 (20.5-32.5)24.2 (21-30.6)24.1 (20-47.5)
Dose intensity (mg/m2/week)0.7 (0.2-1.0)0.6 (0.4-1.1)0.6 (0.3-1.0)0.7 (0.5-1.0)0.7 (0.2-1.1)
Median PFS (months) [95% Confidence interval]7.6 [3.3-NR]5.9 [3.4-11.2]6.8 [3.4-10.2]8.3 [5.5-11.4]5.9 [4.9-7.8]
Objective response rate (ORR) (Complete + partial response) [95% Confidence interval]6 (23.1) [9.0-43.6]9 (31.0) [15.3-50.8]15 (21.7) [12.7-33.3]10 (38.5) [20.2-59.4]58 (26.6) [20.9-33.0]
Disease control rate (DCR) (ORR + stable disease) [95% Confidence interval]17 (65.4) [44.3-82.8]20 (69.0) [49.2-84.7]48 (69.6) [57.3-80.1]22 (84.6) [65.1-95.6]143 (65.6) [58.9-71.9]
Time to progression (TTP), median (months) [95% Confidence interval]7.8 [4.9-NR]6.9 [4.2-11.2]6.8 [3.4-10.2]8.3 [5.5-11.4]5.9 [4.9-8.1]
Overall survival (OS), median (months) [95% Confidence interval]20.3 [9.6-NR]27.3 [9.2-NR]22.5 [19.0-NR]20.0 [18.2-23.6]21.3 [18.8-24.3]
Growth modulation index (GMI), median a Range (min-max) ≤1.1, n (%) >1.1 - <1.33, n (%) ≥1.33, n (%)0.7 (0.1-16.3) 15 (65.2) 1 (4.3) 7 (30.4)0.9 (0.0-15.0) 12 (50.0) 2 (8.3) 10 (41.7)0.7 (0.0-16.7) 35 (62.5) - 21 (37.5)2.3 (0.1-15.4) 11 (42.3) - 15 (57.7)0.8 (0.0-42.5) 110 (56.1) 10 (5.1) 76 (38.8)

The GMI (TTP trabectedin/TTP prior chemo) was assessed on 196 patient: France, n = 23; Germany, n = 24; Italy, n = 56; UK, n = 26. NR, not reached.

Conclusions

In real-life setting T confers meaningful benefits to patients with multiple STS histotypes with a manageable safety profile regardless of small country variations.

Clinical trial identification

Y-IMAGE; ET-D-020-12

Legal entity responsible for the study

PharmaMar

Funding

PharmaMar

Disclosure

C. Benson: Honoraria from PharmaMar for speaking and travel grants. B. Kasper: Honoraria from PharmaMar. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1501P - Safety and efficacy of pazopanib (PAZ) in advanced soft tissue carcinoma (aSTS) by prior lines of therapy, age, and dose modifications: PALETTE subgroup analyses (ID 5064)

Presentation Number
1501P
Lecture Time
13:15 - 13:15
Speakers
  • A. Le Cesne
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

PALETTE was a randomized phase 3 trial (NCT00753688) that demonstrated single-agent activity of PAZ in advanced STS (aSTS). We evaluated the relationship between age, prior lines of therapy, and dose modifications on the safety and efficacy of PAZ in aSTS.

Methods

Median progression-free survival (mPFS) was evaluated in subgroups of prior lines of therapy (1 prior line; 2+ prior lines), age (<65 y; ≥65 y), and dose reductions and interruptions (no dose reduction/interruption; ≥1 dose reduction/interruption). Adverse events (AEs) were also compared in subgroups of prior lines of therapy and age. All analyses were descriptive and exploratory and require cautious interpretation.

Results

A total of 246 patients received pazopanib in the PALETTE study. Median PFS and median overall survival (OS) were longer in patients receiving PAZ who had only 1 prior line of therapy vs 2+ prior lines of therapy (mPFS, 24.7 vs 18.9 weeks [Table]; OS, 13.7 vs 11.3 months). In patients receiving PAZ, mPFS was similar in ages <65 and ≥65 y (20.0 and 20.1 weeks, respectively). In patients receiving PAZ, mPFS was maintained in patients requiring dose reductions or dose interruptions to manage toxicities (Table). Regardless of number of prior lines of therapy, patients receiving PAZ had similar AE rates. The AEs leading to study discontinuation in patients receiving PAZ were higher in ≥ 65 vs < 65 y group (30% vs 17%, respectively). Rates of dose reductions, dose interruptions, and serious AEs leading to study discontinuation were similar between the 2 age groups.

1501P

SubgroupsNPazopanib, mPFS, weeks (95% CI)
Lines of therapy
1 prior line11024.7 (19.6-27.4)
2+ prior lines13618.9 (11.9-20.1)
Age
<65 y (range: 18-64)18420.0 (17.9-22.0)
≥65 y (range: 65-83)6220.1 (11.7-31.6)
Dose reduction
No dose reduction15411.9 (8.9-19.3)
≥1 dose reduction9227.7 (21.1-35.7)
Dose interruption
No dose interruption10711.0 (8.1-19.3)
≥1 dose interruption13921.3 (20.1-27.7)

mPFS, median progression-free survival; CI, confidence interval

Conclusions

Longer mPFS was observed in patients receiving PAZ following only 1 line of therapy. Additionally, mPFS with PAZ was maintained regardless of patient age or if dose modification was required to manage toxicity.

Clinical trial identification

NCT00753688

Legal entity responsible for the study

Novartis Pharmaceutical Corporation

Funding

Novartis Pharmaceutical Corporation

Disclosure

A. Le Cesne: Pharmamar, Lilly, Pfizer, Novartis, Amgen. S. Bauer: Research support: Novartis, Blueprint Medicines, Ariad. Ad board: GSK, Novartis, Pfizer, Bayer, Fresenius, Lilly, Blueprint Medicines, Deciphera. CME-Honoraria: Pharmamar, GSK, Bayer, Novartis. Travel support: Pharmamar, Bayer. G.D.S. Demetri: Grants and personal fees from Novartis, during the conduct of the study; grants and personal fees from Janssen, Eisai, Eli Lilly and PharmaMar, outside the submitted work. W.T.A. van der Graaf: Research grants: GSK, Novartis. Advisory board: Bayer. G. Han, L. Dezzani, Q. Ahmad: Employee of Novartis. H. Gelderblom: Grants to institution LUMC, during the conduct of the study.

Collapse
Poster display session Poster Display session

1502P - Evolution in neutrophil-to-lymphocyte ratio (NLR) among advanced soft tissue sarcoma (STS) patients treated with pazopanib within EORTC 62043/62072 trials (ID 4511)

Presentation Number
1502P
Lecture Time
13:15 - 13:15
Speakers
  • E. De Maio
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

A high NLR has been shown to be associated with a poor prognosis in several solid tumors including STS and might be helpful for patient stratification and individual risk assessment. The aims of this study were to confirm that higher NLR at baseline is associated with worse prognosis in STS and to evaluate if an early decline of NLR during treatment with pazopanib is associated with a more favorable prognosis.

Methods

The single-arm phase II EORTC 62043 and placebo-controlled phase III EORTC 62072 were both investigating the effect of pazopanib in patients with advanced STS. We evaluated NLR at baseline and 50 days later. Multivariate analyses on pazopanib-treated patients investigated the prognostic value on both Progression-Free Survival (PFS) and Overall Survival (OS) of NLR at baseline as well as the predictive value of changes in NLR from baseline to the 50-days landmark. Sensitivity analyses were conducted on the placebo-treated patients.

Results

Among the 333 eligible patients treated with pazopanib, a NLR at baseline ≥3 was associated with shorter PFS and OS in comparison to NLR <3 (HR 1.44; 95% confidence interval {CI} = 1.14-1.82; p-value 0.002 and HR 1.86; 95% CI = 1.43-2.41; p-value <0.001, respectively). Changes in NLR ratio were defined as a difference of at least 40% with baseline. Compared with no changes, an increase or decrease in NLR did not affect PFS (HR 1.19; 95% CI = 0.71-1.98 and HR 0.94; 95% CI = 0.71-1.24; p-value 0.685, N = 262 patients) or OS (HR 1.40; 95% CI = 0.90-2.16 and HR 0.96; 95% CI = 0.7 2- 1.28; p-value 0.255, N = 302 patients). Thresholds other than 40% difference to define NLR change did not impact the result and no association between changes in NLR and outcome was seen in placebo-treated patients. The median NLR change in patients treated with pazopanib was a decrease of 30.4% compared to an increase of 14.5% in placebo.

Conclusions

In this study, limited by its retrospective design, the prognostic value of NLR at baseline was confirmed in advanced STS patients, irrespective of treatment. Changes in NLR during the first 50 days of treatment with pazopanib were not associated with patient outcome and can therefore not be used as an early marker for response.

Legal entity responsible for the study

EORTC

Funding

EORTC STBSG

Disclosure

A. Le Cesne: Pfizer, Lilly, Amgen, Novartis, Pharmamar Honoraria, myself, compensated. P.G. Casali: Consultant/Advisory, Honoraria and Research funds (for the institution) from Amgen, Dompé, Bayer, Blueprint Medicines, Eisai, Eli Lilly, Daiichi Sankyo Pharma, Epizyme Inc., Merck SD, Merck Serono, Nektar Therapeutics, Novartis, Pfizer and PharmaMar. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1503P - Benefit of the use of tyrosine kinase inhibitors (TKIs) in patients (pts) with METAstatic Soft Tissue SARComa (STS) in a Real-Life Setting: an ancillary analysis of the METASARC Study (ID 3727)

Presentation Number
1503P
Lecture Time
13:15 - 13:15
Speakers
  • S. Cousin
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Treatment options for pts with advanced STS are limited. STS, like other proliferating malignancies, are dependent on the formation of new blood vessels to support their growth, invasion and metastasis. Growth Modulation Index (GMI) has been demonstrated as a relevant endpoint to assess clinical in patients with advanced STS. There are no data related to GMI in STS patients treated with anti-VEGFR targeted therapy.

Methods

Pts with metastatic STSs diagnosed between 1990 and 2013 and documented in the prospectively maintained database of the French Sarcoma Group who have received at least one TKI during their treatment were analysed. GMI, defined as the ratio of the Time To Progression (TTP2) under the TKI/TTP under the previous line of treatment (TTP1) was calculated.

Results

209 pts (102 male) were included in this study. Median age was 50 (11-83). 234 lines of TKI were administrated. The drugs used were: Pazopanib (77, 33%), Sorafenib (70, 30%), Sunitinib (45, 19%), Regorafenib (30, 13%), Others (12, 5%). The most frequent histology subtypes were: leiomyosarcoma (64, 30.6%), undifferentiated sarcoma (28, 13.4%), synovialosarcoma (20, 9.6%), desmoplastic round cell tumor (14, 6.7%) and angiosarcoma (11, 5.3%). Median of previous lines was 2 (0 – 4). Median TTP under TKI was 4.1 months (0 – 131.1). GMI was available for 201 pts. Median GMI was 0.76 (0.02 - 12.49). GMI was ≥ 1 in 87 (41.6%) pts and ≥ 1.3 in 65 (31.1%) pts. 17 pts received 2 consecutive lines of TKI. For these pts, median GMI (TTP under TKI2/TTP under TKI1) was 1 (0.12 – 8.77). Seven (41.2%) had a GMI ≥ 1.3.

Conclusions

Targeting VEGFR was associated with significant clinical benefit (GMI ≥ 1.3) in about one third of STS pts. Up to 41% of patients progressing on a TKI experienced clinical benefit with another TKI suggesting the lack of absolute cross-resistance between TKI.

Legal entity responsible for the study

French Sarcoma Group

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1504P - Response to apatinib in advanced alveolar soft part sarcoma (ID 4391)

Presentation Number
1504P
Lecture Time
13:15 - 13:15
Speakers
  • C. Tu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Alveolar soft part sarcoma (ASPS) is a rare, hypervascular and chemo-resistant soft tissue sarcoma. Apatinib, a novel tyrosine kinase inhibitor that highly selectively binds to the ATP-binding site of VEGFR-2 within cells resulting in inhibition of tumor angiogenesis, has shown a substantial potential in angiosarcoma, malignant fibrous histiocytoma and round cell liposarcoma. This study aimed to review the clinical efficacy and safety of apatinib in ASPS.

Methods

The clinical information of 6 patients with advanced ASPS who received apatinib were collected. The median age of them was 26.5 years old (17y-32y). Five patients were found with multiple lung metastases and one (case 4) was with locally advanced unresectable tumor. The maximum diameters of locally advanced tumor and metastatic nodules were measured by MRI and thin-section CT, respectively. All cases received apatinib at initial continuous daily dosing of 500 mg every 4 weeks. Clinical efficacy was evaluated according to RECIST v1.1. The adverse events (AEs) were graded according to CTCAE v4.03.

Results

Median follow-up from start of apatinib treatment was 10.2 months (range, 1-21 months). Five of 6 patients who received at least 1 complete cycle of apatinib treatment were eligible for the efficacy analysis (Table). One patient achieved RECIST complete response and stop apatinib treatment after six cycles. Four patients got partial response. No disease progression was found. The current objective response rate to apatinib treatment was 100% (5/5). The most common grade 3/4 treatment-related AEs were hand-foot syndrome (60.0%), hypertension (20.0%), and hepatotoxicity (20.0%). No drug-related severe AEs occurred. At the time of analysis, all patients were still alive and five patients continued to receive apatinib.

1504P Clinical data of six patients with alveolar soft part sarcoma who received apatinib

CaseAge/genderLocal tumor locationLocal tumor size (cm)Local tumor resection (Yes/No)ChemotherapyCyclesClinical efficacy
117/maleLeft thigh4.5YesGemcitabin + DocetaxelStop 6CR
227/femaleRight thigh10.5NoNoneOngoning 2PR
329/maleLeft thigh11.3NoIfosfamideOngoing 4PR
432/maleRight thigh16.9NoIfosfamide+ CisplatinOngoing 4PR (Local tumor volume decrease)
526/maleLeft thigh9.6YesNoneOngoing 6PR
628/FemaleRight thigh5.8NoNoneOngoing <1

Conclusions

Our analysis confirms the short-term efficacy and safety of apatinib in patients with advanced ASPS. This result supports future randomized controlled trial to further verify anti-tumor activity of apatinib in stage IV sarcomas.

Legal entity responsible for the study

Chongqi Tu

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1505P - Analysis of expression of immunomodulation factors in alveolar soft-part sarcoma: a retrospective study from the Spanish Group for Research on Sarcoma (GEIS) (ID 5140)

Presentation Number
1505P
Lecture Time
13:15 - 13:15
Speakers
  • N. Hindi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Alveolar soft-part sarcoma (ASPS) is an exceedingly rare entity. We previously reported on a series of 12 ASPS the expression of PD-1/PD-L1 and lymphocytic infiltrates. PD-L1 staining was positive in 50% of the cases, suggesting a role for immunomodulatory interventions. We now enlarge the series and explore the expression of immune-related genes with the HTG EdgeSeq System and analyze their possible prognostic role.

Methods

Patients (pts) diagnosed with ASPS from Dec 1994 to Jul 2016 were reviewed. Clinical characteristics and outcome were collected. Immunohistochemical expression was tested on archived Formalin-Fixed Paraffin-Embedded (FFPE) blocks using PD-L1 (ab205921; Abcam), CD8 (ab4055; Abcam) antibodies. An ImmunOncology (IO) panel of 549 mRNAs was evaluated with the HTG EdgeSeq System from one 5µm FFPE section. This novel technology consists in a tissue digestion followed by a pre-hybridization with specific probes using quantitative nuclease protection assay (qNPA) and quantified by a standard RNA-seq protocol in a NGS sequencer.

Results

We identified 16 ASPS pts: median age 23y (6-62), M/F=5/11. Stage localized/metastatic in 11/5. With a median FU of 91 mos (4-151), 6/11 pts (54%) relapsed, with a median RFS of 19 mos (95% CI 1-75) and 2 pts died. PD-L1 was pos in tumor in 10/16 (62%) pts. HTG showed differential expression of immune-related genes according to stage (localized vs metastatic) in MNDA (log2 fold change: -3.01, p < 0.00001) and PRAME (log2 fold change: 1.9, p < 0.0002). Pts relapsing differentially expressed ABCC2 (log2 fold change -1,64, p < 0.04) when compared with those not relapsing. PD-L1 pos vs neg tumors (pos>5% membrane staining for PD-L1) differentially expressed TNFSF11 (fold change: -1,77, p = 0.05). Tumors infiltrated/not for CD8 lymphs (pos>10% of CD8) had a differential expression of CCL18 (log2 fold change: 4,89, p < 0.0001) and SLAMF7 (log2 fold change 2,37, p < 0.0089).

Conclusions

PD-L1 is expressed in more than a half of ASPS of our series. Differential expression in immune-associated genes suggest an immune related gene profile on this entity with clinical and pathological implications. Further exploration of immune-modulation in ASPS is ongoing.

Legal entity responsible for the study

Spanish Group for Research on Sarcoma

Funding

Spanish Group for Research on Sarcoma (GEIS).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1506P - PD-L1 inhibition – a new therapeutic opportunity in cutaneous angiosarcoma? (ID 5244)

Presentation Number
1506P
Lecture Time
13:15 - 13:15
Speakers
  • R. Hamacher
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Cutaneous angiosarcomas (cAS) represent a particularly challenging sarcoma subtype that frequently occurs in pretreated patients as a consequence of radiation therapy or chronic lymphedema. Local treatments are complicated as affected sites are often compromised by previous surgery and irradiation and tumor lesions lack a clear delineation. Novel treatments are therefore urgently needed to improve the outcome of these patients.

Methods

We identified 130 angiosarcomas from our institutional database of which 37 originated in the skin. Clinical characteristics and outcome was analyzed and available tumor tissue was tested for expression of PD-L1 and infiltration of immune cells, including CD8+ cells.

Results

Response to classical chemotherapeutic drugs was highest for taxanes, but frequent responses were also seen with liposomal doxorubicin, gemcitabine or oral cyclophosphamide. Despite promising chemotherapy responses, most patients eventually died of disease with often dismal local complications. Using IHC we observed several cases of cAS that showed a significant expression of PD-L1 between 10-20% on tumor cells. In addition, infiltration of immune cells with the presence of CD8+ cells was regularly observed. One of the patients with PD-L1-positive angiosarcoma of the scalp who had progressed after several lines of chemotherapy was treated with pembrolizumab. Following the second application the patient already showed a dramatic improvement of symptoms with complete healing of a large area of ulcerating and bleeding skin and ongoing remission after 6 months of treatment.

Conclusions

Based on this study, PD-L1-directed therapy may represent a particular opportunity for angiosarcomas of the skin given the frequency of PD-L1 expression and infiltration of immune cells. Our findings suggest prospective immunotherapy studies in this sarcoma subtype.

Legal entity responsible for the study

Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany

Funding

None

Disclosure

M. Schuler: Funding, honoraria and reimbursements from Alexion, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Lilly, MSD, Novartis, AstraZeneca, Roche. H-U. Schildhaus: Honoraria and reimbursements from MSD, BMS, Roche, Abbott Molecular and ZytoVision. S. Bauer: Funding, honoraria and reimbursements from Novartis, Pfizer, Bayer, PharmaMar, GlaxoSmithKline, Blueprint Medicines, Lilly, Deciphera Pharmaceuticals. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1507P - Pembrolizumab (PEM) in patients with advanced/metastatic bone sarcoma (BS) or soft tissue sarcoma (STS): Named patient use by the Medical University of Vienna (ID 3911)

Presentation Number
1507P
Lecture Time
13:15 - 13:15
Speakers
  • S. Schur
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Treatment options in locally advanced/metastatic BS and STS are limited. PEM has shown first signs of promising activity in some histologic subtypes. In this named patient use BS and STS patients who either failed standard therapy or where no standard therapy was established were treated with PEM.

Methods

This retrospective analysis includes efficacy/safety data from 18 pts. with advanced/metastatic BS/STS treated with PEM 200mg d1, q21d between May 2016 and April 2017.

Results

10 pts. were female (56%), 8 pts. male (44%). Median age was 45 yrs. (range 18-84 yrs.). Extent of disease at initial diagnosis was localized in 15 pts. (83%) and advanced/metastatic in 3 pts. (17%). The median number of previous lines of systemic treatment before PEM was 3 (range 0-7 lines). In total, 71 cycles of PEM were administered (median 3 cycles per pt., range 1-11 cycles). Immune-related side effects were hypothyroidism in two pts. and uveitis in one pt. PD-L1 assessment on tumor samples is ongoing.

1507P

patient IDhistologyn PEM cyclesstatusstatus PEM
1fibromyxoid sarcoma4*1
2OSA1100 NED
3fibrosarcoma3*1
4myxofibrosarcoma800 PR
5myxofibrosarcoma2*1
6angiosarcoma301
7dedifferentiated LPS700 PR
8EMC301
9EMC301
10chondrosarcoma500 SD
11myxoid LPS300 PR
12angiosarcoma1*1
13dedifferentiated LPS500 SD
14synovialsarcoma300 SD
15epitheloid sarcoma300 PR
16OSA200 ie
17OSA300 ie
18myxoid LPS200 ie

Abbreviation: OSA = osteosarcoma, EMC = extraskeletal myxoid chondrosarcoma, LPS = liposarcoma, status 0 = alive, * = dead; status PEM 0 = PEM ongoing, 1 = PEM discontinued to PD (progressive disease); NED = no evidence of disease, PR = partial remission, SD = stable disease, ie = response in evaluation.

Conclusions

In this unselected cohort, PEM seems to have some activity in advanced/metastatic BS/STS. However, longer follow up of treated patients and prospective clinical trials of PEM in BS/STS patients will define the value of PEM in this patient cohort. Updated efficacy and toxicity data as well as PD-L1 expression levels will be presented at the meeting.

Legal entity responsible for the study

Thomas Brodowicz, Clinical Division of Oncology, Medical University of Vienna

Funding

None

Disclosure

S. Schur: Personal fees from Eli Lilly (advisory board) outside the submitted work. T. Brodowicz: Personal fees from Roche and PharmaMar for lecture fees and from Amgen, Bayer, Novartis, Eisai and Eli Lilly for lecture fees and advisory boards. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1509P - Characteristics and prognosis of gastrointestinal stromal tumor in the pre-imatinib era: An analysis based on the Kinki GIST registry in Japan (ID 2178)

Presentation Number
1509P
Lecture Time
13:15 - 13:15
Speakers
  • T. Hashimoto
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Recent breakthroughs regarding the oncogenesis of gastrointestinal stromal tumor (GIST) have led to the wider use of imatinib (IM). In addition, since perioperative IM has been established, more accurate information regarding the clinical behavior of GIST is mandatory. However, there is no big data about the clinicopathological characteristics and prognosis of GIST in Japan. The aim of this study was to clarify them based on an analysis of the GIST registry conducted by the Kinki GIST Study Group in Japan.

Methods

The registry was designed to collect data on background characteristics, treatment methods, pathologic characteristics, and prognosis of primary GIST from 2003 through 2007 at 40 participating institutions.

Results

The study enrolled 346 male patients and 332 female patients. The median [range] age was 66 [18-93] years. The primary sites were stomach (74%), small intestine (19%), rectum (3%), esophagus (1%), colon (1%), and others (1%). Fifty-eight percent were asymptomatic and 42% were symptomatic e.g. bleeding (17%), pain (10%), and digestive symptoms (9%). None of the patients was received perioperative IM therapy. Pathological examination revealed that the tumor size was 4.0 [0.1-35]cm and the mitotic count was 3 [0-300] per 50 high-powered fields. There were 91.0% KIT positive GISTs and 82.9% CD34 positive GISTs. Ninety-seven (14.5%) patients showed recurrence and the common recurrent sites were liver (n = 58) and peritoneum (n = 33). According to the modified-Fletcher criteria, the recurrence rates were 0% (0/93, very low-risk group), 2.6% (6/230, low-risk,), 4.6% (4/87, intermediate-risk), and 38.9% (75/193, high-risk), respectively. The 5-years overall survival rate was 89.0%. The 5-years recurrent free survival rate (RFS) of gastric GISTs was significantly better than that of other sites’ GISTs (5-years RFS:82.7% vs. 63.9%, P < 0.001).

Conclusions

We reported the clinicopathological characteristics of GIST in multi-center registry study in Japan. Currently applied GIST risk classification system is comparable to predict high- or low-risk patients with primary non-metastatic and completely resected GIST in pre-IM era.

Legal entity responsible for the study

Kinki GIST registry

Funding

Kinki GIST registry

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1510P - Early response evaluation by 18F-FDG-PET influences management in gastrointestinal stromal tumor patients treated with neo-adjuvant intent (ID 1413)

Presentation Number
1510P
Lecture Time
13:15 - 13:15
Speakers
  • S. Farag
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Early response evaluation by 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) is effective in gastrointestinal stromal tumors (GISTs) treated with imatinib and recommended in GISTs treated with neo-adjuvant intent. Yet, it is unclear whether this effects treatment decisions.

Methods

All patients in the Dutch GIST Registry treated with imatinib with neo-adjuvant intent were identified. Only FDG-PETs made within 8 weeks after initiation or change (in dose or switch) of imatinib were included. Responses were derived from radiological reports and defined in 3 categories: 1) complete response; 2) partial response; 3) no response. Change in management was defined as a difference between pre-PET and post-PET treatment plans. Four categories were defined: change in 1) surgical management; 2) systemic treatment; 3) treatment objective (from curative to palliative); 4) management regarding a second tumor.

Results

Seventy FDG-PETs for early response evaluation in 63 patients treated with neo-adjuvant intent were identified. Forty-one patients (65.1%) had a KIT exon 11 and 22 (34.9%) had a non-KIT exon 11 mutation (15 other and 7 unknown mutations). Of the 70 scans 64 (87.1%) had a baseline, 50 (71.5%) showed metabolic response (partial and complete), and 18 (25.7%) led to change in management. Change in management was strongly correlated with a lack of response (p < 0.001) and a non-KIT exon 11 mutation (p < 0.001). Mutational status and response were strongly correlated (p < 0.001). Out of 29 FDG-PETs conducted in non-KIT exon 11 GISTs, 15 (51.7%) led to change in management: 1 (3.4%) in surgical management, 6 (20.7%) in systemic treatment, 7 (24.1%) in both and 1 (3.4%) regarding a second tumor. Out of 51 FDG-PETs conducted in KIT exon 11 GISTs, change in management was seen 3 times (5.9%): twice in systemic treatment (dose increase after partial response was seen) and once regarding a second tumor. No change in treatment objective was seen.

Conclusions

In contrast to GIST patients harboring a KIT exon 11 mutation, in non-KIT exon 11 mutated GISTs treated with neoadjuvant intent early response evaluation by FDG-PET often leads to change in management.

Legal entity responsible for the study

Neeltje Steeghs

Funding

Novartis, Pfizer and Bayer

Disclosure

N. Steeghs: Research grant for the Dutch GIST Registry from Novartis, Pfizer and Bayer. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1511P - Surgery in the treatment of metastatic and recurrent Gastrointestinal Stromal Tumors (ID 4579)

Presentation Number
1511P
Lecture Time
13:15 - 13:15
Speakers
  • M. Gluzman
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Gastrointestinal stromal tumors (GISTs) are the most frequent mesenchymal tumors of the gastrointestinal tract. Surgery is the method of choice in treatment of localized GISTs but it also plays a great role in the treatment of advanced forms. The main goal of our study is to define the role of surgery for locally advanced and metastatic/recurrent lesions.

Methods

We have performed a retrospective analysis from a prospectively documented database. All histologically proven GISTs, diagnosed and treated between 2003 and 2016, were enrolled from 4 clinics in Saint-Petersburg, Russia. Cases of recurrent or metastatic GISTs were selected from the registry, and baseline characteristics and survival outcomes were analyzed. Patients were classified into two groups. The surgical treatment group (ST group) included those who underwent surgical treatment in addition to tyrosine kinase inhibitor (TKI) therapy after recurrence or metastasis, whereas the drug treatment group (DT group) included those who were treated only with TKI therapy.

Results

Metastasis or recurrence developed in 34 (22,8%) of the 149 patients with GISTs who had undergone surgery for primary localized or locally advanced tumors, 13 (38,2%) of whom were assigned to the ST group and 21 (61,8%) to the DT group. Median follow-up was 68 (4-162) months. In the ST group the 3-year overall survival rate (OS) was significantly higher than in DT group (92,3 vs. 61,9%, p < 0,05) but these groups didn't differ with respect to the 5-year overall survival rate (61,5 vs. 52,4%, p > 0,05). Median time to progression (TTP) was 23,9 months in the DT group and 29,7 months in the ST group (p > 0,05). OS was correlated with the pattern of recurrence: local-regional recurrence vs. distant metastasis – 3-year OS (94,4 vs. 50%, p < 0,05), 5-year OS (77,8 vs. 31,25%, p < 0,05). Median TTP didn't differ in both groups (18,5 vs. 20 months, p > 0,05).

Conclusions

Continuous TKI therapy appears to be important primarily for the prognostic improvement of patients with recurrent/metastatic GISTs. Surgical resection may have benefits when combined with TKI therapy for patients with stable disease or disease responsive to TKI therapy especially in the cases of local-regional recurrence.

Legal entity responsible for the study

Orlova Rashida

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1512P - Clinicopathologic features and long-term follow-up of metastatic gastrointestinal stromal tumor (GIST) patients (pts) with durable response (≥ 5 years) to frontline imatinib (IM): A case-control study from GEIS (ID 1239)

Presentation Number
1512P
Lecture Time
13:15 - 13:15
Speakers
  • C. Serrano
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

IM achieves disease control in most metastatic GIST pts, typically about 18 months. Interestingly, ∼15% GIST pts remain on IM beyond 5 years of continuous treatment. To date, clinicopathologic features predictive of long-term benefit to IM remain largely unknown.

Methods

We analyzed clinical, pathological and molecular characteristics, and long-term outcomes in metastatic GIST pts treated with continuous daily dosing of frontline IM in a cohort of pts benefiting ≥ 5 years, compared with a control group obtained from a national GEIS database.

Results

We found 41 IM long-term responders (IM-LTR) and 71 control cases (CC) with a median time on IM of 90.6 and 18.2 months, respectively. Compared to CC, IM-LTR were younger (59 vs 64 years, p = 0.040), fitter at diagnosis (performance status (PS) 0-1: 100% vs 82.2%, p < 0.001), had fewer metastasis prior to IM initiation (2.6 vs 7.5, p < 0.001), and primary tumors were bigger (10.4 vs 9 cm, p = 0.005) but had lower mitotic count (5.5 vs 15 per 50HPF, p = 0.005). There were no differences in terms of primary tumor location, disease status or metastases location at diagnosis. Mutational profile from IM-LTR (KIT ex 11, 81%; KIT ex 9, 0%; PDGFRA, 8%; wild-type, 11%) did not differ significantly from CC. IM-LTR had significantly better response pattern (complete response 34.1%; partial response 43.9%; stable disease 22%) and overall survival (not reached) over CC (9.2%, 40%, 26.2%; and 63 months, respectively). Only 7 pts (18%) receiving IM ≥ 5 years withdrew IM due to progression (69% CC, p < 0.001). Eight pts (25%) developed ≥ 1 new toxicities after ≥ 5 years on continuous IM; only 1 pt withdrew IM due to toxicity (grade 3 anemia). Univariate analyses show that pts with better PS (p = 0.002), low mitotic count (p = 0.025), low number of metastases (p < 0.001), and better response to IM (p < 0.001) achieve durable benefit from frontline IM.

Conclusions

Clinical and inherently biological tumor characteristics define a subset of GIST pts with increased likelihood to achieve durable benefit from IM. Molecular studies are needed to better identify these pts at treatment initiation.

Legal entity responsible for the study

Spanish Group of Sarcoma Research (GEIS)

Funding

Spanish Group of Sarcoma Research (GEIS)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1513P - Tumor growth rate analysis of progression-free survival (PFS) and overall survival (OS) for patients with metastatic and/or unresectable gastrointestinal stromal tumors (GIST) receiving placebo or regorafenib in the phase 3 GRID trial (ID 2541)

Presentation Number
1513P
Lecture Time
13:15 - 13:15
Speakers
  • C. Kappeler
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In the randomized, controlled phase 3 GRID trial (NCT01271712), regorafenib (REG) significantly improved PFS versus placebo (PBO) in patients with advanced GIST (HR 0.268; P < 0.0001). Here we report an analysis exploring prognostic characteristics of early tumor growth rate (eTGR) for PFS and OS.

Methods

The primary endpoint of GRID was PFS; OS was a secondary endpoint. Target lesions were assessed by central radiologic review based on RECIST (v1.1). Changes in target lesions over time were approximated by a parabola-like 3-parametric model. eTGR was defined as the percentage change per month of the sum of target lesion diameters from the start of double-blind treatment. To explore the association between eTGR and PFS and OS, values of eTGR were split into quartiles (Q) separately by treatment arm. PFS (cut-off in 2012) and OS (cut-off in 2015) were compared in each subgroup population by median times derived from Kaplan–Meier curves and from modeling with a Weibull distribution.

Results

For PBO and REG, there is a nearly inverse relationship between eTGR and median times of PFS and OS from Q1 to Q4 eTGR. For the REG subgroup in Q1 eTGR, this trend is lost, as it has similar or worse median times than the subgroups around zero eTGR, which show the best prognosis.

1513P

PBO
REG
Q1Q2Q3Q4Q1Q2Q3Q4
Mean eTGR–0.0630.0640.1470.279–0.128–0.0220.0240.135
Median PFS*2.71.51.00.85.57.86.01.6
Median OS*45.521.812.39.729.128.917.411.5

By Weibull model of Kaplan–Meier curves; durations in months.

Conclusions

In this exploratory analysis, stabilization of tumor lesions at treatment start seems to be prognostic for better PFS and OS outcomes. A strong reduction of eTGR, which is seen for some REG patients, is not necessary for further disease stabilization and improvement. eTGR may be an additional efficacy parameter to consider when monitoring REG and other tyrosine kinase inhibitor (TKI) treatments in TKI-resistant GIST.

Clinical trial identification

NCT01271712

Legal entity responsible for the study

Bayer

Funding

Bayer

Disclosure

C. Kappeler, A. Wagner: Employment and Stock Ownership: Bayer AG.

\r\n

G.D.S. Demetri: Consulting/Advisory Role: Bayer, Pfizer, Novartis, Eli Lilly, EMD Serono, Sanofi, Janssen Oncology, PharmaMar, Daiichi Sankyo, ARIAD, Blueprint Medicines, Kolltan Pharmaceuticals, WIRB-Copermicus Group, ZIOPHARM Oncology, Polaris, Nektar, Genocea Biosciences, G1 Therapeutics, Caris Life Sciences, Adaptimmune, Kyocera, Eisai; Stock Ownership: Blueprint Medicines, G1 Therapeutics, Bessor Pharma, Caris Life Sciences, Champions Oncology, N-of-One; Research Funding: Novartis, Pfizer, Bayer, AbbVie, Janssen Oncology, Eisai, Amgen; Leadership: Blueprint Medicines; Other: Patents licensed to Novartis from Dana-Farber, with royalty paid to Dana-Farber.

Disclosure

C. Kappeler, A. Wagner: Employment and Stock Ownership: Bayer AG.

G.D.S. Demetri: Consulting/Advisory Role: Bayer, Pfizer, Novartis, Eli Lilly, EMD Serono, Sanofi, Janssen Oncology, PharmaMar, Daiichi Sankyo, ARIAD, Blueprint Medicines, Kolltan Pharmaceuticals, WIRB-Copermicus Group, ZIOPHARM Oncology, Polaris, Nektar, Genocea Biosciences, G1 Therapeutics, Caris Life Sciences, Adaptimmune, Kyocera, Eisai; Stock Ownership: Blueprint Medicines, G1 Therapeutics, Bessor Pharma, Caris Life Sciences, Champions Oncology, N-of-One; Research Funding: Novartis, Pfizer, Bayer, AbbVie, Janssen Oncology, Eisai, Amgen; Leadership: Blueprint Medicines; Other: Patents licensed to Novartis from Dana-Farber, with royalty paid to Dana-Farber.

Collapse
Poster display session Poster Display session

1514P - Serum miRNA abundances discriminate imatinib-naive patients with advanced gastrointestinal stromal tumors (GIST) from those in remission on Imatinib therapy (ID 4087)

Presentation Number
1514P
Lecture Time
13:15 - 13:15
Speakers
  • H. Kosela Paterczyk
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Deregulation of microRNAs (miRNAs) expression is observed virtually in all major types of neoplasm and miRNAs level in blood circulation are investigated as a potential diagnostics or prognostics biomarkers for neoplastic disorders. Gastrointestinal stromal tumors (GISTs) is the most common sarcoma of the gastrointestinal tract and to date performed studies on GISTs have provided mounting evidence on altered miRNA association with clinical, pathological features and Imatinib resistance in GIST. However, the utility of circulating miRNA as response markers of GIST progression and for Imatinib treatment have not been evaluated

Methods

36 metastatic or unresectable CD-117-positive GIST patients, were enrolled and serum sample was collected prior to Imatinib treatment. All patients responded initially to imatinib therapy. In 12 patients an additional serum sample was collected following targeted treatment at the time of remission. Control group comprised 30 healthy individuals. MiRNAs were isolated from serum with MirVANA miRNA Isolation Kit and then analyzed using deep sequencing on Ion Torrent PGM. Reads were mapped to miRBase miRNA collection with miRDeep2. Differential expression was evaluated with edgeR.

Results

Deep sequencing identified 1284 miRNAs. The pair-wise comparison between Imatinib treated and Imatinib-naive GIST samples uncovered 22 miRNAs with differential expression (adjusted p value <0.05) of which 10 (miR-142-5p, let-7d-3p, miR-30e-5p, miR-194-5p, miR-223-5p, miR-223-3p, miR-125a-5p, miR-199b-5p, miR-24-2-5p, miR-641) yielded AUCs (areas under Receiver Operating Characteristic curves) ranging 0.81 and 0.9, thus having a high discriminative properties. A comparison of imatinib-naive GIST and control healthy samples revealed 99 differentially expressed miRNAs (adjusted p value <0.05) of which four (miR-582-5p, miR-150-5p, miR-450b-5p, miR-450a-5p) reached AUCs with high discriminatory power ranging 0.81-0.84.

Conclusions

Circulating miRNA abundances can distinguish GIST patients from those in remission following Imatinib therapy as well as from the healthy controls. However, further studies evaluating the potential of designated microRNAs as response markers for treatment or as predictive markers of GIST are warranted.

Legal entity responsible for the study

Piotr Rutkowski

Funding

the grant from National Science Center [2013/11/B/NZ5/03165 to PR

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1515P - LncRNA H19, HOTAIR and MALAT1 as prognostic molecular biomarkers in GIST (ID 4892)

Presentation Number
1515P
Lecture Time
13:15 - 13:15
Speakers
  • N. Barraco
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Long non-coding RNAs (lncRNAs) are emerging as essential regulators of genetic and epigenetic networks. Their deregulation may underlie carcinogenesis suggesting their potential involvement in tumorigenic and metastatic processes, as well as their role as prognostic/predictive biomarkers for clinical use in patients with several solid tumors. Few studies evaluated lncRNAs expression in rare tumors such as Gastrointestinal Stromal Tumors (GISTs). Indeed, the upregulation of HOTAIR has been associated with tumor aggressiveness and metastasis, and poor survival of GIST patients. In order to gain more detailed insight on the molecular role of lncRNAs, we analyzed the expression levels of lncRNAs H19, MALAT1 and HOTAIR in tissue specimens of surgically resected GIST patients to evaluate the potential role of lncRNAs as prognostic biomarkers.

Methods

The expression of the lncRNAs H19, MALAT1 and HOTAIR was evaluated in a total of 40 pairs of disease formalin-fixed paraffin-embedded tissue and adjacent normal tissue from 40 GIST patients with localized and locally advanced disease using quantitative real-time reverse transcriptase.

Results

H19 was overexpressed in 50% GIST patients (p-value: 0.0496). MALAT1 was overexpressed in 45% GIST patients (p-value: 0.032). None of them had the related date with HOTAIR. Furthermore, the up-regulation of H19 has been found in 74% patients harboring cKIT mutations compared to 57% wild type patients (p-value: 0.042). Conversely the up-regulation of MALAT1 has been found in 76% patients harboring cKIT mutations compared to 100% wild type patients (p-value: 0.027). Finally, the up- regulation of H19 has been found in 100% patients with TTP < 3 months compared to 25% patients with TTP >3 months, while the up-regulation of MALAT1 has been found in 25% patients with TTP < 3 months compared to 75% patients with TTP >3 months.

Conclusions

H19 and MALAT1 appear upregulated in GIST patients according to the KIT- mutation status. These data would suggest a potential opposite prognostic value of both H19 and MALAT1 lncRNAs in these patients. The results of HOTAIR expression levels were indeterminated in all analyzed tumor samples, probably because HOTAIR has been degraded during its isolation. Further analyses are needed to confirm these data.

Clinical trial identification

Not applicable

Legal entity responsible for the study

University of Palermo

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1516P - Predictive factors of response to Sunitinib in metastatic Gastrointestinal Stromal Tumors (mGISTs): A retrospective analysis (ID 4815)

Presentation Number
1516P
Lecture Time
13:15 - 13:15
Speakers
  • L. Incorvaia
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Imatinib is currently the standard therapy for first line treatment of metastatic GIST. Although this treatment has demonstrated durable responses with PFS and OS benefit, most patients develop resistance and experience subsequent disease progression. Current treatment available in second line are Imatinib high-dose (800 mg/day) or Sunitinib. The presence of two options in this setting, in the absence of direct comparisons, raises many questions on the choice.

Methods

A total of 128 patients with metastatic GIST were collected in our analysis in this large database. We analyzed the validity of several parameters as possible predictors of response to treatment with Imatinib high-dose vs Sunitinib in patients progressing at the standard dose of Imatinib 400 mg/day. The parameters analyzed were: anatomic site of primary GIST, site of metastasis, KIT and PDGFRa mutational status, and FDG-PET status at progressing disease. Every factor has been correlated with Progression Free Survival (PFS) for Imatinib 800 mg/day and Sunitinib treatment, measured in months. Datas collected have been analyzed with software “Medcalc”, performed by using the Kaplan-Meier method.

Results

Univariate analysis showed Sunitinib more active then Imatinib in gastric GISTs (median PFS: Sun 12 months vs Ima 800 6 months; p < 0,0001), in pts with peritoneal metastasis (median PFS: Sun 10 months vs Ima 800 5 months; P = 0,2429), in wild-type (median PFS: Sun 20 months vs Ima 800 17 months; P = 0,1361) and PET-negative GIST patients (median PFS: Sun10 months vs Ima 800 7 months; p = 0,0874).

Conclusions

with the limitations of a retrospective analysis, this study identifies the gastric site of primary tumor as a predictive factor to efficacy of Sunitinib treatment in second line. The mutational status (GIST WT), the site of metastasis (peritoneum) and the FDG-PET status (negative), although not statistically significantly, seem to be elements of increased activity for Sunitinib treatment in second line.

Clinical trial identification

NA

Legal entity responsible for the study

University of Palermo

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1517P - Analysis of PD-L1 Expression in Patients with Gastrointestinal Stromal Tumors (ID 4996)

Presentation Number
1517P
Lecture Time
13:15 - 13:15
Speakers
  • A. Kinupe Abrahao
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The immune system is believed to have an important role in solid tumor progression. The development of monoclonal antibodies targeting immune checkpoints, such as programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1), have revolutionized the treatment of some cancers. Recent efforts have attempted to elucidate the relevance of the PD-1/PD-L1 pathway in gastrointestinal stromal tumors (GIST).

Methods

Formalin-fixed, paraffin-embedded specimens were obtained from resected GIST at Sunnybrook Health Sciences Centre between March 2008 and August 2015. PD-L1 analysis was based off a tissue microarray of the cases using the Roche Ventana SP263 antibody. Each case had 1mm cores taken from different areas of the tumor block. Normal controls used for PD-L1 were placenta and tonsil (epithelial and inflammatory). CD117 was assessed via immunohistochemistry in all tumor specimens.

Results

Of twenty-nine patients who underwent surgical resection, eight had insufficient tumor tissue for analysis, and three cases were excluded due to CD117 negativity after preoperative imatinib treatment, leaving 18 patients for analysis. Three of these 18 cases were positive for PD-L1 expression: 2 patients with moderate PD-L1 staining in 85% of the stromal cells and 1 with weak staining in 15% of the stromal cells. Fifteen patients were negative for PD-L1 expression. Analysis of PD-L1 expression in tumor-infiltrating lymphocytes was not feasible due to the lack of inflammatory cells in tumor environment. The patients whose samples had significant PD-L1 expression had gastric primaries, with tumour size <10cm. They did not require preoperative treatment, and did not have metastatic disease despite two having a high mitotic rate. The clinicopathologic characteristics of patients by PD-L1 expression status is demonstrated in Table 1.

1517P

PD-L1 negative (N = 15)PD-L1 positive (N = 3)
Age (± SD)62 (14)63 (14)
Gender M F10 51 2
Location Gastric Small Bowel Colon Rectum7 5 - 33 - - -
Metastasis No Yes11 43 -
Preoperative Imatinib No Yes7 83 -
Size (cm) Preoperative imatinib Upfront surgery6.2 (1.3-16) 5.8(1.5-11)− 9.5 (1.5-9.5)
Mitotic Rate (50 HPF) < 5 = or > 512 31 2

Conclusions

In this cohort, PD-L1 may be a favorable prognostic biomarker. Further studies to examine the clinical benefit of immunotherapy in GIST patients with or without PD-L1 expression are warranted.

Legal entity responsible for the study

Ana Beatriz Kinupe Abrahao

Funding

Glenita Mungcal GIST Research Award from the Life Raft Group Canada

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1518P - Gastrointestinal stromal tumors secrete Ghrelin and express ghrelin receptors – needs linguistic correction (ID 675)

Presentation Number
1518P
Lecture Time
13:15 - 13:15
Speakers
  • C. Zhu
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Gastrointestinal stromal tumor (GIST) mainly occurs in the fundus ventriculi. Ghrelin is a multifunctional protein polypeptide, which can promote the occurrence and development of tumor. It is mainly produced in the fundus ventriculi. But the misdiagnosis rate and postoperative recurrence rate is high. This study was to determine the serum ghrelin level in the normal population, patients with GIST before and after operation. And The expression of ghrelin and its receptor was determined in GIST tissue and The expression of ghrelin in adjacent normal tissue in the same patient. We hope that ghrelin can provide a new idea for the diagnosis and prognosis of GIST.

Methods

Preoperative and postoperative serum of 78 patients with GIST and 69 normal persons was collected and determined the level of serum ghrelin by ELISA. Expression of ghrelin and ghrelin receptors in GIST tissue (47 cases) was determined and Expression of ghrelin in adjacent normal tissue was determined at same time by immunohistochemical. We observed whether there were statistically significant differences in statistical analysis.

Results

the average ghrelin in the serum of normal people was 20.14 pg/ml, and 31.25 pg/ml for GIST patients. P = 0.034. The mean value of serum ghrelin in 78 patients with GIST was 31.25 pg/ml before operation and 22.5 pg/ml after operation . P = 0.023. Ghrelin and its receptor expression positive rate was 100% in GIST (47 cases). Among them, Ghrelin: (+) 12%, ( ++) 29%, ( +++) 38%, ( ++ ++) 20% Ghrelin receptors:(+) 4%, ( ++) 23%, ( +++) 27%, ( ++ ++) 46% 4) The expression of ghrelin in tumor tissue was about 2.23 times than that of adjacent normal tissues.

Conclusions

Gastrointestinal stromal tumors secrete Ghrelin and express ghrelin receptors. Ghrelin may serve as an effective indicator for the diagnosis and prognosis of GIST.

Clinical trial identification

This experiment has been examined and approved by the ethics committee.there is no trial protocol number.the release date is oct 2015.

Legal entity responsible for the study

Peking Union Medical College Hospital

Funding

Beijing Municipal Commission of Health and Family Planning

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1519P - Primary pleomorphic sarcoma (PS) and leiomyosarcoma (LMS) of bone: Retrospective analysis of an original series (ID 4127)

Presentation Number
1519P
Lecture Time
13:15 - 13:15
Speakers
  • G. Baldi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

To describe the clinico-pathological features of 23 patients affected by primary PS and LMS of bone, to confirm the diagnosis by molecular analysis, to evaluate the clinical outcome and to explore the prognostic impact of these features on disease-free (DFS) and overall survival (OS).

Methods

Primary PS and LMS of bone surgically treated from 2004 to 2015 were retrospectively reviewed. We analysed: age, sex, stage, histotype, histological-grade and surgical and/or medical therapy. IDH1 mutational status was evaluated and immunohistochemical staining was performed for smooth muscle actin and desmin. For molecular analysis tumor DNA was extracted from freshly cut FFPE sections by GeneRead™ DNA FFPE (Quiagen) and ddPCR (Bio-rad) was used to determine the presence of IDH1H and IDH1C mutations. DFS and OS rates were calculated according to the Kaplan-Meier method. The differentiation (myogenic, MD, versus non-myogenic, NMD) was correlated with the outcome using Kaplan-Meier method.

Results

23 patients with primary PS or LMS of bone were included in the study. Median age was 49 years (range 13-90), male/female 14/9, 18 had localised disease and 5 metastatic disease, 17 received surgery, 14 received adjuvant therapy, 1 received neoadjuvant chemotherapy and 5 received up-front chemotherapy for advanced disease. All cases were histologically and radiologically reviewed: 17 PS and 6 LMS were identified. All cases were high-grade (FNCLCC grading system). Mutational analysis is currently underway and it will be presented at the meeting. 5-year OS of the whole series was 60% (95% CI; 3,1 – NE) and 5-year DFS was 50% (95% CI; 1,6 – 12,2). Patients with advanced disease are 13: 5-year OS in this subgroup was 38% (95% CI; 2,5 – NE). We identified MD in 11 cases. There were no significant differences between the MD and NMD groups in terms of DFS (logrank p-value=0,6788) and OS (logrank p-value=0,7389).

Conclusions

These primary malignant bone tumours are very rare with poor prognosis after relapse or when radical surgery is not feasible. MD did not predict a worse outcome than NMD in terms of OS and DFS.

Legal entity responsible for the study

Giacomo Giulio Baldi

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1520P - Development of a new promising rescue agent for high dose methotrexate (HDMTX) treatment in osteosarcoma: A safety and dose finding study (ID 3598)

Presentation Number
1520P
Lecture Time
13:15 - 13:15
Speakers
  • M. Eriksson
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

HDMTX followed by calcium folinate (CF) rescue is established as part of MAP chemotherapy to manage toxicity during osteosarcoma treatment. A problem with HDTMX is the variability in plasma exposure of both MTX and CF leading to an unpredictable response. A potentially superior rescue agent methylenetetrahydrofolate (Modufolin®), containing the active metabolite of CF, has been evaluated to identify a safe and effective dose for further development.

Methods

This exploratory study performed in Hungary, Poland, Sweden and Czechia involved osteosarcoma patients, 12-40 years, planned for MAP chemotherapy. All patients received one MAP cycle (two HDMTX courses) with standard CF rescue of 15 mg/m2. Those that completed this MAP cycle successfully according to six defined criteria subsequently received Modufolin® in the following cycle (two HDMTX courses). There were two Modufolin® dose cohorts, 15 mg/m2 (1) and 7.5 mg/m2 (2). A Data and Safety Monitoring Board evaluated safety before initiation of the second dose cohort and suggested the dose for further development.

Results

Eight patients 12-17 years were included. Four patients were treated in cohort 1 and four in cohort 2. In cohort 1, no MTX toxicity or delayed elimination with subsequent treatment delay was reported. In cohort 2 one patient reported mucositis grade 3 and failed successful advancement after first course of Modufolin®. In both cohorts and after both types of rescue, there were cases with significantly increased s-creatinine levels.

Conclusions

Modufolin® seems to be a safe and effective rescue agent after HDMTX. The study design however precludes a comparison between Modufolin® and CF, since only patients with successful CF rescue received Modufolin®. The higher dose of 15 mg/m2 seemed more effective as rescue and was selected for further development.

Clinical trial identification

EudraCT Nr 2013-001280-23

Legal entity responsible for the study

Isofol Medical AB

Funding

Isofol Medical AB

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1521P - Role of intraarterial cisplatin and intravenous adriamycin as neoadjuvant and adjuvant chemotherapy in non-metastatic osteosarcoma (ID 3696)

Presentation Number
1521P
Lecture Time
13:15 - 13:15
Speakers
  • A. Mishra
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Primary bone sarcomas are rare tumors, comprising approximately 0.2% of all cancers. Because of distant metastasis, cure after surgical treatment alone is uncommon. The development of effective adjuvant or induction chemotherapy regimens has dramatically improved the prognosis and cure rate of 50%–70% and limb salvage in > 90% of cases. For nonmetastatic osteosarcoma(NMO), optimal treatment consists of multiagent neoadjuvant/adjuvant chemotherapy and limb-sparing surgical procedures. The degree of tumor necrosis (TN) after neoadjuvant chemotherapy is one of the most important pronostic indicators. Intraarterial cisplatin and intravenous adriamycin (IC-IA) could achieve a good tumor response.

Methods

Based on achievement of a maximized angiographic response, 106 patients with NMO of the extremities received IC-IA monthly for 3-6 courses between January 1995 and December 2008 followed by limb salvage surgery then adjuvant intravenous (IV) chemotherapy with adriamycin and cisplatin. After resection, if patients had a good response (the extent of TN was ≥ 90%), the same regimen was administered IV every 3 weeks for a total of 6 courses of chemotherapy. Poor responders (tumor necrosis < 90%) were treated with a regimen of high-dose methotrexate with leucovorin rescue (HD-MTX) or ifosfamide, cisplatin, and etoposide (ICE).

Results

Patients received an average of 5 cycles of neoadjuvant IC-IV chemotherapy. 96 patients underwent limb-preservation surgery and 72 had > 90% TN. With an average follow-up of 8years, 60 patients were continuously disease free, 32 died of disease and 14 had no evidence of disease within 5 years after relapse. The 5-year overall survival rate was 70%. No patient developed clinically detectable cardiac toxicity or ototoxicity after adriamycin and cisplatin administration. Febrile neutropenia occurred in few.

Conclusions

This study shows the effectiveness of treating primary NMO of extremities with IC-IA infusion. Advanced bone and soft tissue sarcomas are challenging diseases to treat with an unmet need for effective systemic therapy. Checkpoint inhibitors and Adoptive T cell therapy based immunotherapy could be a new ray of hope in treating bone sarcomas.

Clinical trial identification

Trial Protocol Number IND18745J

Legal entity responsible for the study

Gandhi Medical College and Hamidia Hospital, Bhopal, India

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1522P - Biomarkers of malignant cell growth used to assess the risk of osteosarcoma development (ID 617)

Presentation Number
1522P
Lecture Time
13:15 - 13:15
Speakers
  • B. Sultonov
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The aim of our study was a comprehensive study of molecular-genetic, pathologic and clinical symptoms in patients with osteosarcoma to enhance the effectiveness of early diagnosis, treatment and prevention of osteosarcomas.

Methods

We studied the immunophenotypic characteristics of tumor cells in 212 patients with osteosarcoma. Results of antibody reactions to Ki-67, bcl-2, p53 (mutant gene) localized in the nuclei and the mitochondrial matrix expressed in % based on the number of stained cells per 100 examined.

Results

Our results revealed that the expression profile of p53 +, bcl-2-, Ki 67+ in 34.9% (74/212) patients with osteosarcoma is considered as poor prognosis, presented as early metastasis, progression of tumor growth and early relapses (15 months), advanced processes (III and IV stage), low grade pathomorposis (1 and 2), the relative duration of lifespan in patients (up to 3 years), it is associated with a low degree of differentiation (G3), an increase in tumor size up to 550 cm3 with chondroblastic type of osteosarcoma. These data should be considered when looking for and isolating the most promising groups for molecular genetic markers that are would be predictive valuable in the clinic for monitoring the treatment of patients with osteosarcoma.

Conclusions

Thus, it is obvious need for clinical medicine in the implementation and expansion of molecular testing for effective decision-making on the appointment of molecular targeted therapy of patients with osteosarcoma. These requires optimization the approach based on consideration of the specifics of the population, the objective conditions related to the presence of intratumoral characteristics. The data reveals a group of patients with osteosarcoma at high risk with unfavorable prognosis at the stage of examination and choose for these kind of patients effective therapy.

Legal entity responsible for the study

National Cancer Research Center of Uzbekistan

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1523P - Efficacy of second line treatment with etoposide and ifosfamide in adult patients with advanced Ewing Sarcoma family tumors (ID 5294)

Presentation Number
1523P
Lecture Time
13:15 - 13:15
Speakers
  • M. Nardo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Ewing Sarcoma family tumors (ES) are rare subtypes of sarcomas, and even less common in adult patients. For those not amenable to treatment with curative intent, sequential therapy with multi-agent combinations is the standard of care, usually followed by ifosfamide/etoposide (IE) at the time of progression, largely based on protocols that included pediatric patients. Nevertheless, less is known about the efficacy of this approach for adult patients with ES refractory to first-line therapy.

Methods

We assembled a retrospective cohort of patients aged 18 or older diagnosed with metastatic/inoperable ES refractory to first-line combinations, treated with IE between 2010 and 2016. Patient´s characteristics, tumor variables, treatment outcomes and toxicity data were evaluated. Kaplan-Meier method was used to estimate overall survival and uni/multivariate analysis were carried out to identify factors associated with survival.

Results

Among 18 adult patients, the mean age of diagnosis was 22 years, 73% were male and 84% had an ECOG of 0-1 at commencement of IE. Pelvis and thorax were the most common primary sites. The mean number of cycles of IE was 4. The disease control rate was 27%, with partial responses occurring in 16% of the patients (there were no complete responses). The median OS was 4,8 months (IC 95% 0,7-8,8). Toxicities ³ grade 3 occurred in 61% of the patient, including two treatment-related deaths. The main grade 4 toxicity was febrile neutropenia. Hospitalization were required in 55% of the cases.

Conclusions

IE has limited efficacy and significant toxicity when used in the second-line setting for adult patients with advanced ES, and different approaches should be investigated for these patients.

Legal entity responsible for the study

Instituto do Câncer do Estado de São Paulo

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1524P - AG-120, a novel IDH1 targeted molecule, inhibits invasion and migration of chondrosarcoma cells in vitro (ID 5111)

Presentation Number
1524P
Lecture Time
13:15 - 13:15
Speakers
  • V. HEREDIA
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Chondrosarcoma (CS) is the second most frequent primary malignant bone tumor in adults. Surgery is the best treatment option for these patients since most subtypes are resistant to chemotherapy and radiotherapy, thus novel systemic therapies are needed for patients with unresectable tumors. The majority of cases correspond to the conventional central CS histology, were recurrent mutations in isocitrate dehydrogenases (IDH1/2) coding genes are found. Therefore, IDH1 has been reported as a potential therapeutic target and several selective inhibitor molecules, such as AGI-5198 and, more recently, AG-120, have been developed and are currently being evaluated in clinical trials. In this work, we have explored the in vitro effects of AG-120 on a central CS cell line, JJ012, which carries a mutation in IDH1.

Methods

JJ012 cells were cultured both in monolayer and three-dimensional (3D) spheroids and treated with increasing concentrations of AG-120. IDH1 mutation in arginine residue R132G was verified by PCR sequencing. Proliferation and cytotoxic screening were done with Sulforhodamine B (SRB) assay. Monolayer invasion and migration assays were performed with FluoroBlok and wound healing assays respectively and 3D experiments were developed using a Matrigel matrix.

Results

R132G mutation of IDH1 was confirmed by PCR sequencing. Previous reports with AGI-5198 inhibitor show contradictory results regarding their effect on CS cells. In this work, we show how novel molecule AG-120 inhibits both invasion and migration of CS IDH1 mutated JJ012 cell line in monolayer and 3D cell culture, although it does not affect their proliferation. Minor effects on viability were only detected at high dose (100 µM).

Conclusions

These results support AG-120 as a new possible therapeutic agent for patients with metastatic CS, and further research is needed to understand its action mechanisms in this pathology.

Legal entity responsible for the study

Fundación para la Investigación Biomédica del Hospital Universitario La Paz (FIBHULP)

Funding

Fundación para la Investigación Biomédica del Hospital Universitario La Paz (FIBHULP)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1525TiP - Geriatric assessment of elderly chemotherapy-naïve patients treated with trabectedin for advanced soft tissue sarcomas (STS): The E-TRAB study of the German Interdisciplinary Sarcoma Group (GISG-13) (ID 1521)

Presentation Number
1525TiP
Lecture Time
13:15 - 13:15
Speakers
  • B. Kasper
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

More than half of soft tissue sarcoma (STS) patients are diagnosed older than 60 years and prognosis for advanced disease is unfavourable with a median overall survival (OS) of ∼12 months remaining substantially unchanged during the last 20 years. Quality of Life (QoL), especially when collected directly from the patients as Patient Reported Outcome (PRO) measures, has gained increasing interest in cancer care; however, existing data among elderly STS patients are scarce. The rationale of the E-TRAB study is to analyze QoL and PRO data in the elderly population (aged ≥ 60 years) with advanced STS, considered to be unsuited to receive anthracycline-based chemotherapy, and treated with trabectedin as a 1st line therapy.

Trial design

The E-TRAB (GISG-13) study is designed as a non-interventional trial (n = 110). Primary endpoints to be evaluated are OS and QoL. It is hypothesized that OS will not be < 10 months in this population. As secondary endpoints a comprehensive geriatric assessment is performed, and whenever feasible captured as PRO, in order to predict for safety parameters consisting of the Instrumental Activities of Daily Living (IADL), the Mini Nutritional Assessment (MNA), the Charlson Comorbidity Index (CCI), the Geriatric Depression Scale (GDS) and Time up & Go. The predictive value of two geriatric screening tools (G8, CARG prediction tool) is investigated with regards to unplanned hospitalizations, grade 4 toxicities and early death within the first six months. The EORTC QLQ-C30 and 18 PRO-CTCAE items are used to evaluate PRO. As translational research the tumour microenvironment is analyzed immunohistochemically using a new multicolor-immunofluorescence-histology technology (OPAL). This trial benefits from a cooperation of sarcoma centers in Austria, Germany and Switzerland under the German Interdisciplinary Sarcoma Group (GISG). The recruitment of patients is ongoing.

Clinical trial identification

NCT03022448

Legal entity responsible for the study

GWT-TUD GmbH

Funding

This investigator-initiated (IIT) study was sponsored by GWT-TUD GmbH, Dresden, Germany, with support from PharmaMar, S.A., Madrid, Spain.

Disclosure

B. Kasper: Honoraria and research funding by PharmaMar. P. Reichardt, M. Schuler, P. Hohenberger: Honoraria and research funding from PharmaMar.

Collapse
Poster display session Poster Display session

1526TiP - CDK4/6 inhibition in locally advanced/metastatic chordoma (NCT PMO-1601) (ID 4549)

Presentation Number
1526TiP
Lecture Time
13:15 - 13:15
Speakers
  • S. Fröhling
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Chordoma is a rare bone tumor with slow growth. The standard treatment is en-bloc excision, but the site of origin of the disease (skull base, sacrum) often prevents complete resection. For these patients, debulking surgery followed by radiation therapy (RT) or high-dose RT alone can be an alternative. However, local relapses and, more rarely, metastatic disease occur, and there is no efficient systemic therapy available. Only very limited responses are seen with chemotherapy or targeted agents (e.g. imatinib, lapatinib). In chordoma cell lines and patient biopsies, the p16 (cyclin-dependent kinase inhibitor 2a, encoded by CDKN2A) tumor suppressor is consistently deleted. Experiments with patient-derived chordoma cell lines demonstrated aberrant CDK4/6 activity downstream of p16 loss, which can be efficiently inhibited by the CDK4/6 inhibitor palbociclib, resulting in reduced proliferation and growth of neoplastic cells (von Witzleben et al. Cancer Res 2015;75(18):3823-31).

Trial design

Patients ≥18 years of age with locally advanced or metastatic chordoma with at least one measurable tumor lesion, ECOG Performance Status 0-2, adequate organ function, and loss of p16 (as determined by immunohistochemistry) or CDKN2A (as determined by genetic analysis) who have no response or have lost response to treatment with a tyrosine kinase inhibitor are eligible. Key exclusion criteria are prior treatment with palbociclib, uncontrolled CNS involvement, cytopenia/s, significant cardiovascular disease including prolongation of the corrected QTc interval >470 ms. Based on previous experience with 125 mg palbociclib once daily for 21 days followed by 7 days of rest in patients with other solid-organ malignancies, this regimen is chosen. Based on a Simon optimal 2-stage design, the disease control rate is the primary endpoint, whereby response is defined as complete response, partial response, or stable disease according to RECIST v1.1 after 6 cycles. For sample size calculation, we estimate a poor response with 10% and a good response with 25% (power, 80%; alpha, 5%), resulting in a first stage of 18 patients and, if 3 or more patients respond, a second stage with 25 additional patients (total, n = 43).

Clinical trial identification

NCT03110744 EudraCT Number: 2016-004660-19

Legal entity responsible for the study

University Hospital Heidelberg

Funding

NCT Heidelberg, Pfizer

Disclosure

R. Schlenk: Research funding: Novartis, Pfizer, Amgen, AtraZeneca, PharmaMar Speakers bureau: Novartis, Pfizer Advisory board: Daiichi Sankyo, Novartis, Pfizer. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

TRANSLATIONAL RESEARCH (ID 5679)

Lecture Time
13:15 - 13:15
Speakers
  • R. Giavazzi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15
Poster display session Poster Display session

1634P - FGFR pathway genomic aberrations and response to FGFRs inhibitors (ID 1711)

Presentation Number
1634P
Lecture Time
13:15 - 13:15
Speakers
  • M. Faure
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Fibroblast growth factor receptors (FGFRs) are broadly distributed transmembrane tyrosine kinase receptors. They promote cell development, differentiation, survival, migration, angiogenesis, and carcinogenesis. Specific FGFR aberrations was observed in certain cancers, some are known to be driver for tumor progression, and related to prognosis or sensitivity to cancer treatments. It is consistent to hypothesize that targeting these cancers with FGFR inhibitors would be therapeutically beneficial.

Methods

We lead a retrospective, descriptive, monocentric study, involving results of three phase I trials of Institut Gustave Roussy, for patients with refractory various solid tumors, previous selected on FGFR abnormality (amplification, single nucleotid variant (SNV), translocation). Primary endpoint was tumor response. Secondary endpoints were difference in tumor according to the type of mutation, and time to failure (TTF).

Results

55 patients with median age of 55 years and various solid tumors were enrolled between February 16, 2011 and October 26, 2016. All patients had advanced or metastatic cancer with a median of 3.5 metastatic sites. They were heavily pretreated, in median 3 prior regimens of chemotherapy, 0 to 2 lines of targeted therapy, and one patient had immunotherapy. For patients with FGFR fusion proteins, 43% achieved partial response (PR). Median tumor response was - 46%, sustained response until 17 months. Response of SNV group depended on the type of mutation and degree of pathogenicity, in the different tumor locations. 20% had PR. Whereas some pathogenic mutations lead to dramatic response until 18 months; others failed to achieve tumor shrinkage or stabilization. 5% of patient with FGFR amplification had tumor response for 3 months only. Main clinical and biological toxicities were grade ½ and resolved after interruption. Treatment was resumed, at same or lower dose, excepted for 3 patients with permanently discontinuation. There was no death due to toxicity.

Conclusions

We identified genetic alterations in various members of the FGF/FGFR system that represent suitable predictive biomarkers to guide patient selection for treatment with selective pan FGFRs targeting agents.

Legal entity responsible for the study

Institut Gustave Roussy

Funding

None

Disclosure

A. Hollebecque, R. Bahleda, J-C. Soria: Amgen, Astellas, Astra Zeneca, Bayer, Cellgene, Genentech, Ipsen, Jansen, Lilly, Novartis, Pfizer, Roche, Sanofi Aventis All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1635P - FGFR 360° resistance: Establishing a translational research framework in FGFR-altered (FGFRalt) patients (pt) treated with fibroblast growth factor receptor inhibitors (FGFRinh) (ID 5154)

Presentation Number
1635P
Lecture Time
13:15 - 13:15
Speakers
  • C. Hierro
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Pt selection is critical for the future development of FGFRinh. Our institution aimed to implement a translational platform to obtain samples of FGFRalt pt included in phase 1 trials.

Methods

Prospective generation of a collection of pt samples with molecularly-selected FGFRalt tumors [amplified(amp)/mRNA high expression(mRNAh)/mutated(mut)/translocated(trans)]. We developed a protocol to obtain serial biopsies (bx) during therapy with an FGFRinh, including warm autopsies, for patient-derived xenografts (PDXs) generation. We collected plasma for analysis of circulating tumor DNA (ctDNA). Clinical benefit (ClinBen) was defined as any tumor shrinkage or disease control for 4 months.

Results

From 2014 to 2017, 40 FGFRalt pt were included [FGFRamp (20)/mRNAh (7)/mut(17)/trans (3)]. 30 cases received an FGFRinh [multi-tyrosin kinase (7), selective reversible-(8) or irreversible-FGFR1-4inh (14) or FGFR4inh (1)]. 8 cases achieved ClinBen (5 breast - 2 FGFR1amp, 2 FGFR1mut, 1 11q+FGFR2amp-/1 biliary tract FGFR2trans/1 head&neck FGFR1mRNAh/1 mullerian carcinosarcoma FGFR2mut). PDXs/bx after progression to FGFRinh (10) and warm autopsies of responding pt (2) will serve to study tumor heterogeneity and resistance mechanisms using novel high-throughput technologies. All PDXs (16 growing/14 in observation) will help in identifying potential predictive biomarkers and further characterizing the mechanism of action of FGFRinh in vivo. In vitro functional profiling of oncogenic activity of FGFRmut (17) will be performed. Blood samples will serve for developing in-house cfDNA analysis to monitor genomic evolution of these 40 pt.

Conclusions

We have succesfully developed a powerful precision medicine framework for linking the molecular biology with the best tumor models in parallel with early clinical research. By integrating the knowledge obtained from the analysis of relevant samples, we aim to validate future hypothesis-driven therapies for selected FGFRalt pt and guide the successful development of FGFRinh. Co-funded by ISCIII-FEDER (PI15/00360).

Legal entity responsible for the study

Vall d'Hebron Institute of Oncology

Funding

ISCIII-FEDER

Disclosure

C. Hierro: Research fundings from Bayer A. Vivancos: Member of advisory boards for AztraZeneca, Sysmex, Merck. J. Tabernero: Member of advisory boards for Amgen, Bayer, Boehringer, Celgene, Chugai, Lilly, MSD, Merck Serono, Novartis, Pfizer, Roche, Sanofi, Symphogen, Taiho and Takeda V. Serra: Research fundings from Bayer HealthCare J. Rodon: Member of advisory boards for Novartis, Lilly, Orion and has received research fundings from Principia and Bayer All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1636P - EPHA2 receptor is involved in in vivo acquired resistance to anti-Epidermal Growth Factor Receptor (EGFR) treatment in metastatic colorectal cancer (mCRC) (ID 3891)

Presentation Number
1636P
Lecture Time
13:15 - 13:15
Speakers
  • G. Martini
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

EPHA2 tyrosine kinase receptor is implicated in cell growth, migration, and invasiveness in a wide range of cancers. We studied its role as a potential marker of resistance to anti-EGFR drugs in colorectal cancer (CRC). We previously demonstrated that EPHA2 was differently activated among a panel of CRC cell lines with primary and acquired resistance to cetuximab and the use of ALW-II-41-27 (EPHA2 selective inhibitor) in combination with cetuximab was able to revert this resistance in in vitro experiments (abstract presented at 2016 ESMO Congress in Copenhagen). Here we present the study on in vivo models.

Methods

EGFR dependent SW48 and LIM1215 cell lines were engrafted into nude mice and treated with cetuximab until disease progression. Once tumors became resistant (SW48-CR and LIM1215-CR) mice were randomized in groups of 10 mice each and assigned to receive ALW- II-41-27 as single agent or in combination with cetuximab, no treatment and cetuximab alone group served as control. ALW- II-41-27 was administered daily at 30 mg/kg by oral gavage and cetuximab intraperitoneally at 1 mg/kg two days a week. Treatment was performed for three weeks, then mice were euthanized and protein expression in tumors was analysed by Western Blot.

Results

The combination of the two drugs induced a significant reduction of tumor volume since the first administration. A reduction of 50% of tumor volume was found in 5 out 10 LIM1215-CR mice treated with ALW-II-41-27 as single agent. This effect was maintained after cessation of therapy and induced prolonged survival. Tumor protein analysis by WB demonstrated a strong reduction of EPHA2 expression and activation in mice treated with the combination of ALW-II-41-27 and cetuximab, accompanied by a significantly inhibition of activated pMAPK and pAKT.

Conclusions

These results highlight the role of EPHA2 as a potential therapeutic target in mCRC treatment.

Legal entity responsible for the study

Università della Campania Luigi Vanvitelli

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1637P - Eph A2 expression is a predictive biomarker of poorer activity and efficacy of FOLFIRI + cetuximab in RAS WT metastatic colorectal cancer (mCRC) patients (pts) in the CAPRI GOIM trial (ID 3956)

Presentation Number
1637P
Lecture Time
13:15 - 13:15
Speakers
  • C. Cardone
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Eph A2 promotes tumor growth, invasiveness and angiogenesis in mCRC. Targeting Eph A2 could overcome resistance to anti-epidermal growth factor receptor treatment in colon cancer preclinical models.

Methods

Formalin-fixed paraffin-embedded tumor specimens from 82 RAS wild type (WT) mCRC pts treated with cetuximab + FOLFIRI as first line therapy in the CAPRI GOIM trial were assessed for Eph A2 expression by immunohistochemistry. Eph A2 levels were evaluated developing an HSCORE [1 × (% cells 1+) + 2 × (% cells 2+) + 3 × (% cells 3+)] (range: 0-300). A cut off was set by ROC analysis to define high (>50) and low (≤50) Eph A2 levels.

Results

Eph A2 expression was found in 55/82 (67%) cases. According to HSCORE Eph A2 levels were low in 54 (66%) and high in 28 (34%) samples. Eph A2 expression resulted in mostly complete membranous staining. Tumor stroma was positive in 15/82 (18%) cases. In most of these cases an intense immune infiltrate was observed. Non-tumor adjacent normal mucosa was assessable in 34/82 samples. Eph A2 was expressed in 16/34 (47%), more frequently in dysplastic epithelial areas. A significant correlation between Eph A2 expression in tumor and stroma was found (p < 0.0001). Eph A2 was more frequently expressed in less differentiated tumors (p = 0.02), as well as in left-sided tumors compared to right-sided tumors [17/28 (61%), 11/28 (39%), respectively p = 0.04]. Eph A2 expression was associated with higher rate of disease progression (PD) 8/28 (29%) vs 5/54 (9%) (p = 0.02), and with worse median PFS [8.6 m (CI95% 6.4-10.8) vs 12.3 m (CI95% 10.4-14.2) p = 0.030], both in left and right-sided tumours. Moreover, median OS was 28.4 m (CI95% 13.1-43.7) vs 39.8 m (CI95% 30.2-49.4), although this result did not reach statistical significance (p = 0.23).

Conclusions

Eph A2 levels were significantly associated with a worse PFS and an increase in PD in RAS WT mCRC pts treated with cetuximab + FOLFIRI as first line therapy in the CAPRI GOIM trial, in both right- and left-sided tumors. A similar trend was observed for OS. Eph A2 might represent an additional predictive biomarker of lack of efficacy in RAS WT mCRC pts treated with cetuximab + FOLFIRI.

Legal entity responsible for the study

Department of Clinical and Experimental Medicine ‘F. Magrassi’ Università degli studi della Campania “Luigi Vanvitelli”, Naples, Italy.

Funding

AIRC

Disclosure

F. Ciardiello: Advisory boards: Merck Serono, Lilly, Roche, Bayer, Amgen, Pfizer. E. Martinelli: Advisory boards: Merck Serono, Amgen. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1638P - Clinical value of cfDNA and CTCs in EGFR mutations detected in advanced NSCLC (ID 5257)

Presentation Number
1638P
Lecture Time
13:15 - 13:15
Speakers
  • S. Calabuig Fariñas
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Targeted inhibition of EGFR represents a milestone in lung cancer treatment. Development of sensitive and accurate techniques allows the detection of EGFR mutations in liquid biopsies. CTCs and ctDNA analysis may be useful in treatment selection, response monitoring, and early resistance detection. The aim of this study was to correlate the EGFR mutational status of CTCs and cfDNA at diagnosis and during follow-up in non-small-cell lung cancer (NSCLC) patients.

Methods

The study included 22 EGFR mutated NSCLC patients, blood samples were collected and repeated sampling was performed during follow-up and at progression. cfDNA was obtained from plasma; whereas CTCs were isolated by size using a filtration-based device (ScreenCell), characterized and enumerated by H&E. CTC and cfDNA genotyping was performed by PNA-Taqman assay for EGFR 19del, L858R, G719X and T790M detection.

Results

Patient’s median age was 65 years, 81.8% were female, 70% never-smokers and 94% were ADC. The follow-up ranged from 3 to 48 months. Out of the 22 EGFR mutated tumors identified, 12 harbored exon 19 deletion, 7 L858R mutation in exon 21, 2 G719X mutation and one presented exon 19 deletion and T790M together at diagnosed. All patients were treated with EGFR-TKIs. 110 blood samples were evaluated at baseline and during follow-up. CTCs were observed by H&E with a range 1-30/3 ml. Our results confirm that detected mutations can provide early outcome information. Early undetectable blood mutations after EGFR-TKI might predict a large clinical response, whereas in TKI-responders patients, EGFR mutation remained undetectable, its reappearance preceded disease progression. In case of persisted mutation during treatment, a rapid progression and exitus was observed. A baseline T790M mutation in EGFR TKI-naïve patient has been reported with rapidly progression and exitus.

Conclusions

Results suggest that analyses of EGFR mutations in CTC and cfDNA have important clinical implications and can be a useful biomarker of diagnoses, response to therapy and early detection of mechanisms of TKIs resistance, in advance of clinically detection. This work was supported by Astra Zeneca (ISSRES0110), the RD12/0036/0025 ISCIII, grants from the FEDER and López-Trigo Grant.

Legal entity responsible for the study

Fundación para la Investigación del Hospital General Universitario de Valencia

Funding

This work was supported by Astra Zeneca (ISSRES0110), the RD12/0036/0025 ISCIII, grants from the Fondo Europeo de Desarrollo Regional (FEDER) and López-Trigo Grant.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1639P - Tucatinib, a HER2 selective kinase inhibitor, is active in patient derived xenograft (PDX) models of HER2-amplified colorectal, esophageal and gastric cancers (ID 5051)

Presentation Number
1639P
Lecture Time
13:15 - 13:15
Speakers
  • S. Peterson
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Tucatinib, an orally bioavailable and HER2 selective small molecule tyrosine kinase inhibitor, is currently being developed for HER2+ metastatic breast cancer in combination with capecitabine + trastuzumab (HER2CLIMB study). In addition to breast cancer, HER2 is amplified in subsets of patients with other malignancies, including gastrointestinal cancers (colorectal, esophageal and gastric cancers). To test whether tucatinib might have utility in treating HER2-amplified cancers originating from the gastrointestinal tract, tucatinib was tested alone, and in combination with trastuzumab, in cell line derived and PDX models of colorectal, esophageal and gastric cancer.

Methods

In vitro assays were performed to evaluate the combination of tucatinib and trastuzumab in HER2-amplified cell lines by measuring changes in signal transduction (pHER2, pHER3, pAKT) and cell survival. The in vivo activity of tucatinib (50 mg/kg BID) and trastuzumab (20 mg/kg Q3D) was evaluated alone, or in combination, in established HER2-amplified tumor models, including PDX models of colorectal, esophageal and gastric cancers.

Results

As a single agent, or in combination with trastuzumab, tucatinib demonstrated significant anti-tumor activity, including tumor regressions, in the N87 gastric cancer cell line xenograft model and in PDX models of HER2 amplified colorectal, esophageal and gastric cancers. The combination of tucatinib and trastuzumab was consistently more active than either single agent alone, and resulted in tumor growth inhibition from 85-139%, including complete tumor regressions in HER2+ gastric PDX models.

Conclusions

The activity of tucatinib in HER2-amplified colorectal, esophageal and gastric tumor xenograft models supports the exploration of using tucatinib to treat HER2+ gastrointestinal cancers in the clinical setting. To this end, an open label phase II clinical study combining tucatinib with trastuzumab in HER2+/RAS wild type metastatic colorectal cancer (MOUNTAINEER) has recently been initiated.

Legal entity responsible for the study

Cascadian Therapeutics

Funding

Cascadian Therapeutics

Disclosure

S. Peterson: Employee and shareholder of Cascadian Therapeutics, corporate officer of Cascadian Therapeutics. P. de Vries, J. Piasecki, R. Rosler: Employee and shareholder of Cascadian Therapeutics

Collapse
Poster display session Poster Display session

1640P - PI3K/RICTOR-mTORC2 axis as a driver of prognosis and potential druggable target in squamous cell lung carcinoma (SqCLC) (ID 3164)

Presentation Number
1640P
Lecture Time
13:15 - 13:15
Speakers
  • S. Pilotto
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

We built a risk classification model for resected SqCLC (R-SqCLC) by combining clinicopathological predictors to discriminate patients' (pts) prognosis (Pilotto JTO 2015), we externally validated this model in a pts’ cohort of > 1,300 R-SqCLC (Bria WCLC 2016) and we performed an integrated multi-platforms genome analysis of prognostic outliers to identify potentially druggable modulators as the PI3K/RICTOR-mTORC2 axis (Pilotto WCLC 2016). Here, we validate our molecular findings and we enhance our rationale with in vitro studies.

Methods

Next Generation Sequencing (NGS) analysis of somatic mutations (SM) and copy number alterations (SCNA) was performed (Ion AmpliSeq Lung & Colon Cancer Panel: 22 genes, a SqCLC customized targeted NGS panel: 20 genes, the commercial Ion AmpliSeq Comprehensive Cancer Panel: 409 genes). In vitro experiments were performed using the SqCLC cell line H-1703 (Rictor amplified-6 copies). PF-05212384 [PI3K/mTOR inhibitor (inh)], AZD2014 (mTORC1/2 inh), MK-2206 (panAkt inh), everolimus (mTOR inh) and chemotherapeutic drugs (Docetaxel, Gemcitabine) were tested. Cell viability was assessed by crystal violet assay and the half maximal inhibitory concentration (IC50) was estimated to evaluate drug efficacy.

Results

Main results of overall 97 pts (Training/Validation: 60/37) are presented in the Table.

1640P

GeneTraining Set [%]Validation Set [%]
SMTP5353 [88.3]27 [72.9]
TIE14 [6.7]2 [5.4]
PTEN6 [10]4 [10.8]
PIK3CA3 [5]3 [8.1]
SCNA GainsRICTOR13 [35.1]14 [23.3]
PIK3CA17 [45.9]26 [43.3]
FRS28 [21.6]7 [11.7]
FGFR114 [37.8]18 [30]
SCNA LossesPTEN5 [13.5]19 [31.7]
TSC28 [21.6]7 [11.7]

The in vitro results support a significant inhibition of H-1703 cells proliferation by Gemcitabine, Docetaxel, PF-05212384, MK-2206 and AZD2014 with IC50 values of 0.4 nM, 0.45 nM, 10 nM, 66 nM, 110 nM, respectively. Everolimus was not effective.

Conclusions

Our multi-step genomic analysis performed in almost 100 R-SqCLC pts allowed us to identify altered pathways with a biological impact in SqCLC oncogenesis, as the PI3K/RICTOR-mTORC2 axis. Moreover, our in vitro results justify pursuing mTOR inhibition, focusing on mTORC2 complex, in RICTOR-aberrant tumors.

Clinical trial identification

Not Applicable

Legal entity responsible for the study

Emilio Bria

Funding

My First AIRC (Associazione Italiana per la Ricerca sul Cancro) Grant (MFAG) project no. 14282. Young Investigational Award of the International Association for the Study of Lung Cancer (IASLC) 2015

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1641P - Combination of duligotuzumab, anti HER3 antibody or taselisib, Pi3k inhibitor with trastuzumab shows synergistic antitumoral activity in HER2 positive gastric cancer cells (GCC) (ID 3747)

Presentation Number
1641P
Lecture Time
13:15 - 13:15
Speakers
  • M. Laterza
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The anti-HER2 monoclonal antibody Trastuzumab is central to the treatment of HER2-positive gastric cancer (GC); however, its responses are limited due to some poorly understood mechanisms of resistance. The aim of this study was to asses the antitumortal activity of Duligotuzumab, an anti HER3 antibody or Taselisib, a Pi3k inhibitor combined with Trastuzumab IN a panel of HER2 positive human gastric cancer cell lines (GCC), to improve anti HER2 treatment efficacy.

Methods

We evaluated in vitro the effect of Duligotuzumab, Taselisib and Trastuzumab in single agent and in combination treatments in HER2-positive GCG (NCI-N87, KATOIII, OE19) and in negative HER2 GCC (MKN28), through proliferation, migration and apoptosis assays. We also investigated the effect of combined treatment on downstream intracellular signaling, by western blot analysis.

Results

After establishing, through a dose response curve, the IC50 for each drug used (≈ 0.5 µM), a significant synergistic effect of Duligotuzumab, Taselisib and Trastuzumab treatments in HER2-positive GCG was observed by reduction of proliferation and migration in KATOIII, OE19 and N87 cell lines; the same effect was found analyzing the apoptotic rate. At cellular level, in particular in KATOIII and OE19 cell lines, the combined treatment with Duligotuzumab or Taselisib plusTrastuzumab completely inhibited the activation of proteins downstream of HER3, PI3K and MAPK pathways.

Conclusions

Targeting both HER2 and HER3 or HER2 and PI3K with the combination of anti-HE3 antibody or Pi3k inhibitor with Trastuzumab results in an improved treatment effects on HER2-positive GCG. These important findings can be utilized to facilitate the design of future clinical trials.

Legal entity responsible for the study

University of Campania Luigi Vanvitelli

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1642P - The TLR9 agonist lefitolimod modulates tumor microenvironment and improves anti-tumor effect of checkpoint inhibitors in vivo (ID 4841)

Presentation Number
1642P
Lecture Time
13:15 - 13:15
Speakers
  • K. Kapp
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Preclinical and ongoing clinical studies support the use of TLR9 agonists for immunotherapeutic approaches. Lefitolimod (MGN1703) is a covalently-closed dumbbell-like immune surveillance reactivator with broad immunomodulatory effects on the innate and adaptive immune system. Lefitolimod is currently evaluated in a phase 3 trial in mCRC patients (IMPALA), a phase 2 trial in SCLC patients (IMPULSE) and in two phase 1/2 trials, (i) in solid tumors in combination with the checkpoint inhibitor ipilimumab and (ii) in HIV patients (TEACH).

Methods

In several murine tumor models lefitolimod reduced tumor growth. Since the mode-of-action of lefitolimod starts upstream of the initiation points of checkpoint inhibitors like anti-CTLA-4 or anti-PD-1/anti-PD-L1 combinatory approaches may result in an enhanced anti-tumor effect. Therefore, two syngeneic murine models – the colon carcinoma CT26 and the lymphoma A20 model – were used for evaluation of the anti-tumor effect of lefitolimod with checkpoint inhibitors. In the CT26 model the influence of lefitolimod on tumor microenvironment (TME) was analysed.

Results

Treatment with anti-PD-L1 (i.p.) had no effect on tumor growth in the CT26 model, whereas addition of lefitolimod (s.c.) to anti-PD-L1 led to a clear anti-tumor effect (tumor growth inhibition, TGI 48%). This combinatory effect was even more pronounced in the A20 model where treatment with anti-PD-1 (i.p.) alone had a moderate anti-tumor effect which was vastly increased by the combination (TGI – anti-PD-1: 46%, anti-PD-1/lefitolimod 99%). Moreover, in the CT26 model an anti-tumor response to lefitolimod (i.tu.) was associated with increased infiltration of CD3 T-cells –more specifically CD8 T-cells – into the tumor of a CT26 model.

Conclusions

We showed that the member of dSLIM family of TLR9 agonists, lefitolimod, can enhance the limited anti-tumor effects of checkpoint inhibitors in murine colon carcinoma and lymphoma tumor models in vivo. The anti-tumor effect of lefitolimod is associated with TME modulation with increased T-cell infiltration. These data clearly support the combination of lefitolimod with checkpoint inhibitors in clinical trials.

Legal entity responsible for the study

Mologen AG

Funding

Mologen AG

Disclosure

K. Kapp, B. Volz, D. Oswald, M. Schmidt: Employee of Mologen AG. B. Wittig: Consults Mologen AG and also receives funding from Mologen AG.

Collapse
Poster display session Poster Display session

1643P - Circulating immune-profile as predictor of outcome in advanced NSCLC patients treated with nivolumab (ID 2657)

Presentation Number
1643P
Lecture Time
13:15 - 13:15
Speakers
  • M. Tiseo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Detection of predictive markers of anti-PD-1/PD-L1 antibodies activity is of pivotal interest in non-small cell lung cancer (NSCLC). This study aimed to identify a circulating immuno-profile as predictor of outcome in NSCLC patients treated with nivolumab.

Methods

A peripheral blood immuno-profile evaluation was performed at baseline (T0), after 2 (T1) and 4 cycles (T2) of bi-weekly nivolumab in advanced pre-treated NSCLC patients from two Italian Institutions. First tumor assessment was performed after 4 cycles and then every 2 months. FACS analysis of lymphocyte subpopulations [CD3, CD4, CD8, NK (CD56), Treg (FOXP3) and MDSC] was performed. Absolute and % changes of lymphocyte subsets together with their functional and proliferative activity were assessed. Quali-quantitative leucocyte composition at baseline and its variation during therapy were correlated with tumor response and survival.

Results

In the overall population of 54 treated patients, baseline Neutrophil-to-Lymphocyte ratio and NK count, lymphocyte count and CD4 variations during therapy showed a statistically significant prognostic role (p < 0.001; p = 0.012; p < 0.001; p = 0.010, respectively). Among 31 patients (squamous carcinoma, n = 17; adenocarcinoma, n = 14) in which all 3 time-points samples were available, 19 were responders (response and stable disease) and 12 non-responders. In responders, absolute numbers of total NK and NKCD56dim subset were higher at baseline and their increase between T0 and T1 was statistically significant (p < 0.05). Responders also displayed increased cytotoxic capability as shown by a higher baseline expression of CD3ζ, perforin and granzyme in NKCD56dim subset. No significant variation was documented in absolute number and functional activity of CD4+ and CD8+ lymphocytes. A higher percentage of CD8+PD-1+ cells at baseline was observed in responders, while non-responders showed a statistically significant increase in the absolute number of MDSC during therapy (p < 0.05).

Conclusions

The number and function of NKs and the frequency of PD-1 expression in CD8+ cells could represent predictive peripheral immuno-biomarkers for nivolumab treatment in advanced NSCLC.

Clinical trial identification

NA

Legal entity responsible for the study

University Hospital of Parma

Funding

AIRC

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1644P - Monitoring the effect of cytostatic treatment by immune activity (ID 3586)

Presentation Number
1644P
Lecture Time
13:15 - 13:15
Speakers
  • T. Hansen
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Early prediction of effect remains an unsolved problem in cytostatic treatment of malignant tumors. The aim of the present study was to analyze the potential relationship between immune activity as measured by the NK Vue® system and response in patients with different tumors and different cytostatic regimens.

Methods

The study included six different trials encompassing patients with breast, prostate, ovarian and colorectal cancer. All protocols are still recruiting and so far 108 patients have been included. The preliminary results are based on 54 patients with response data available from the first evaluation. Blood samples were collected at baseline and prior to each treatment cycle into NK Vue™ Promoca tubes and placed in an incubator at 37 °C within 15 minutes of sampling. Following 24 hours of stimulation the plasma in each tube was harvested and analyzed for the level of interferon-gamma, as a surrogate for immune activity, by enzyme-linked immunosorbent assay using the NK Vue® Gold Kit.

Results

Similar results were seen between immune response and tumor types receiving different treatments. Consequently, data were pooled for preliminary evaluation. The outcome suggested a classification into three groups. The interferon-gamma dropped to an abnormal level (<200 pg/ml) in group 1 (27 patients) or remained at an abnormal level during treatment. In group 2 (12 patients) the level remained within a normal range (>500 pg/ml), while in group 3 (15 patients) it was raised from an abnormal to a normal level. The response rates were 11%, 42%, and 80% in the three groups, respectively. The difference was highly significant (p < 0.001). Accordingly, the positive and negative predictive values of a raising level were 80% and 79%.

Conclusions

The results suggest a relationship between the ability to mount an immune response upon stimulation and treatment effect comparable among different tumor types and treatments. Increasing levels of interferon-gamma shortly after initiation of treatment seems to predict treatment effect. Updated results will be presented at the meeting.

Legal entity responsible for the study

Vejle Hospital, Department of Oncology

Funding

ATGen

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1645P - Analysis of programmed death-ligand 1 (PD-L1) expression, transforming growth factor (TGF)-β gene expression signatures (GES) and tumor-infiltrating immune cells (IC) in hepatocellular carcinoma (HCC): Rationale for targeting PD-L1- and TGF-β (ID 3187)

Presentation Number
1645P
Lecture Time
13:15 - 13:15
Speakers
  • Y. Zhang
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

HCC evades antitumor immune responses via multiple mechanisms, including the PD-L1 and TGF-β pathways. PD-L1 expression correlates with tumor aggressiveness and recurrence. Increased TGF-β activity corresponds with poor clinical outcomes. Using immunohistochemistry (IHC), we previously showed that PD-L1 expression in HCC stems primarily from IC. To further assess the HCC immune milieu, we measured IC, TGF-β-associated GES, and PD-L1 expression using IHC/RNAseq.

Methods

We assessed protein expression in 50 resected HCC specimens by quantitative (Q) IHC (primary antibodies: PD-L1, CD8, CD68) using standard techniques and automated software. For RNAseq, we prepared strand-specific libraries from extracted RNA, which were sequenced and compared to GES from published papers, CIBERSORT and Ingenuity Pathway Analysis.

Results

All cases had typical morphology (low- to high-grade trabecular, pseudoglandular, or solid with common cytoplasmic features). Q CD8 IHC significantly correlated with CD8 mRNA expression and CD8 T cell GES, supporting the utility of RNAseq to evaluate the role of CD8+ T cells in HCC. RNAseq identified TGF-β1 as the main TGF-β isoform in HCC. Predefined TGF-β GES correlated strongly with EMT GES. There was a trend toward increased TGF-β1 activity and EMT marker expression in the S1 molecular subtype, which has previously been associated with TGF-β-driven aberrant Wnt signaling. Q CD8 IHC correlated with PD-L1 mRNA and protein levels in IC. In samples with high CD8, there was a trend of increased tumor-associated macrophages (TAMs); the presence of TAMs strongly correlated with TGF-β GES. Interestingly, few tumor cells displayed membranous PD-L1 staining as confirmed by PD-L1/pan-cytokeratin double labeling.

Conclusions

We used RNAseq and IHC to better understand the immunosuppressive environment in HCC driven by TGF-β and PD-L1, which may mediate different mechanisms to inhibit preexisting CD8+ IC.

Clinical trial identification

N/A

Legal entity responsible for the study

Funding was provided by Merck KGaA, Darmstadt, Germany.

Funding

Funding was provided by Merck KGaA, Darmstadt, Germany. Disclosure: Y. Zhang, B.J. Naughton, P.A. Rolfe, I. Dussault: Employee of EMD Serono Research & Development Institute, Billerica, MA, USA. E. Frick-Krieger, C. Ihling: Employee of Merck KGaA, Darmstadt, Germany. L. Terracciano: Consulting/advisory role to Merck AG

Collapse
Poster display session Poster Display session

1646P - Clinical factors associated with mutation burden in non-small cell lung cancer (ID 2174)

Presentation Number
1646P
Lecture Time
13:15 - 13:15
Speakers
  • A. Ono
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

PD-L1 expression is associated with clinical benefit from anti-PD1/anti-PD-L1 therapies in advanced NSCLC. However, additional biomarkers are needed to predict which patients will benefit most. The aim of this study is to correlate specific genomic alterations with immunological biomarkers in a cohort of NSCLC.

Methods

Patients diagnosed with NSCLC from 2000 to 2005 were retrospectively reviewed. Genetic mutations and copy number of selected genes were determined by Sanger and FISH. Immunophenotype was defined by PD-L1, HLA-1 and TILs CD8+ immunostaining and scored as follows: PD-L1 positivity ≥ 5% on membrane tumor cells; HLA-1 intensity: 0+,1+,2+; TILs CD8+ score: low or high infiltration. Statistical analysis using Chi-square test and logistic regression were performed.

Results

From 150 patients: 87% males; stage: 90% I-II, 10% III-IV; histology: 42% adenocarcinoma (ADC), 44% squamous (SCC), 14% sarcomatoid carcinoma (SaC). Genomic alterations according to histologic subtype are summarized in Table. PD-L1 was positive in 47% of tumors (49% of ADC, 43% of SCC, 58% of SaC), and correlated with TILs CD8 + (p <.001) and HLA-1 (p=.002). PD-L1 positivity was associated with MET alterations (4.5% MET amp, 2% METexon14 skipping), OR = 5.4 (1.4-21.2), p=.015. ADC harbouring STK11 loss of function were correlated with negative PD-L1 (p = .01) and associated with immunosupressive phenotype (negative PD-L1, low CD8+), OR = 11.6 (2.1-64), p=.005. Distinct PD-L1 expression was evidenced in KRAS mutant tumors according to additional co-mutations: 25% of KRAS+STK11 were PD-L1+ whereas 75% of KRAS+TP53 were PD-L1+, despite no statistically significance.

1646P Genetic alterations according to histologic subtype (N = 150).

ADCn = 59 (100%)SCCn = 62 (100%)SaCn = 19 (100%)
KRASMET ampMETex14
mut (exons 2-3)14 (20%)GA3 (5%)Mut1 (5%)
wt45 (80%)NC53 (85%)wt16 (94%)
Unknown6 (10%)Unknown2 (1)
EGFRMETex14KRAS
mut (exon18-21)6 (10)Mut0Mutation4 (21)
wt49 (83)wt58 (94)wt12 (63)
Unknown4 (7)Unknown4 (6)Unknown3 (16)
ALKPIK3CA ampSTK11
Transloc.1 (2)GA6 (10)Mutation1 (5)
wt57(96)NC20 (32)wt15 (79)
Unknown1 (2)Unknown36 (58)Unknown3 (16)
STK11FGFR1 amp
mut10 (17)Amp9 (15)
wt46 (78)NA51 (82)
Unknown3 (5)Unknown2 (3)
METex14
Mut2 (3)
wt50 (95)
Unknown7 (12)
MET amp
GA4 (7)
wt51 (86)
Unknown4 (7)
Co-mutation KRAS+STK11
Mut4 (7)
wt55 (93)
Co-mutation KRAS+P53
Mut4 (7)
wt54 (91)
Unknown1 (2)

Conclusions

MET and STK11 alterations were correlated with differential expression of tumor PD-L1. STK11 mutant tumors were more likely to have an immunosupressive phenotype. Tumors harbouring specific genomic alterations might be enriched for distinct immunophenotypes which might contribute to rational use of immunotherapies.

Legal entity responsible for the study

IDIBELL-Institut Català d'Oncologia

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1647P - Integrative analysis of NSCLC identifies tumor genetic profiles associated with PD-L1 expression. (ID 3535)

Presentation Number
1647P
Lecture Time
13:15 - 13:15
Speakers
  • M. Saigi Morgui
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

PD-L1 expression is associated with clinical benefit from anti-PD1/anti-PD-L1 therapies in advanced NSCLC. However, additional biomarkers are needed to predict which patients will benefit most. The aim of this study is to correlate specific genomic alterations with immunological biomarkers in a cohort of NSCLC.

Methods

Patients diagnosed with NSCLC from 2000 to 2005 were retrospectively reviewed. Genetic mutations and copy number of selected genes were determined by Sanger and FISH. Immunophenotype was defined by PD-L1, HLA-1 and TILs CD8+ immunostaining and scored as follows: PD-L1 positivity ≥ 5% on membrane tumor cells; HLA-1 intensity: 0+,1+,2+; TILs CD8+ score: low or high infiltration. Statistical analysis using Chi-square test and logistic regression were performed.

Results

From 150 patients: 87% males; stage: 90% I-II, 10% III-IV; histology: 42% adenocarcinoma (ADC), 44% squamous (SCC), 14% sarcomatoid carcinoma (SaC). Genomic alterations according to histologic subtype are summarized in table 1. PD-L1 was positive in 47% of tumors (49% of ADC, 43% of SCC, 58% of SaC), and correlated with TILs CD8+ (p <.001) and HLA-1 (p=.002). PD-L1 positivity was associated with MET alterations (4.5% MET amp, 2% METexon14 skipping), OR=5.4 (1.4-21.2), p=.015. ADC harbouring STK11 loss of function were correlated with negative PD-L1 (p = .01) and associated with immunosupressive phenotype (negative PD-L1, low CD8+), OR=11.6 (2.1-64), p=.005. Distinct PD-L1 expression was evidenced in KRAS mutant tumors according to additional co-mutations: 25% of KRAS+STK11 were PD-L1+ whereas 75% of KRAS+TP53 were PD-L1+, despite no statistically significance.

Table 1. Genetic alterations according to histologic subtype (N=150).
ADC n= 59 (100%) SCC n= 62 (100%) SaC n=19 (100%)
KRAS MET amp METex14
mut (exons 2-3) 14 (20%) GA 3 (5%) Mut 1 (5%)
wt 45 (80%) NC 53 (85%) wt 16 (94%)
Unknown 6 (10%) Unknown 2 (1)
EGFR METex14 KRAS
mut (exon18-21) 6 (10) Mut 0 Mutation 4 (21)
wt 49 (83) wt 58 (94) wt 12 (63)
Unknown 4 (7) Unknown 4 (6) Unknown 3 (16)
ALK PIK3CA amp STK11
Transloc. 1 (2) GA 6 (10) Mutation 1 (5)
wt 57(96) NC 20 (32) wt 15 (79)
Unknown 1 (2) Unknown 36 (58) Unknown 3 (16)
STK11 FGFR1 amp
mut 10 (17) Amp 9 (15)
wt 46 (78) NA 51 (82)
Unknown 3 (5) Unknown 2 (3)
METex14
Mut 2 (3)
wt 50 (95)
Unknown 7 (12)
MET amp
GA 4 (7)
wt 51 (86)
Unknown 4 (7)
Co-mutation KRAS+STK11
Mut 4 (7)
wt 55 (93)
Co-mutation KRAS+P53
Mut 4 (7)
wt 54 (91)
Unknown 1 (2)

Conclusions

MET and STK11 alterations were correlated with differential expression of tumor PD-L1. STK11 mutant tumors were more likely to have an immunosupressive phenotype. Tumors harbouring specific genomic alterations might be enriched for distinct immunophenotypes which might contribute to rational use of immunotherapies.

Collapse
Poster display session Poster Display session

1648P - B7-H3 (CD276) on circulating epithelial tumor cells (CETCs) correlates with proliferation marker Ki-67 and may be associated with aggressiveness of tumor in breast cancer patients (ID 4722)

Presentation Number
1648P
Lecture Time
13:15 - 13:15
Speakers
  • M. Pizon
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

CETCs in the peripheral blood are a prerequisite for the development of metastases. B7-H3 is an important immune checkpoint member of the B7 family and inhibits T-cell mediated anti-tumor immunity. It is highly overexpressed on a wide range of solid cancers and often correlates with both negative prognosis and poor clinical outcome in patients. Based on the clinical success of the inhibitory immune checkpoint blockade, mAbs against B7-H3 appear to be promising therapeutic strategy. In order to better understand the role of B7-H3 in cancer development we used a non-invasive, real-time biopsy for determining B7-H3 on CETCs in breast cancer patients.

Methods

Blood from 50 patients suffering from breast cancer were analyzed for CETCs. The number of vital CETCs and the expression of B7-H3 and Ki-67 were evaluated using the maintrac® method.

Results

CETCs were detected in all examined patients (ranged from 2-676 CETCs in 100 µl of blood). B7-H3 expression on the surface of CETCs was found in 82% of patients. Triple negative breast cancer patients had statistically significantly more B7-H3 positive CETCs than patients with hormone receptor positive tumor tissue (median 50 vs. 26.3; p < 0.05). The frequency of B7-H3 positive CETCs was significantly higher in patients who received radiation therapy compared to patients without irradiation (mean 42 vs. 29; p < 0.05). B7-H3 positive CETCs seem to be more aggressive because the percentage of B7-H3 positive CETCs correlated with the percentage of proliferation marker Ki-67 positive CETCs (r = 0.689 and p < 0.001). Interestingly, a significant relationship between Ki-67 expression level on the CETCs and nodal status was found.

Conclusions

Breast cancer patients have detectable CETCs with high frequency of B7-H3 expression regardless of stage of disease. B7-H3 seems to be an important factor in immune evasion and may be a promising target of anticancer therapies. Furthermore, radiation leads to an up-regulation of B7-H3 expression on CETCs, which could be a possible mechanism of acquired radio-resistance.

Legal entity responsible for the study

N/A

Funding

None

Disclosure

U. Pachmann, K. Pachmann: Holder of patent. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1649P - Survival of non-small cell lung cancer patients predicted from expression of PD-L1, HLA class I and MICA/B on tumor cells (ID 1898)

Presentation Number
1649P
Lecture Time
13:15 - 13:15
Speakers
  • R. Okita
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Several groups have reported that programmed death-1 (PD-1) ligand 1 (PD-L1) overexpression on tumor cells predicts a poor prognosis in patients with non-small cell lung cancer (NSCLC). Although recent studies have shown that PD-L1 overexpression on tumor cells predicts for improved clinical outcome in NSCLC patients treated with anti-PD-1/PD-L1 immunotherapy, PD-L1 low/negative tumors also benefit from anti-PD-1/PD-L1 immunotherapy. These findings suggest that study on multiple immune parameters should be considered. We recently reported that the overexpression of PD-L1 in tumor predicted a poor prognosis while overexpression of NK cell activating ligand MICA/B predicted improved clinical outcome in patients with resected NSCLC. It is well known that both T cell- and NK cell-mediated tumor recognitions are influenced by HLA class I molecules, however, the roles of HLA class I molecules are different between T cells and NK cells; HLA class I/T cell receptor immune synapse induces antigen-specific cytotoxicity by T cell, while HLA class I/killer cell immunoglobulin-like receptor synapse attenuates NK cell-mediated cytotoxicity. Here, we assessed the multiple immune parameters (PD-L1, MICA/B, and HLA class I) in NSCLC tissues to assess the prognostic factors in patients with resected NSCLC.

Methods

We examined resected tumor tissues from 91 patients with pathological stage IA-IIIA NSCLC using immunohistochemical reaction for the expression of PD-L1, MICA/B, and HLA class I then assessed whether the multiple immune parameters impact on the clinical outcome of patients with NSCLC.

Results

PD-L1low/MICA/Bhigh tumors have an excellent disease free survival time (DFS) compared with PD-L1low/MICA/Blow (p=0.010 by log-rank test) or PD-L1high (p<0.01) tumors. Additionally, MICA/Bhigh/HLA class Ihigh tumors have improved DFS compared with MICA/Blow/HLA class Ihigh tumors (p=0.035).

Conclusions

Multiple immune parameters using the expression status of MICA/B and PD-L1 or HLA class I on tumor cells are useful prognostic factors for NSCLC. We should have more concerns to NK cell-mediated tumor elimination in anti-PD-1/PD-L1 immunotherapy.

Legal entity responsible for the study

Okita Riki, Kawasaki Medical School

Funding

Japan Society for the Promotion of Science (JSPS) Kakenhi Grant (25462189 and 16K10696) (to R.O)

Disclosure

M. Nakata: Research funding from Kyowa Kirin, Taiho Pharma, and Ono Pharma. The sponsors had no control over the interpretation, writing, or publication of this work. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1650P - YKL-39 induces monocytes migration and angiogenesis and inversely correlates with metastasis in patients with breast cancer (ID 2310)

Presentation Number
1650P
Lecture Time
13:15 - 13:15
Speakers
  • I. Mitrofanova
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Human chitinase-like proteins are considered as biomarkers of cancer and chronic inflammation.YKL-39 is a unique member of chitinase-like protein family due to its presence only in humans but not in rodents both on gene and protein levels. However, its biological activity and association with tumor progression remains unknown.

Methods

YKL-39 expression and secretion in human monocytes-derived macrophages was measured by RT-PCR and ELISA. Monocyte migration was analyzed in a trans-well system. In vitro tube formation assay was performed using HUVEC cells.112 female patients with nonspecific invasive breast cancer of stage IIA-IIIC (T1-4N0-3M0) were included in the study. Confocal microscopy analysis was used to identify cell type expressing YKL-39 in tumor samples.YKL-39 expression level was measured by RT-PCR in tumor biopsy specimens.

Results

Human monocytes-derived macrophages differentiated in the presence of IL4 and TGFbeta, but not IL4 alone, were found to express high levels of YKL-39 mRNA and protein. Purified YKL-39 significantly enhanced the migration of human CD14+monocytes by 1.9 fold (p < 0.01) after 1 h, and 4.9 fold (p < 0.01) after 3 h that was comparable with the effect of MCP-1. In vitro tube formation assays using HUVEC cells demonstrated that YKL-39 has a strong pro-angiogenic effect. In human samples of breast cancer YKL-39 was found to be expressed in CD68+ macrophages but not in cancer cells or other stromal cell types. In breast cancer biopsy specimens it was found that high YKL-39 gene expression correlated with the significantly reduced frequency of lymphatic and hematogenous metastasis. Furthermore, high level of YKL-39 expression associated with 100% metastatic-free survival rate (p = 0.015).

Conclusions

TGFbeta is a key cytokine inducing production of YKL-39 in macrophages. YKL-39 stimulates critical for tumor progression processes: chemotaxis of monocytes and angiogenesis. However high levels of YKL-39 expression in tumor samples are predictive for metastatic-free survival in patients with breast cancer, suggesting that YKL-39 can program monocytes and newly growing vessels to inhibit metastatic spread.

This study was supported by grant RNF №14-15-00350.

Legal entity responsible for the study

Tomsk State University, Tomsk, Russian Federation

Funding

Tomsk State University, Tomsk, Russian Federation. This study was supported by grant RNF №14-15-00350.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1651P - Pre-treatment neutrophil lymphocyte ratio/platelet lymphocyte ratio as surrogate markers of survival in non-metastatic head and neck cancer patients: An observational study (ID 4271)

Presentation Number
1651P
Lecture Time
13:15 - 13:15
Speakers
  • V. Agarwal
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Neutrophil lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) are known to be surrogate markers of inflammation and have been shown to predict mortality in patients with heart disease and cancer. In this study, we evaluate the influence of pre-treatment NLR and PLR on overall survival in head and neck cancer patients.

Methods

In this observational correlational study, subjects with a diagnosis of non-metastatic head and neck cancer were analyzed for neutrophil-lymphocyte ratio and the platelet-lymphocyte ratio at baseline before the start of their cancer-directed therapy.

Results

In this study, 189 subjects were analyzed for neutrophil-lymphocyte ratio and platelet-lymphocyte ratio before their treatment. The mean age of the study sample was 54.5±11.8. Forty-two percent underwent surgery followed by adjuvant chemoradiation while remaining underwent concurrent chemoradiation. Neoadjuvant chemotherapy was done in 29.4% of the study population. Mean NLR was 3.4±3.13 and PLR was 12.7±8.8. ROC analysis revealed 2.23 as the cutoff for NLR and 9.49 as the cutoff for PLR. Based on these cutoffs a Kaplan-Meir analysis on overall survival showed significantly improved survival (67.5% vs 58% at their mean estimates of 52 and 36 months) in those with <2.23 NLR ratio compared to > 2.23 (Log Rank χ2=5.3, p = 0.02). Similarly, those with <9.49 PLR had better overall survival (69% vs 56% at their mean estimates of 46 and 39 months) compared to > 9.49 (Log Rank χ2=8.1, p = 0.005). Lower NLR also showed better disease-free survival (44 vs 33 months, Log Rank χ2=4.8, p = 0.03) and lower PLR also showed better disease-free survival (44 vs 33 months, Log Rank χ2=8.2, p = 0.004).

Conclusions

Both NLR and PLR are inflammatory biomarkers in cancer. The results from this study suggest pretreatment NLR and PLR as predictive markers of survival in non-metastatic head and neck cancer patients.

Legal entity responsible for the study

Vijay Agarwal

Funding

HCG Foundation

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1652P - Enhanced antitumor activity of fixed-dose combinations of celecoxib and antihypertensives (ID 5449)

Presentation Number
1652P
Lecture Time
13:15 - 13:15
Speakers
  • C. Lee
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Inflammation and hypertension have recently emerged as causal factors for tumor progression and anti-hypertensive agents have been shown to reduce inflammation and suppress tumor growth and metastasis. Cyclooxygenase-2 (COX-2) is upregulated in most human tumors and is a potent inducer of cancer-associated inflammation. This preclinical study evaluated a novel combination of a COX-2 inhibitor with three antihypertensive drugs to suppress tumor growth and metastasis.

Methods

Three anti-hypertensive drugs: i) Lisinopril [LIS], an inhibitor of Ang I converting enzyme (ACE); ii) Olmesartan medoxomil (OLM), an Ang II receptor blocker (ARB); and iii) Hydrochlorothiazide (HCTZ), a thiazide diuretic along with Celecoxib [CEL], a COX-2 inhibitor were evaluated either alone or in combination for tumor growth suppression and metastatic spread in an orthotopic inflammatory breast cancer (IBC)/SUM149 model and subcutaneous melanoma/MDA-MB-435, glioblastoma/U87, and IBC/SUM159 models. Luciferase-tagged SUM149 and MDA-MB-435 cell lines were used to determine the incidence and the burden of locoregional and systemic spread. Mice were monitored for weight loss, tumor volume and survival outcome. Metastasis was measured as luciferase expression in lymph nodes and lungs and normalized to total protein.

Results

In the orthotopic SUM149 model, OLM and CEL plus OLM, had a statistically significant decrease in tumor burden (2.4 ± 0.6 x104 RLU/mg of protein, p = 0.01 by Mann-Whitney test) in the ipsilateral lymph nodes versus the saline-treated control (17.6 ± 8.6 x104 RLU/mg of protein). Similar trend was observed for LIS, but not for HCTZ. In the subcutaneous model, synergistic antitumor activity was observed with OLM (p = 0.026) at low dose but not with LIS and CEL (p = ns). At high dose, LIS, OLM, and CEL showed significant inhibition of tumor growth but no synergy. HCTZ, an antihypertensive diuretic which has no direct impact on the vascular wall had no effect on tumor growth.

Conclusions

These preclinical data strongly suggest a hitherto unappreciated role of ACE/ARB in tumor growth control and support the further exploration of combinations of CEL with ACE/ARB in cancer, especially inflammatory breast cancer.

Legal entity responsible for the study

Marina Biotech Inc

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1653P - Overexpression of toll-like receptor 9 (TLR9) in elderly cancer patients when compared to cancer-free elderly control group (ID 5568)

Presentation Number
1653P
Lecture Time
13:15 - 13:15
Speakers
  • L. Torres
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

the genes of toll-like receptors (TLR) have been described as related to the immunosenescence process and carcinogenesis. The relationship of this gene family with carcinogenesis and immunoregulatory responses seems a promising field. The aim of this study is to compare the expression of TLR9 between geriatric cancer patients and elderly patients without any personal or familiar history of cancer, establishing if there are significant differences that may be explained by the carcinogenesis process rather than the immunosenescence one.

Methods

Between 2015 and 2016, a prospective cohort study including 195 elderly patients (≥ 60 years), 120 with incident cancer at the time of admission and 75 without any personal or familiar cancer history, assessed and collected sociodemographic and clinical variables and performed analysis of the peripheral blood in translational exploratory study. Determination of TLR9 was performed by flow cytometry with monoclonal antibodies anti-TLR9. Statistical analysis of the data was performed with GraphPad Prism.

Results

120 patients with incident prostate or breast cancer and 75 patients without any personal or familiar history of cancer, both with ≥ 60 years, were included. Most of the cancer patients were male (60%), while most of the patients without any cancer history were female (75%). Comparing the percentage and fluorescence values of TLR9 expression, there are significant differences (p < 0,0001) between the cancer patients group and the one without any personal or familiar cancer history.

Conclusions

There is significant difference between the expression of TLR9 in elderly cancer patients and elderly patients without personal or familiar history of cancer.

Clinical trial identification

Not applicable

Legal entity responsible for the study

Jurema Telles de Oliveira Lima

Funding

FACEPE/CNPq

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1654P - NSCLC patient immune cell profiling and response of tumor antigen specific CD8 T cells to checkpoint receptor antagonists (ID 2563)

Presentation Number
1654P
Lecture Time
13:15 - 13:15
Speakers
  • J. Lee
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The breakthrough in cancer immunotherapy of targeting PD-1 or PD-L1 and enabling T cells to attack tumor has opened new window for cancer treatment of many tumor types. Many of these immunotherapeutic approaches involve targeting specific immune checkpoints. To better understand the role of checkpoint receptors in cancer immunotherapy and explore new treatments, we analyzed cancer patient PBMCs and dissociated tumor samples from non-small cell lung cancer (NSCLC).

Methods

Transcripts of Cancer Testis (CT) antigens (NY-ESO-1, MAGE-A1, and MAGE-A3), a novel T cell inhibitory checkpoint receptor (TIGIT), and its respective ligands (PVR and PVRL2) as well as PDL1 from a cohort of NSCLC patients was analyzed. Immune cells from these patient samples were profiled and NYESO1/HLAA2 tetramer was used to detect NYESO1 specific CD8 T cells in HLAA2+ patients. Intracellular cytokine secretion was analyzed using a specially designed multiparameter flow cytometry panel for both general and NYESO-1+ CD8 T cells.

Results

The correlation between CT antigens, immune checkpoints, and effector T cell signature (CD8A, IFN-gamma, and Granzyme A) may help us understand 1) why certain patients have inflamed tumors and others have “cold” tumors; 2) why certain patients respond to anti-PD(L)1 therapy and others do not respond. Antibodies targeting both PD1 and another novel immune checkpoint receptor TIGIT showed best stimulatory effect on both antigen specific CD8 T cell number and intracellular IFN-gamma staining of those cells.

Conclusions

The data suggest that immune checkpoint receptor antibodies can be screened in real patient immune cells. Cotargeting multiple immune checkpoint receptors may provide superior efficacy to targeting single immune checkpoint receptor.

Clinical trial identification

Not applicable

Legal entity responsible for the study

Eli Lilly and Company

Funding

Eli Lilly

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1655P - Effect of neoadjuvant chemotherapy on correlation of tumor-associated macrophages with angiogenesis and lymphangiogenesis in human breast cancer (ID 3077)

Presentation Number
1655P
Lecture Time
13:15 - 13:15
Speakers
  • I. Mitrofanova
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Promoting role of tumor-associated macrophages (TAM) in angiogenesis and lymphangiogenesis was demonstrated in mouse tumor model sand human cancers. However, our latest studies revealed that amount of TAM in patients with breast cancer before as well after neoadjuvant chemotherapy (NAC) reversely correlated with lymphatic metastasis. Our aim was to analyse the effect of NAC on correlation of TAM in intratumoral compartments with angiogenesis and lymphangiogenesis.

Methods

115 female patients with breast cancer T1-4N0-3M0 were included in the study. 36 patients did not receive NAC, 79 received NAC. Expression levels of CD68 (general macrophage marker), stabilin-1 (marker of М2 macrophages), CD31 (marker of blood vessels) and LYVE1 (marker of lymphatic vessels) were identified by immunohistochemistry in 5 distinct areas of tumors: 1) soft fibrous stroma; 2) coarse fibrous stroma; 3) areas of maximum stromal-and-parenchyma lrelationship; 4) parenchymal elements; 5) gaps of ductal tumor structures.

Results

In breast cancer samples of patient who did not receive NAC direct correlation of CD68 expression in soft fibrous stroma and CD31 expression in coarse fibrous stroma (r = 0,87;р=0,02) was identified. However, reverse correlation was found between CD68 expression in gaps of ductal tumor structures and LYVE1 expression in soft fibrous stroma (r=-0,89;р=0,04). In contrast, in patients after NAC we identified a direct correlation between expression of CD68 and LYVE1 expression in the gaps of ductal tumor structures (r = 0,80;р=0,02). Expression of stabilin-1 in coarse fibrous stroma directly correlated with amount of LYVE1+ cells in areas with maximum stromal-and-parenchymal relationship (r = 0,76;р=0,04), but reversely correlated with the amount of CD31+ vessels in soft fibrous stroma (r= - 0,52;р=0,001).

Conclusions

Our data suggest that TAM before treatment support tumor angiogenesis however protect against lymphangiogenesis. After NAC TAM can switch their functional phenotype, do not support angiogenesis anymore but support lymphangiogenesis. The mechanism of chemotherapeutic programming of TAM remains to be identified.

This study was supported by grant RNF №14-15-00350.

Legal entity responsible for the study

Tomsk State University, Tomsk, Russian Federation

Funding

Tomsk State University, Tomsk, Russian Federation. This study was supported by grant RNF №14-15-00350.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1656P - Myeloid-derived suppressor cells are associated with a decrease of tumor antigen-specific Th1 immunity in non-small cell lung cancer (ID 3381)

Presentation Number
1656P
Lecture Time
13:15 - 13:15
Speakers
  • O. Adotevi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Myeloid Derived Suppressor cells (MDSC) are immune suppressive cells associated with poor survival in several cancers. In this study, we investigated their impact on spontaneous tumor antigen specific Th1 responses in Non-Small Cell Lung Cancer (NSCLC).

Methods

Monocytic MDSC (M-MDSC) (Lin-HL-DR-/loCD11b+CD14+CD33+) were measured in peripheral blood of 122 NSCLC patients and 34 healthy donors by flow cytometry. The presence of spontaneous anti-tumor Th1 response was measured by IFN-ɣ ELISPOT assay using mixture of peptides derived from lung cancer-associated tumor antigens such as telomerase, NY-ESO1 and Wilms Tumor-1. Patients’ samples were collected at baseline before any treatment.

Results

Higher percentage of circulating M-MDSC was found in NSCLC patients compared to healthy subjects (mean: 3.9±0.4% vs 1.5±0.4%, p < 0.01). Most patients presenting high level of M-MDSC in blood (≥ 5%: M-MDSChi) belonged to metastatic stage (34%, 21/61) compared to 14%, 9/61 in localized disease (p < 0.01). However, M-MDSChi status was associated with poor survival regardless the tumor stage (median OS: 11 vs 27 months in M-MDSChi and M-MDSClo groups respectively, p < 0.001). The IFN-ɣ+ antitumor specific T cell response was detected in 55.8% of patients and this frequency significantly dropped to 20% in M-MDSChi group. Furthermore, the magnitude of this response changed according to M-MDSC level (231 vs 116 IFN-ɣ+ specific T cells in M-MDSClo and M-MDSChi respectively p < 0.05). Finally, patients having both M-MDSChi status plus low anti-tumor T cell response exhibited a very poor survival (median OS of 6 months).

Conclusions

Our results show that high levels of circulating M-MDSC is associated with a decrease of pre-existing antitumor T cell response. The level of M-MDSC combined with antitumor T cell responses could predict distinct clinical outcome. Thus, monitoring M-MDSC in blood could be used as relevant immune biomarker in NSCLC.

Legal entity responsible for the study

Adotevi Olivier

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1657P - Revealing potential immune responses (IRs) in patients with advanced colorectal cancer (aCRC) on first line chemotherapy: A prospective study of neutrophil to lymphocyte ratio, immune function and outcome (ID 1951)

Presentation Number
1657P
Lecture Time
13:15 - 13:15
Speakers
  • S. Turnbull
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Neutrophil to lymphocyte ratio (NLR), a broad measure of inflammation and immune function, predicts outcome including overall survival (OS) in aCRC but the underlying mechanisms are unclear. Better understanding of IR in these patients may identify potential responders to novel immunotherapy. We investigated whether immune function correlated to NLR and how this altered during chemotherapy.

Methods

Peripheral blood was taken from 29 aCRC patients receiving 1st line chemotherapy (baseline and 6 weeks). NLR of ≥ 5 was defined as high. Immune function of peripheral blood mononuclear cells (PBMCs) was determined by NK cell activity (degranulation by CD107 expression and cytotoxic potential by 51Chromium release) against target tumour cells, T cell activity by IFN-γ ELISpot, cytokine secretion (Luminex) and immune cell activation (flow cytometry).

Results

High baseline NLR was associated with shorter OS compared to low NLR (6.6 vs. 18.8 months; HR = 3.6 [1.25 to 10.35] p = 0.0024). High NLR also correlated with a depressed IR, including decreased cytolytic activity of PBMCs (p = 0.046), NK cell degranulation at baseline and decreased levels of certain immune stimulatory cytokines. Low baseline NLR correlated with increased T cell activity against tumour-associated carcinoembryonic antigen (CEA) after 6 weeks of chemotherapy. Higher cytotoxic activity of PBMCs against target tumour cells at baseline (seen in the majority of patients with NLR <5) was associated with increased OS (p = 0.04). A drop in NLR during chemotherapy was associated with increased innate immune function as determined by NK cell degranulation (p = 0.004). Irrespective of NLR, frequency of Tregs reduced during chemotherapy and there was an increase in PD-1 expression on CD8+ T cells (p = 0.043), NK cells (p = 0.035) and monocytes (p = 0.016).

Conclusions

This study supports the poor prognosis of a high baseline NLR in aCRC and demonstrates its association with an attenuated IR. Chemotherapy can partially reverse this phenomenon, potentially enhancing anti-tumour immunity. If chemotherapy leads to a more effective anti-tumour IR, sequential immunotherapy could exploit this.

Legal entity responsible for the study

University of Leeds

Funding

Cancer Research UK, Yorkshire Cancer Research

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1658P - Somatic loss of the wild-type BRCA1 allele is not necessarily the first event in the pathogenesis of hereditary ovarian cancer: Implications for novel mechanism of acquired platinum resistance (ID 2662)

Presentation Number
1658P
Lecture Time
13:15 - 13:15
Speakers
  • E. Imyanitov
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Ovarian cancers (OC) arising in BRCA1 germ-line mutation carriers usually demonstrate high sensitivity to neoadjuvant chemotherapy (NACT), but almost inevitably relapse even after complete cytoreduction and continuation of systemic platinum treatment after surgery.

Methods

Changes in the somatic BRCA1 loss-of-heterozygosity (LOH) status were monitored in OC samples obtained before NACT, after NACT and at disease relapses.

Results

Loss of the wild-type BRCA1 allele was documented in 19 out 28 chemonaive OC samples included in the study. Surprisingly, 13 (68%) of these 19 OC demonstrated the retention of BRCA1 heterozygosity in the tumor tissue, which was surgically removed after median 3 cycles of NACT. TP53 mutations were easily detectable in some of the post-NACT samples thus confirming the good quality of microdissection. FISH assay and the analysis of adjacent SNPs revealed that the reversion of LOH status was attributed to selection of preexisting BRCA1-proficient clones but not to the second mutation in BRCA1 gene. Four tumor relapses were available for analysis; 3 out of these 4 tumors “restored” BRCA1 LOH during platinum-free interval. Next-generation sequencing analysis identified additional molecular events associated with evolution of OC clones upon platinum exposure and during treatment-free periods.

Conclusions

Isolated BRCA1 proficient cells are still present in chemonaive ovarian carcinomas with BRCA1 LOH, indicating that the somatic loss of the wild-type BRCA1 is not necessarily the first event in the pathogenesis of hereditary OC. These clones rapidly expand during even short-term systemic therapy. BRCA1-deficient cells have selective advantage in the absence of drug exposure and repopulate the tumor mass during platinum-free intervals. These fluctuations of BRCA1 LOH status explain why conventional platinum-based therapy, being capable to produce excellent tumor responses in BRCA1 germ-line mutation carriers, is not curative when considering long-term outcomes.

Legal entity responsible for the study

Laboratory of Molecular Oncology, N.N. Petrov Institute of Oncology, St.-Petersburg

Funding

Russian Scientific Fund (grant 14-25-00111)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1659P - Role of UBR5 mutations in DNA damage response in mantle cell lymphoma (ID 3915)

Presentation Number
1659P
Lecture Time
13:15 - 13:15
Speakers
  • O. Kutovaya
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Mantle cell lymphoma (MCL) accounts for 7% of non-Hodgkin lymphomas and represents a particularly challenging disease with patient outcomes inferior to most other lymphoma subtypes. We recently reported frequent mutations (18%) in UBR5, a gene encoding an E3 ubiquitin-protein ligase that has not been previously implicated in lymphomagenesis. All mutations were clustered within 100bp in or around exon 58 of UBR5 and are predicted to result in the loss of the conserved cysteine residue, which is responsible for binding the ubiquitin molecule. The recurrence and clustering of UBR5 mutations suggest their critical pathogenic nature in MCL that might be therapeutically targetable. The aim of this study is to determine the specific role of UBR5 mutations in the pathogenesis of MCL.

Methods

Mutations clustering in exon 58 of UBR5, as seen in MCL patients, were generated in three MCL cell lines (Granta-519, Jeko-1, and Mino) using the CRISPR-Cas9 genome engineering tool. First, global proteomes of UBR5 mutants and WT were analyzed by Tandem Mass Tag (TMT)-based mass spectrometry to identify proteins with differential expression due to the UBR5 mutations. Next, mass spectrometry-based immunoprecipitation proteomics (IP-MS) was employed to identify UBR5 interacting partners. Candidate UBR5 interacting proteins were functionally validated by flow cytometry, western blotting, co-immunoprecipitation, and immunofluorescence.

Results

The global proteome and IP-MS analyses identified a number of DNA damage response, chromosome organization, and cell cycle response proteins as the predominant proteins affected (p < 0.05). Our functional validation experiments show differential G2/M checkpoint activation and aberrant DNA damage response in UBR5 mutants vs WT through association of UBR5 with ATM interactor ATMIN.

Conclusions

The proteome and functional analyses are consistent with UBR5 functioning as a key regulator of cell signalling and point to the critical role of UBR5 as a novel regulator of DNA damage response. Next, our goal is to develop mouse xeno-transplantation MCL models and identify therapeutic agents that render sensitivity in xeno-transplantation models with UBR5 mutations.

Legal entity responsible for the study

Centre for Lymphoid Cancer, Vancouver, British Columbia, Canada

Funding

The Terry Fox Foundation

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1660P - ATM role in prostate cancer (PrCa) progression and survival (ID 4570)

Presentation Number
1660P
Lecture Time
13:15 - 13:15
Speakers
  • Y. Cendón Flórez
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Germline and/or somatic aberrations in ATM gene have been recently identified in up to 5% of PrCa cases. It has been also described that mutations in DNA repair genes predispose individuals to more aggressive and lethal phenotypes. For these reasons, our goal is to investigate the role of ATM in PrCa progression.

Methods

To study the cooperation of Atm in prostate cancer progression in vivo, we crossed the transgenic mouse model TRAMP with Atm null mice in C57BL/6 background. This model allows us to elucidate the progression of prostate cancer in wild-type (+/+), heterozygous (+/-), and homozygous (-/-) Atm loss in mice. PIN, invasive and metastatic prostate cancer as well as survival curves were compared for the three arms. In addition, in a large cohort of mCRPC (n = 419) from the prospective PROREPAIR-B study (NCT03075735), in which a large panel of germline DNA repair genes were studied, we compared the clinico-pathological characteristics at baseline and mCRPC diagnosis between germline ATM mutation carriers and non-ATM carriers. Chi-Square and Exact Fisher test, the Kaplan-Meier method and Long-rank test were used for statistical analyses.

Results

Twenty eight TRAMP(T/+); Atm(+/+) and 45 TRAMP(T/+); Atm(+/-) mice were follow-up until sacrifice-endpoint. Heterozygous Atm loss mice presented higher frequency of metastasis in the necropsy compared to Atm wild-type (44% vs. 21%, p = 0,045) and shorter median survival (26 vs. 32 weeks, p = 0,008). There were not significant different observed in PIN or invasive tumour prevalence. TRAMP(T/+); Atm(-/-) mice were excluded from analyses due to the early development of lethal thymomas requiring sacrifice before week 16. On the other hand, 8 patients out of 419 were found to harbour germline pathogenic ATM mutations (1.9%), and compared with non-ATM carriers presented higher frequency stage IV at diagnosis (63% vs. 34%, p = 0.2), bone metastasis (100% vs. 82%, p = 0.4) without other relevant differences found in these preliminary analyses.

Conclusions

Aberration in the ATM gene may favour metastatic progression in PrCa in prostate cancer preclinical models, although its clinical implication will require further clarification in the future.

Clinical trial identification

Part of the results came from the prospective PROREPAIR-B study (NCT03075735)

Legal entity responsible for the study

Spanish National Cancer Research Centre

Funding

Prostate Cancer Unit-Spanish National Cancer Research Centre

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1661P - Synergistic inhibition of CEP55 induces mitotic catastrophe and specifically targets aggressive breast cancer (ID 4473)

Presentation Number
1661P
Lecture Time
13:15 - 13:15
Speakers
  • D. Sinha
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Triple negative breast cancers (TNBCs) are the most aggressive and profoundly heterogeneous form of breast cancer (BC), treatment of which is a prevalent challenge faced in clinics. CEP55, discovered first by our laboratory, is a key regulator of cytokinesis, error in which roots to multi-nucleation. Function of CEP55 is critically delimited by ERK2/PLK1 dependent phosphorylation, for accurate cytokinesis. Research has demonstrated connotation of CEP55 with numerous cancers including BC as higher CEP55 mRNA expression is allied to worse prognosis and poor survival. We hypothesised that, CEP55 controls fate of aneuploid cell population among aggressive BC that are heavily reliant on mitotic genes for tumour progression, thus can be targeted for therapy development.

Methods

Using in vitro studies we demonstrated that depletion of CEP55 sensitizes TNBC cells to anti-mitotic drugs like PLK1 inhibitor to induce CDK1-Caspase 3-dependent mitotic catastrophe due to unscheduled CDK1/Cyclin B activation. Also we showed ERK1/2 transcriptionally controls CEP55 hence inhibition of MEK1/2 using the small molecule inhibitor Selumetinib, can mimic depletion of CEP55 in vivo.

Results

We rationalised the usage of a MEK1/2 inhibitor in combination with a PLK1 inhibitor across a series of BC cell lines. We observed synthetic lethality among the aggressive hormone receptor negative lines with higher CEP55 expression compared to normal like and receptor positive lines with lower CEP55 level. The combination synergistically amplified apoptosis of aneuploid population via premature entry of these cells into mitosis in the presence of antimitotic drugs due to exhaustion of CEP55. We have also validated this synergistic effect of MEK1/2 and PLK1 inhibition using xenograft models, results of which imitated the in vitro findings.

Conclusions

We propose a novel treatment tactic of MEK1/2 -PLK1 dual combination for selectively targeting CEP55 over-expressing BC in the clinics.

Legal entity responsible for the study

QIMR Berghofer Medical Research Institute

Funding

Cancer Council Queensland (CCQ) and National Health & Medical Research Council

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1662P - Synergistic antitumor effects of OT-101 (trabedersen), a transforming growth factor-beta 2 (TGF-β2) antisense oligonucleotide (ASO) and chemotherapy in preclinical tumor models (ID 5465)

Presentation Number
1662P
Lecture Time
13:15 - 13:15
Speakers
  • O. D'Cruz
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Overexpression of TGF-β2 has been implicated in the malignant progression of tumors by inducing immunosuppression, proliferation, angiogenesis and metastasis. OT-101 (Trabedersen) is a phosphorothioate ASO designed to specifically target human TGF-β2 mRNA. Herein, we report the synergizing effect of OT-101 with chemotherapy in multiple human tumor xenograft models for further exploration of clinical combination strategies.

Methods

OT-101 was administered as single agent (1-64 mg/kg, qdx3/wk or qdx21) and in combination with Gemcitabine (GEM, 15 mg/kg, qdx2/wk), Dacarbazine (DTIC, 1-10 mg/kg, qdx4/wk) or Paclitaxel (PTX, 10 mg/kg, qdx5) to nude mice (10/subgroup) bearing either (i) orthotopic human L3.6pl pancreatic cancer (PAC), (ii) human metastatic C8161 melanoma, (iii) SC glioblastoma (U87) or (iv) SC ovarian (SKOV-3) tumors. Mice were monitored for adverse effects, body weight loss, tumor size and survival outcome. Lymph node and liver surface and micro-metastases as well as size and weight of the pancreatic tumors were determined. Tumor sections were stained with anti-BrdUrd and CD31 antibodies to determine tumor cell proliferation and vascularization, respectively.

Results

OT-101 significantly reduced tumor growth (p = 0.0084), lymph node metastasis (p = 0.023), and tumor angiogenesis (p < 0.0001) versus untreated control in the PAC model. OT-101 demonstrated synergy in tumor growth inhibition and increased survival in human malignant melanoma (C8161, p = 0.038, vs. DTIC alone), glioblastoma (U87, p = 0.001 vs. PTX) and ovarian (SKOV-3, p < 0.05 vs. PTX) cancer models when combined with either DTIC (C8161) or PTX (U87 and SKOV-3). No synergy was observed with GEM (PAC). The combination regimen tested was effective and tolerable. Significant antitumor activity was achieved at HED of 80 mg/m2/day which is well below the optimized clinical dose used for IV infusion of patients at 140 mg/m2/day.

Conclusions

The preclinical data laid the groundwork for establishing combination therapies in the clinic. Of interest is the preferential synergy between OT-101 and PTX or DTIC, but not with GEM.

Legal entity responsible for the study

Autotelic Inc

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1663P - Targeting thioredoxin reductase 1 in novel combination therapies in p53 mutant triple negative breast cancer (ID 4470)

Presentation Number
1663P
Lecture Time
13:15 - 13:15
Speakers
  • P. Raninga
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The TP53 gene is frequently mutated in human cancers including triple negative breast cancers (TNBCs) (∼84% patients). Although TP53 mutation is the only oncogenic driver in TNBCs, no targeted therapies for mutant p53 (mtp53) TNBCs are available. We aim to identify novel therapeutic targets and combination therapies for mtp53 TNBCs.

Methods

A large-scale genomic analysis was performed using the TCGA database to analyse the expression of various antioxidant genes in Mt and wild-type (wt) p53 BC cells. Thioredoxin reductase 1 (TrxR1) protein levels and redox activity were measured by western blot and DTNB reduction assay, respectively. Mt and wt p53 cells were treated with gold-based TrxR1 inhibitor and APR-246 and subsequently analysed for cell proliferation, apoptosis, and cell cycle progression. Phospho-histone H3 (pHH3) Ser10 expression was analysed by FACS.

Results

We observed significant upregulation of TrxR1, a redox gene, in mtp53 BC patients compared to wt patients. TrxR1 protein levels and redox activity were higher in mtp53 cells compared to wt cells. Notably, TrxR1 inhibition selectively induced apoptosis in mtp53 BC cells, but not in wt cells. Upon treatment with TrxR1 inhibitor, a significant proportion of mtp53 cells arrested in the G2/M phase with a concomitant increase in pHH3 Ser10, a marker of mitotic chromatin condensation. Thus, TrxR1 inhibition may lead to mtp53 TNBC cell death by causing mitotic catastrophe. APR-246, known to restore wild-type activity of mtp53 in many cancers, alone failed to induce apoptosis in mtp53 BC cells. However, co-treatment of APR-246 with a sub-lethal concentration of TrxR1 inhibitor resulted in a synergistic effect in mtp53 cells.

Conclusions

Inhibiting TrxR1 may represent an effective therapeutic strategy for mtp53 TNBCs. These results warrant the clinical evaluation of a novel combination therapy using APR-246 and TrxR1 inhibitors for mtp53 TNBC patients.

Legal entity responsible for the study

QIMR Berghofer Medical Research Institute

Funding

National Health and Medical Research Council, Australia

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1664P - Liprin-α4 could be a potential therapeutic target for pancreatic cancer (ID 1343)

Presentation Number
1664P
Lecture Time
13:15 - 13:15
Speakers
  • A. Yamasaki
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In pancreatic cancer whose microenvironment is extremely hypoxic condition, the analysis of signal transduction under hypoxia is thought to be significantly important. By investigating microarray analysis of pancreatic cancer cultured between under normoxia and under hypoxia, we found that the expression of leukocyte common antigen related (LAR)- interacting protein (liprin)-α4 was extremely increased under hypoxia compared to under normoxia. In the present study, the biological significance of liprin-α4 in pancreatic cancer was investigated and whether liprin-α4 could be a therapeutic target for this refractory cancer was estimated.

Methods

Three pancreatic ductal adenocarcinoma cell (PDAC) lines (ASPC-1, SUIT-2, and PANC-1) were cultured under normoxia (20%O2) and under hypoxia (1%O2), and were used as target cells. Inhibition of liprin-α4 was performed using liprin-α4 siRNA. Expression of liprin-α4 was analyzed by real time RT-PCR, western blot and immunofluorescent staining. Proliferation was estimated by cell count and MTT assay. Invasion was estimated by matrigel invasion assay. Mice xenograft experiments were performed using BALB/c nude female mice. Surgically resected human pancreatic cancer specimens were used for immune staining.

Results

1) Expression of liprin-α4 was increased in PDAC under hypoxia compared to normoxia. 2) Liprin-α4 suppression decreased invasion through inhibition of endothelial mesenchymal transition in PDAC under hypoxia. 3) Liprin-α4 inhibition decreased proliferation of PDAC under hypoxia In vitro. 4) Tumor volume in mice injected with liprin-α4-inhibited PDAC was significantly lower than that in control mice. 5) Signaling from liprin-α4 was through PI3K and MAPK signaling pathways. 6) Relation between hypoxia inducible factor-1α (HIF-1α) expression and liprin-α4 expression was observed by immunofluorescent staining using surgically resected pancreatic cancer specimen.

Conclusions

These results suggest that liprin-α4 which is more expressed under hypoxia, plays pivotal role for inducing malignant phenotype such as proliferation and invasion in pancreatic cancer, and that liprin-α4 could be an effective therapeutic target for pancreatic cancer.

Legal entity responsible for the study

Cancer Therapy and Research, Kyushu University Hospital,

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1665P - Dual targeting of cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE) by miR-4317 displays a synergistic efficacy in repressing breast cancer progression (ID 2512)

Presentation Number
1665P
Lecture Time
13:15 - 13:15
Speakers
  • R. Youness
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Recently, hydrogen sulphide (H2S) and its synthesizing enzymes, CBS and CSE, have been casted as pleiotropic regulators in the malignant transformation process. H2S paradoxically acts as oncogenic mediator in ovarian and liver cancers, and as tumor suppressor in prostate and gastric carcinomas. However, the link between H2S and Breast cancer (BC) remains unclear. Thus we aimed at unraveling the association between H2S and its synthesizing enzymes in BC progression. Furthermore, it was essential to evaluate their possible adoption as therapeutic targets in BC through their dual targeting by short non-coding RNAs.

Methods

Breast tissues were collected from 30 BC patients. Ki67 levels were quantified using immunohistochemistry. MDA-MB-231 and MCF7 cells were cultured and transfected with different oligonucleotides and/or treated with NaHS, an exogenous source of H2S. Total RNA was extracted and quantified by qRT-PCR. Cellular viability, proliferation, and migation were measured using MTT, BrdU and scratch assays respectively. Bioinformatic analysis was performed to predict novel miRNAs that could target both CBS and CSE.

Results

CBS and CSE were significantly upregulated in BC tissues. Patients with high Ki67 scores showed the highest expression levels of CBS and CSE. Knocking down of CBS and CSE using siRNAs resulted in a significant attenuation of different hallmarks of BC. On the other hand, NaHS resulted in an increase in BC progression. miR-4317 was found to putatively target both CBS and CSE oncogenes with high binding scores. Ectopic expression of miR-4317 in BC cell lines resulted in a simultaneous reduction of CBS and CSE transcripts which was associated with a concomitant reduction in cellular viability, proliferation and migration. Finally, co-treatment of miR-4317 and NaHS resulted in abrogation of miR-4317 tumor suppressor activity.

Conclusions

This study showed a marked upregulation of CBS and CSE in BC tissues and characterized them as aggressive oncogenic drivers in BC. Moreover, miR-4317, a novel tumor suppressor in BC, displayed a synergistic effect in halting BC progression via twin-targeting CBS and CSE and diminishing H2S levels in BC cell lines.

Legal entity responsible for the study

German University in Cairo

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1666P - Met/Axl system as a dual target in the mesothelioma pathway and invasiveness (ID 3243)

Presentation Number
1666P
Lecture Time
13:15 - 13:15
Speakers
  • G. Viscardi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Malignant pleural mesothelioma is an aggressive and highly lethal disease. Conventional chemotherapies and radiation therapy have limited efficacy. Many evidences suggest the roles of receptors tyrosine kinase (RTKs) in mesothelioma pathogenesis, in particular epidermal growth factor (EGFR), Met and Axl. Axl activation is involved in proliferation and inhibition of apoptosis, and its over-expression represents a key molecular determinant underlying the development of acquired resistance to targeted anticancer agents.

Methods

Different histological types, epithelioid, sarcomatoid and mixed, of human mesothelioma cell lines were used. Protein levels of Met, Axl and its ligand, growth arrest-specific 6 (Gas6) were evaluated by Western Blot analysis. We conducted in vitro treatments with different doses of Foretinib, dual inhibitor of Met and Axl, in order to demonstrated the variation of cell proliferation and migration through MTT and Colony Forming Assay at the range dose 0.5-1 µM of Foretinib. Lastly, the rate of cell apoptosis was quantified by flow cytometry.

Results

The presence of Met, Axl and Gas6 proteins were found in all cell lines analyzed with different expression pattern. The dose escalation of Foretinib from 0.01 µM to 2 µM strongly inhibited cell proliferation and migration of mesothelioma cell lines. Treatment with Foretinib (at the dose 0.5 µM and 1 µM), determining a significantly increase of apoptosis rate (up to 50%) in specific histological type suggesting a different cell sensibility.

Conclusions

The co-activation of MET and AXL in mesothelioma cell lines suggests that these kinases could serve as novel therapeutic targets. MET and AXL inhibitors could be used as novel anticancer therapies influence clinically meaningful end points including metastatic recurrence and survival in the majority of tumour types.

Legal entity responsible for the study

University of Campania “Luigi Vanvitelli”

Funding

Associazione Italiana per la Ricerca sul Cancro (AIRC)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1667P - Targeting CXCR4 and FAK in non-small cell lung carcinomas with co-inactivated p53 and PTEN tumor suppressors (ID 4441)

Presentation Number
1667P
Lecture Time
13:15 - 13:15
Speakers
  • A. Podolski-Renic
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

In this study we evaluated potential of targeting CXCR4 and focal adhesion kinase (FAK) in suppressing metastatic spread of p53/PTEN deficient non-small cell lung carcinomas (NSCLCs).

Methods

We first examined the invasive characteristics of NSCLC cells with suppressed p53 and PTEN activity using wound healing, gelatin degradation and invasion assays. Namely, NCI-H460 cells with applied pharmacological inhibition of wild type p53 and PTEN activity (NCI-H460p53-/PTEN-) were analyzed along with COR-L23 cells that have intrinsically inactive both tumor suppressors. Further, changes in the expression of CXCR4 and FAK were evaluated by RT-qPCR and Western Blot analysis. Finally, we tested the ability of CXCR4 and FAK inhibitors (WZ811 and PF-573228, respectively) to suppress the migratory and invasive potential of p53/PTEN deficient NSCLC cells, in vitro and in vivo using orthotropic metastatic lung carcinoma mouse model.

Results

Our results showed that cells with mutually inactive p53 and PTEN have significantly increased migratory and invasive potential. Such invasive phenotype is associated with hyperactivation of CXCR4 and FAK and their downstream AKT and ERK signaling pathways. Treatments with WZ811 and PF-573228 significantly reduced migratory and invasive capacity of NCI-H460p53-/PTEN- and COR-L23 cells that was accompanied by the downregulation of AKT signaling. In addition, these two inhibitors showed trend to improve survival of SCID mice with orthotopicaly inoculated COR-L23 cells that extensively invaded lung parenchyma and developed distant metastases compared to NCI-H460 derived tumors.

Conclusions

Overall, we demonstrated that p53/PTEN deficient NSCLCs have extremely invasive phenotype and provided a rationale for the use of CXCR4 or FAK inhibitors for the suppression of NSCLC dissemination.

Legal entity responsible for the study

This study was supported by the Ministry of Education, Science and Technological Development of the Republic of Serbia (Grant Nos III41031 and 173020), COST Action CM1106 „Chemical Approaches to Targeting Drug Resistance in Cancer Stem Cells “and COST Action CM1407 „Challenging organic syntheses inspired by nature - from natural products chemistry to drug discovery”.

Funding

This study was supported by the Ministry of Education, Science and Technological Development of the Republic of Serbia (Grant Nos III41031 and 173020), COST Action CM1106 „Chemical Approaches to Targeting Drug Resistance in Cancer Stem Cells “and COST Action CM1407 „Challenging organic syntheses inspired by nature - from natural products chemistry to drug discovery”.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1668P - Pin1 protein: A druggable target in high grade serous ovarian cancer (ID 5001)

Presentation Number
1668P
Lecture Time
13:15 - 13:15
Speakers
  • C. Russo Spena
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Epithelial Ovarian cancer (EOC) is the 5th leading cause of cancer death in the USA and the High Grade Serous-EOC is the most common and aggressive type characterized by mutations in the p53 gene (>90%). Pin1 has been demonstrated to activate a mutant p53 transcriptional program. It binds specific phosphoSer/Thr-Pro-motifs and catalyses the cis/trans conformational switch of target proteins. In HGS-EOC, Pin1 is overexpressed in about 50% of cases suggesting that it may be a potential therapeutic target. The Pin1 inhibition affects cellular proliferation, migration, invasion, new angiogenesis and apoptosis suggesting a pivotal role in cancer. Nevertheless, there are still deficiencies in producing Pin1 ligands: Pfizer has reported inhibitors that have poor permeability and low efficacy. Here, it is reported that the encapsulation of these inhibitors in liposomes increases the cytotoxic activity on ovarian cancer cells.

Methods

The inhibitor was encapsulated in ionisable cyclodextrins via a pH gradient. PEG-liposomes were prepared using different molar ratio of cholesterol and lipids. Cell viability was tested with an MTT-like assay. The liposome characteristics were evaluated by DLS and zeta potential. The loading efficiency of drug was calculated via UV-Visible method and the release with a dialysis membrane.

Results

Pegylated liposomes of about 100 nm were synthesized for the encapsulation of drug. This complex has a promising loading efficiency and release rate. These characteristics allow an efficient delivery in different ovarian cancer cells line achieving the IC50 values in the low micromolar range. Instead, inhibitor alone did not change the cell viability.

Conclusions

In summary, we have created a new formulation of Pin1 liposomal inhibitor that can be used to kill ovarian cancer cells. Therefore, for further application in vivo, the inhibitor retention would enhance antitumor efficacy. In fact, the liposome system could have a high concentration in the tumour, thanks to enhanced permeability and retention effect, overtake the tumoral barrier and represents an option for the therapy of ovarian cancer.

Legal entity responsible for the study

Graduate School in Chemistry - University of Trieste

Funding

AIRC - Associazione Italiana Ricerca sul Cancro

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1669P - RalB GTPase: A potential novel target for RAS mutant colorectal cancer (ID 5369)

Presentation Number
1669P
Lecture Time
13:15 - 13:15
Speakers
  • H. Khawaja
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Colorectal cancer is the 3rd most common cancer in the UK, with around 40,000 new cases diagnosed annually. CRC patients have a 5-year overall survival rate of < 10% and more than 50% will die of metastatic disease. Intrinsic or acquired resistance to chemotherapeutic drugs is a major problem in CRC and developing an effective treatment strategy is therefore of the utmost importance. CRC cases harbouring RAS mutations (>50% cases) are associated with poor prognosis; this mutation has proven to be an important predictive factor for response to EGFR targeted therapies. Failure to target the RAS oncogene has resulted in a concentrated effort to discover targets within the downstream components of this pathway. In this study, we evaluate the roles of the small GTPases, RalA and RalB, as novel targets in RAS mutant (MT) CRC. The RALGDS/RAL pathway constitutes a RAS effector pathway and mediates cell survival, proliferation and tumorigenesis. RalB in particular contributes to cell survival through TBK1 signalling.

Methods

We used an siRNA-based approach silencing RALA and RALB both individually and simultaneously in a panel of KRASMT and WT cells with and without the addition of the MEKi AZD6244 (Selumetinib). Knockdown efficiency and subsequent signalling events were assessed by western blotting. Flow cytometry and MTT assays were used to measure cell death and cell viability respectively. Connectivity mapping using data from microarray experiments was used to identify drugs mimicking the phenotype observed with siRALB, subsequently leading to the investigation of a TBK1 inhibitor which is currently ongoing.

Results

We found that silencing RALB but not RALA, led to the greatest amount of cell death in RASMT but not WT CRC cells. In addition, a significant increase in cell death was observed when RALB silencing was combined with MEK inhibition. Cell death was found to be mediated by Caspase 8 and involved an upregulation of death receptor 5 (DR5). Furthermore, TBK1 inhibition was found to mimic the phenotype observed with siRALB.

Conclusions

RalB but not RalA, is associated with cell survival and may contribute to drug resistance in RASMT CRC. Thus, the development of novel RalB-specific therapies may lead to new treatment strategies for RAS MT CRC.

Legal entity responsible for the study

Queen's University Belfast

Funding

Queen's University Belfast, Cancer Research UK

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1670P - Serial genotypic characterization of circulating tumor cells (CTCs) in patients with metastatic castration resistant prostate cancer (mCRPC) undergoing treatment with abiraterone acetate (abi) or enzalutamide (enza) (ID 1291)

Presentation Number
1670P
Lecture Time
13:15 - 13:15
Speakers
  • N. Dizman
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

While enza and abi have substantially improved outcomes for patients (pts) with mCRPC, de novo and acquired resistance mutations are increasingly recognized.

Methods

Pts receiving abi or enza in the course of routine clinical care were consented for blood collection at weeks 0, 4, 8 and 12 of therapy, and at the time of progression (based on Prostate Cancer Working Group 3 [PCGW3] criteria). CellSearch was used for CTC enumeration; individual cells were isolated and subsequently classified for EpCAM and C45 positivity. RNA sequencing (RNA-seq) was performed on pools of up to 10 CTCs.

Results

Amongst 36 pts enrolled, median age was 71 (range, 54-84) and median PSA was 21.9 ng/dL (range, 0-918.3). Regarding treatment, 21 pts received abi and 15 received enza. By PCWG3 criteria, 23 pts met the definition of progression on abi or enza. Mean/median CTC count was 158/5 (IQR 25%-75%, 0-15). On RNA-seq of CTCs collected at the time of progression, AR was the most mutated gene followed by ATRX, GNAS, FOXA1, KMT2A and CNOT1. Several deleterious mutations in the DNA damage response genes were noted including frameshift mutations in PRKDC, MSH2 and MLH1. Differential gene expression analysis between abi/enza sensitive and abi/enza resistant samples revealed 2100 differentially regulated genes in drug-resistant CTCs. Ingenuity pathway analysis was used to identify pathways altered due to differential regulation of these genes. Among these pathways, TGFβ and CCDN1 signaling were found to be significantly up-regulated in drug resistant CTCs. In vitro enza-resistant models will be presented, offering validation of our clinical findings.

Conclusions

RNA-seq of CTCs representing abi/enza sensitive and resistant states can identify potential mechanisms of resistance. Therapies targeting the downstream signaling mediated by CCND1, such as CDK4/6 inhibitors (e.g., palbociclib or ribociclib), could avert resistance. Targeting TGFβ, another putative mediator of resistance, may be warranted.

Legal entity responsible for the study

Sumanta Kumar Pal

Funding

Janssen Pharmaceutical Company

Disclosure

J. Patel, B. Foulk, V. Bhargava, D.A. Smirnov: Working for Janssen Pharmaceutical Company All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1671P - A novel circulating cell free DNA-based assay can predict tumor response to systematic chemotherapy (ID 1270)

Presentation Number
1671P
Lecture Time
13:15 - 13:15
Speakers
  • T. Toshima
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Although circulating cell-free DNA (cfDNA) in blood is being touted as a frontier noninvasive approaches, its clinical utility still remains questionable. The purpose of this study was to compare the efficacy of cfDNA by comparing with blood CEA levels and radiological evaluation in patients with unresectable metastatic colorectal cancer (mCRC) during treatment of systemic chemotherapy.

Methods

In this study, 12 patients with mCRC who were intended to receiving systemic chemotherapy were enrolled. Methylation status of CpG sites, considered as cancer-specific alteration, and concentration of cfDNA were evaluated from blood plasma obtained before administration of systemic chemotherapy in each treatment cycle. To analyze aberrant cancer-specific methylation, we modified the highly sensitive assay for bisulfite DNA (Hi-SA) followed by fluorescence-based PCR, as reported previously (JNCI 2009). Our modified methodology can detect 8 loci of target promoters, therefore methylation score (MS) could be ranged from 0 to 8 at a given time.

Results

Of the 12 patients enrolled, 10 patients experienced radiological progressive disease (PD). Plasma MS was significantly increased before radiological PD in 8 of 10 patients with PD. Thus MS had the median lead time of 73 days (range: 0-231 days) before documentation of radiological PD. In contrast, serum CEA level could predict PD only in the 2 patients before documentation of their radiological PD. Consequently, plasma MS could predict radiological PD with the median lead time of 9 days (range: 0-21 days) compared with serum CEA. We also examined whether cfDNA concentration level in plasma was associated with radiological PD. Of the 12 patients, only 3 patients increased cfDNA concentration level before radiological PD with the median lead time of 88 days (range; 21-140 days).

Conclusions

Our circulating cell free DNA-based assay is a robust methodology for capturing DNA methylation in circulating cell-free DNA in plasma, and is useful for the early identification of CRC patients that are at risk of developing PD prior to radiographic documentation.

Legal entity responsible for the study

Takeshi Nagasaka

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1672P - ctDNA might expand therapeutic options for second line treatment of KRAS mutant mCRC (ID 3703)

Presentation Number
1672P
Lecture Time
13:15 - 13:15
Speakers
  • P. Gazzaniga
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

KRAS mutations predict failure of anti-EGFR therapies, thus genotyping colorectal cancer (CRC) is crucial for personalized treatments. Cancer heterogeneity hamper the assessment of KRAS mutational status in tumor tissues, leading to the search for alternative sources of cancer genetic information. ctDNA of patients treated with anti-EGFR drugs exhibit pulsatile levels of KRAS mutations, revealing that the CRC genome adapts dynamically to intermittent EGFR blockade. These data support the use of liquid biopsy to monitor the molecular underpinnings of resistance to anti-EGFR agents. Research has been selectively concentrated on the emergence of resistant clones in the blood of patients with wtKRAS CRC as biomarker of anti-EGFR therapy resistance. Conversely our group demonstrated that patients with metastatic CRC harboring mutated primary tumors, thus not candidate to EGFR inhibitors, frequently have wtKRAS circulating tumor cells in blood. To explain the prevalence of wtKRAS clones in these patients, the generation of hypoxia has been suggested. We aimed to determine if anti-angiogenic drugs might drive the biological evolution of mKRAS clones towards a prevalent wtKRAS disease, by ctDNA.

Methods

Ten patients with histologically confirmed mKRAS mCRC candidate to first-line anti angiogenic drugs were prospectively enrolled. To investigate whether wtKRAS clones emerge as dominant under treatments, serial blood draws were performed at baseline and at 3 months months of treatment. Idylla™ (Biocartis) ctKRAS Mutation Assay was used to track KRAS mutational status in serial ctDNA determinations for each patient.

Results

At baseline, KRAS mutational status in ctDNA was found concordant with tumor tissues in all patients analysed. At 3 months,3/10 (30%) of mKRAS CRC patients treated with antiangiogenic drugs switched to wtKRAS ctDNA in peripheral blood.

Conclusions

These preliminary data suggest that patients with mKRAS colon cancer not unfrequently switch to a prevalent wtKRAS disease in course of treatment with anti-angiogenic drugs. if confirmed in a large patients population, these results might shift second-line therapeutic options for KRAS mutant mCRC patients from insufficient to promising.

Clinical trial identification

not applicable

Legal entity responsible for the study

Paola Gazzaniga

Funding

Merck

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1673P - Ex vivo expansion of circulating tumor cells for individualized drug susceptibility in patients with advanced or recurrent oesophageal cancer (ID 3112)

Presentation Number
1673P
Lecture Time
13:15 - 13:15
Speakers
  • V. Kumar
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Esophageal cancer (EC) is the eighth most common cancer in the world. The incident rate of EC is significantly high in Asian countries compared to rest of the world. Circulating tumor cells (CTCs) derived from EC have the potential to be precursors of metastasis. It is therefore of paramount interest to isolate and characterize CTCs from EC patients to monitor and detection of recurrence. The aim of present study is to evaluate drug response using patient-derived CTC cultures obtained from EC.

Methods

Custom microfabricated tapered microwells will be integrated with microfluidics to expand CTC clusters without any prior pre-enrichment. The established CTC cluster assay will be used to screen anticancer drugs. The drug concentrations selected will be centered on the IC50 that had previously established for each drug across EC cell lines. Cluster formation in culture will be correlated with overall patient survival. 50 patients with a proven diagnosis of EC attending the Department of Surgical Oncology, Kidwai Institute of Oncology will be enrolled into the study.

Results

Our initial results showed CTC clusters formation in the patients with metastatic EC. This cluster formation was affected by the presence and duration of systemic therapy. We observed a progressive reduction in cluster formation in samples from patients who had undergone increasing longer treatment.

Conclusions

Our result suggests that CTC cluster can be used to rapid evaluation of drug response. We would further use the CTC cluster assay as a potential tool for evaluating patient prognosis during treatment. The study will be employed to determine the drug susceptibility pattern in individual patients and also provide therapeutic choices for personalized treatment.

Legal entity responsible for the study

Kidwai Memorial Institute of Oncology, Institute of Bioinformatics

Funding

Department of Science and Technology, (DST) Government of India

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1674P - Exploratory study of CK-M30 and pHH3 expression in Circulating Tumor Cells (CTCs) as biomarkers of docetaxel (DOC) efficacy in metastatic castration resistant prostate cancer (mCRPC) (ID 4588)

Presentation Number
1674P
Lecture Time
13:15 - 13:15
Speakers
  • P. Nombela Blanco
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

A drop in CTCs counts as early as 4 weeks following treatment initiation have been suggested as an indicator of overall survival (OS) benefit. DOC remains a pivotal treatment in mCRPC for which there are no early pharmacodynamic (PD) markers of its activity in patients (pts). CTCs may be used as surrogate tumor tissue to study PD markers of apoptosis (CK-M30) or mitosis arrest (pHH3) in mCRPC pts receiving DOC.

Methods

We conducted a prospective 2-cohort multicenter exploratory study in mCRPC pts progressing by PCWG2 criteria who were eligible for DOC 75 mg/mˆ2. Pts were prescreened using the CellSearch system and selected if CTCs≥5/7.5mLs of blood (Basal 1). A 2nd blood sample (Basal 2) was drawn in eligible pts within 0-7 days prior to C1D1 of DOC and further samples were collected at 8h, 24h, 7d and 21d. Directly conjugated mAb against pHH3 and CK-M30 were used in combination with CellSearch. Statistical analyses were performed to evaluate the baseline and post-treatment variability. Increases in % of biomarker CTC+ greater than the median baseline variability were correlated with achieving a 50% PSA response (PSA50) and OS from DOC start using chi-square and long-rank test, respectively.

Results

60 mCRPC pts (CK-M30 = 30; pHH3 = 30), 95% ECOG 0-1, 95% and 17% have bone and visceral metastases respectively, received a median of 7 cycles (range 2-10) of DOC. Biomarker results are summarised in Table.

1674P Variability

Basal 1 (N = 60) Median (Range)Basal 2 (N = 57) Median (Range)%marker Corr. CoeffPost-24h (n = 59) median (Range)P-value Basal vs post-24h
Cohort CK-M30CTC/7.5mL9 (5-1266)8 (3-863)0.54 p = 0.7639 (4-1129)NS
CK-M30+48% (0-76%)35% (0-100%)51% (0-100%)
Cohort pHH3CTC/7.5mL10 (5-567)12 (4-623)0.98 p < 0.0019 (3-584)<0.001
pHH3+0% (0-10%)0% (0-8%)7% (0-67%)
ResponseNPSA50p-valueOS median (CI95%)p-value
CK-M30+ change 24hCKM30 + >50%52NS24 m (–)NS
CKM30 + <50%251121 m (12-30)
pHH3+ change 24hpHH3 + >10%1380.04717 m (12-22)NS
pHH3 + <10%16412 m (6-18)

Conclusions

pHH3 in CTC+ may be a potential PD biomarker of a favourable response to DOC treatment.

Legal entity responsible for the study

Spanish National Cancer Research Centre

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1675P - Risk of recurrence prediction and optimum treatment planning for early stage breast cancer patients: A cost-effective, accurate and broad based solution for Asia (ID 863)

Presentation Number
1675P
Lecture Time
13:15 - 13:15
Speakers
  • S. Somashekhar
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Current molecular risk stratification tests have helped clinicians to optimize Chemotherapy for early stage breast cancer patients leading to huge savings in treatment costs and improved quality of life. However, current tests are not impactful in the Asia due to the extreme cost-sensitivity of the market. Aim of this study was to develop and validate a cost-effective, broad based and robust test to stratify early stage hormone receptor positive patients based on individual risk of recurrence.

Methods

A retrospective cohort of 300 patients, was used to develop ‘CanAssist-Breast’- a Morphometric Immunohistochemistry based test comprising 5 biomarkers plus three clinical parameters (Tumor size, node status and grade) using SVM based algorithm. CanAssist-Breast biomarkers belong to key signaling pathways involved tumor invasion and chemotherapy resistance.

Results

CanAssist-Breast classifies patients into ‘low or high’ risk of recurrence based on “CanAssist-Breast Score” score. Test validation in a 800+ sample cohort demonstrated that it is useful in both node negative and positive patients, as well as chemotherapy naïve and treated patients. CanAssist-Breast Score, is a strong independent predictor of disease recurrence by multivariate analysis. The majority of patients in ‘low risk’ had Stage 2, Grade 2/3 disease over Stage 1, Grade 1 disease. Comparison with commonly used prognostic tools including Ki67, the online tool PREDICT and Oncotype Dx showed that CanAssist-Breast test was superior in determining prognosis.

Conclusions

CanAssist-Breast is a low-cost, prognostic and chemotherapy predictive test to predict risk of recurrence and enable optimal treatment planning in patients with early stage Breast Cancer in Asia.

Legal entity responsible for the study

DCGI registered Ethical Committee based in Bangalore, India.

Funding

Onco Stem Diagnostics Private Limited

Disclosure

M.M. Bakre: OncoStem Diagnostics is start-up biotechnology company privately funded by venture capitalist. The retrospective, non-interventional, observational study was approved by DCGI registered Ethical Committee based in Bangalore, India. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1676P - Comparison of progression-free survival (PFS) on comprehensive multiplatform profiling-guided therapy to PFS on prior therapy: A pooled analysis from 4 contemporary prospective studies (ID 1309)

Presentation Number
1676P
Lecture Time
13:15 - 13:15
Speakers
  • A. Seeber
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

It is expected that the progression-free survival (PFS) for patients with refractory cancers will decline over subsequent lines of therapy. Patients with refractory metastatic cancer have previously been shown to derive some clinical benefit from comprehensive multiplatform profiling (CMP) of tumor tissue. Data from four independent physician-led prospective and prospective/retrospective studies was pooled in an exploratory manner to determine if PFS was improved when patients were treated with molecular profiling-guided therapies compared to PFS on the prior therapies.

Methods

Tumor tissue specimens from 202 patients were submitted for CMP to a certified referral laboratory (Caris Life Sciences, USA) between March 2010 and December 2016. Treatment selections were based on predictive biomarker status associated with agents with potential clinical benefit. Clinical benefit was defined as a PFS ratio (= PFS upon treatment according to CMP/PFS on the prior therapy) ≥ 1.3.

Results

As of December 2016, 157 of 202 (77.8%) profiled patients were treated according to the predictive results, of whom 140 were evaluable. Patients had received a median of three prior therapies (range 1-12). The most common tumor types were breast (n = 35), colorectal (n = 14), non-small cell lung (n = 11) and gastric cancer (n = 9). A median PFS of 120.0 days was observed with CMP-directed therapies compared to 89.5 days for prior therapies (HR = 0.70, p = 0.012). Seventy-three of 140 patients (52%) had a PFS ratio ≥ 1.3. Over 70% of treated patients received chemotherapy alone, while 21% of patients received targeted therapies, either alone or in combination with chemotherapy or hormone therapy.

Conclusions

Contrary to the expected decline in PFS, patients had a better outcome when treated with CMP-guided treatments. This was interestingly driven by the precision use of available chemotherapeutic resources rather than sometimes inaccessible targeted therapies. Further prospective trials in specific tumor types may help to highlight particular patient populations who might benefit most from CMP guidance.

Legal entity responsible for the study

Günther Gastl

Funding

None

Disclosure

A. Seeber: Consultant for Caris Life Sciences All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1677P - Effect of enoxaparin, omeprazole, gemcitabine and bortezomib in refractory patients (ID 4041)

Presentation Number
1677P
Lecture Time
13:15 - 13:15
Speakers
  • J. Marquez-Manriquez
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Repurposing drugs and immunogenic chemotherapy for cancer is an emerging field, especially the combination of drugs with validated data. Several studies have shown that enoxaparin, omeprazole, gemcitabine and bortezomib have immunomodulatory properties that synergize with several chemotherapeutic protocols and decrease chemoresistance in several tumors. We treated refractory patients with ECOG=0 with this combination. We demonstrated significant clinical response that correlated with the immune response after 2 months of weekly treatment.

Methods

CICS IRB approved this protocol and inform of consent was signed. We included 10 patients, median age 45 years old of each tumor with at least 2-4 relapses. The patients receive intravenous 0.5 gr/m2 of gemcitabine, 3.5 mg of bortezomib, 80 mg of omeprazole and enoxaparin was administrated subcutaneously in the area with more tumor activity according with the CT SCAN, Granzyme B ELISPOT and cytokine ELISA that were performed before, during and after the treatment. We analyzed the data with prism graph pad and by multivariate analysis using SAS/STAT.

Results

We had a significant correlation between increased levels of CD8 cells (p = 0.0003) and PFS in the 100% of the patients. The cytokines measured were downregulated after the treatment with significant correlation with IL-6 (p = 0.001), IL-8 (p = 0.001), IL-18, (p = 0.01) and TNF alpha (p = 0.005) and CR after the third CT scans. The laboratory tests before, during and after the treatment did not demonstrate clinical significant toxicity.

Conclusions

The results obtained in this pilot study gave relevant data to prepare a phase I trial. We conclude that this combination is feasible to overcome chemoresistance and improve the anti-tumor immune response by CD8 cells and decreasing cytokines associated with tumor progression.

Clinical trial identification

Does not apply

Legal entity responsible for the study

Centro de Investigacion de cancer en Sonora (CICS) campus Ciudad Obregon, Sonora, Mexico.

Funding

Fundacion del Centro de Investigacion de cancer en Sonora campus Ciudad Obregon

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1678P - A new chemotherapy-based combination to prevent osteosarcoma progression (ID 4487)

Presentation Number
1678P
Lecture Time
13:15 - 13:15
Speakers
  • M. Monchanin
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Despite the intensification of chemotherapy regimen, 5 years survival rates for patients with metastatic or relapsed osteosarcoma (OS) remains of 20%. The secreted factor netrin 1 (Nt1) is overexpressed in many human cancers to block apoptosis. Recent studies showed that blocking Nt1 interaction with its receptors potentiates chemotherapy efficacy suggesting that combining chemotherapies with Nt1 interference could be a promising approach for chemoresistant tumors like OS.

Methods

Analyses of the ATGSarc database (http://atg-sarc.sarcomabcb.org/), indicate that Sarcoma with complex genomic (SCG) with a higher expression of Nt1 have a poorer outcome (p < 0.002). In addition, q PCR performed on human sarcomas samples showed that Nt1 is higher expressed in OS compared to other SCG (7.75 fold increase - p < 0.02). These data indicated that Nt1 could be a potential target for OS treatment. Thus, we evaluated the antitumoral effects of anti Nt1 monoclonal antibody (aNt1) combined to doxorubicin (Dox) in a rat syngeneic and metastatic OS model. In this model, treatments were administered either on progressive OS or post operatively to prevent OS relapse. At the end of the experiments tumors and lung were collected for IHC analyses.

Results

As pre operative treatment, Dox/aNt1 combination caused a marked delay in OS progression (median end point reached at day 17 and day 22 respectively in Dox and Dox/aNt1 group, (p < 0.02) and dramatically slowed down metastatic spreading: lung metastases (d > 5mm) were found respectively in 75% and 17% of Dox and Dox/aNt1 treated rats As post operative treatment, Dox/aNt1combination significantly increased animals survival (median end point reached at day 15 and day 21 respectively in Dox and in Dox/aNt1 group; (p < 0.02). Moreover, 19 days after tumor resection, 10% of the Dox treated tumors hadn’t relapsed versus 40% in the Dox/aNt1 treated group. A variation in tumor vascular density caused by the treatment was found in the Anti Nt1 treated groups as shown by CD146 staining.

Conclusions

Our study reporting the antiproliferative and antimetastatic effects and of Dox/aNt1 Combination in OS indicate that this combined treatment could be a way to overcome OS chemoresistance.

Legal entity responsible for the study

Dutour Aurélie

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1679P - Mutant KIT translocates into the nucleus and induces NFKBIB expression that leads to KIT expression in imatinib-resistant gastrointestinal stromal tumors (ID 3380)

Presentation Number
1679P
Lecture Time
13:15 - 13:15
Speakers
  • Y. Hsueh
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Gastrointestinal stromal tumor (GIST) is a dominantly mutant KIT-driven tumor. Prolonged tyrosine kinase inhibitor (TKI) treatment may result in a resistant phenotype through acquired secondary KIT mutation. Increasing evidences show that membrane-bound receptors, as EGFR, can translocate into the nucleus, mediate genes expression, and lead to tumor survival and drug resistance. However, it’s barely known the nuclear role of KIT in GIST.

Methods

In this study, two imatinib (IM)-resistant GIST cell lines, GIST48 and GIST430, were used as a model.

Results

In this study, we first showed that KIT is distributed both in the cytoplasm and the nucleus in IM-resistant GIST cells. Using ChIP-seq and ChIP assay, we identified that nuclear KIT bound to the NFKBIB promoter region and regulated its expression. The expression levels of NFKBIB and phosopho-KIT were significantly correlated with NCCN-risk category in surgically resected GISTs stained by immunohistochemistry. The cell viabilities were inhibited as accompanying with KIT reduction in GIST cells while NFKBIB was silenced or RELA was overexpressed. Moreover, RELA was activated, translocated into the nucleus, and bound to KIT promoter region in NFKBIB-silenced or RELA-overexpressed GIST cells. Valproic acid, acted as a NFKB inducer, could induce RELA nucleus translocation and binding to KIT promoter region that led to the reduction of protein and RNA expression level of KIT and the cell viabilities of GIST cells. Furthermore, the combination of IM with low-dose valproic acid showed synergistically inhibitory effect on cell viabilities of GIST cells and comparable effects on reducing phospho-KIT level and inhibiting tumor growth as high-dose valproic acid did in GIST430 xenograft model.

Conclusions

Taken together, we first demonstrated that phosphorylated KIT could translocate into the nucleus and drive itself expression in IM-resistant GIST cells through mediating NFKBIB expression. In addition, our findings identified a novel and druggable KIT-NFKBIB-NFKB regulatory axis that provides a new insight on tumorigenesis and therapeutic option for IM-resistant, mutant KIT-expressing GISTs.

Legal entity responsible for the study

National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan

Funding

National Health Research Institutes, Taiwan and the Ministry of Science and Technology, Taiwan.

Disclosure

Y-S. Hsueh: Research funds were provided by the Ministry of Science and Technology, Taiwan. L-T. Chen: Research funds were provided by the National Health Research Institutes, Taiwan. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1680P - Reversion of epithelial–mesenchymal transition (EMT) as a mechanism of action of cabazitazel in castration-resistant prostate cancer (ID 5169)

Presentation Number
1680P
Lecture Time
13:15 - 13:15
Speakers
  • N. Jiménez
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The epithelial to mesenchymal transition (EMT) process is involved in de novo and acquired resistance to hormone-therapy and docetaxel (D) in metastatic castration resistant prostate cancer (mCRPC). Cabazitaxel (CZ) it active after D-progression and prior second-line hormone-therapies. Here we investigated the differences between CZ and D resistance related to the EMT phenotype acquisition and its potential clinical value.

Methods

D and CZ resistant (R) cells lines were derived from parental DU-145 and PC-3. Cell line Molecular characterization was performed using Affymetrix GeneChip Human Gene 2.0 ST microarrays. Gene expression analysis was performed by quantitative real-time PCR in cell lines and in FFPE tumors from mCRPC treated with CZ. Protein levels were measured by Western Blot. Cell migration was assessed using the Cultrex cell migration kit (Trevigen) andcell viability by MTT assay. Gene inhibitory experiments were performed by siRNA transfection.

Results

Microarray data, pathway analysis and EMT gene data in silico validation showed that EMT occurred in both D-R and CZ-R cells, being ZEB1 one of the top deregulated genes. However, we identified 55 EMT genes differentially deregulated between D-R and CZ-R vs parental cells. Among them CDH1, and ESRP1 (lost in D-R but maintained in Cz-R), and AXL (overexpressed in D-R and downregulated in CZ-R). D-R cells presented a more pronounced mesenchymal phenotype (morphology, higher migration and lower proliferation rates, higher expression of EMT markers at mRNA and protein level) than CZ-R. Dose-response experiments showed that CZ induced CDH1 and ESRP1 expression in different cell lines models. ZEB1 inhibition reverted D- resistance, but not CZ-resistance, and restored ESRP1 expression in D-R cells. In 29 CRPC patients treated with CZ, low level of expression of ESRP-1 in tumor correlated with a better PSA-PFS (6.2 vs 2.7 months, P = 0.006; HR: 0.31 P = 0.009) and radiological PFS (7.9 vs 3.3 months, P = 0.047; HR: 0.39 P = 0.055) and the EMT phenotype was not associated to resistance.

Conclusions

The reversion of EMT phenotype, trough induction of CDH1 and ESRP1, may be a novel mechanism of action of CZ, which may explain its activity in patients progressing to prior therapies in CPRC.

Legal entity responsible for the study

Hospital Clínic of Barcelona

Funding

Sanofi-Aventis

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1681P - Globally optimizing therapeutic combinations against bortezomib-resistant multiple myeloma using a quantitative parabolic optimization platform (QPOP) (ID 1166)

Presentation Number
1681P
Lecture Time
13:15 - 13:15
Speakers
  • M. Rashid
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Multiple myeloma is an incurable hematological malignancy that relies on drug combinations as first and secondary lines of treatment. The inclusion of proteasome inhibitors, such as bortezomib, into these drug combination regimens has improved median survival. Resistance to bortezomib, however, is a common occurrence that ultimately contributes to treatment failure. Thus, there remains a need to identify improved drug combinations that may serve as later lines of treatment.

Methods

We have developed the quantitative parabolic optimization platform (QPOP) to optimize drug combinations against bortezomib-resistant multiple myeloma. By mapping phenotypic output data to parabolic response surfaces, QPOP is able to deterministically optimize drug combinations as well as drug dosages.

Results

We have successfully identified potential optimal drug combinations against bortezomib-resistant RPMI 8226 (P100v) cell line as projected via QPOP, with these combinations exhibiting synergistic response surface maps. The drug combinations displayed lower half-maximal inhibitory concentrations (IC50) in vitro as compared to single drug administration. While QPOP does not rely on molecular mechanism prediction, the identified optimal drug combinations can reverse DNA hypermethylation and silencing of tumor suppressors that occurs following acquired bortezomib-resistance. Prolonged survival of P100v tumor-bearing mice was observed when these optimized combinations were validated in vivo, further highlighting the importance of treating in vitro and in vivo as two separate entities. Moreover, the drug combination is broadly effective across a range of primary multiple myeloma patient samples.

Conclusions

These results collectively show that QPOP is a robust platform that is able to eradicate the bortezomib-resistant clones of multiple myeloma. Beyond bortezomib-resistant multiple myeloma, global optimization of drug combinations by QPOP can serve to improve drug combination design across a range of other cancers and diseases through a continuous optimization process across the entire drug development pipeline.

Legal entity responsible for the study

Masturah Rashid

Funding

National Research Foundation Cancer Science Institute of Singapore RCE Main Grant

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1682P - Compartmentalized activities of the pyruvate dehydrogenase complex sustain lipogenesis in prostate cancer (ID 3205)

Presentation Number
1682P
Lecture Time
13:15 - 13:15
Speakers
  • J. Chen
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Metabolism in cancer serves to provide energy and key biomolecules that sustain cell growth, a process that is frequently accompanied by decreased mitochondrial use of glucose. Importantly, metabolic intermediates including mitochondrial metabolites are central substrates for post-translational modifications at the core of cellular signalling and epigenetics. However, the molecular means that coordinate the use of mitochondrial metabolites for anabolism and nuclear protein modification are poorly understood.

Methods

We constructed prostate specific Pten; Pdha1 double knockout mice by crossing transgenic mice with flox elements flanking exon 8 of Pten gene and exon 4 and exon 5 of Pdha1 gene. Gene expression profiling analysis and metabolic analysis between Pten; Pdha1 double knockout and Pten knockout tumours were performed to investigate the metabolic pathways altered upon Pdha1 inactivation. Lipidomics analysis between these two genotypes of tumours were performed to reveal the difference on the lipid and cholesterol ester species in response to Pdha1 inactivation.

Results

We found that genetic and pharmacological inactivation of Pyruvate Dehydrogenase A1 (PDHA1), a subunit of pyruvate dehydrogenase complex (PDC) that regulates mitochondrial metabolism inhibits prostate cancer development in different mouse and human xenograft tumour models. Intriguingly, we found that lipid biosynthesis was strongly affected in prostate tumours upon PDC inactivation. Mechanistically, we found that nuclear PDC controls the expression of Sterol regulatory element-binding transcription factor (SREBF) target genes by mediating histone acetylation whereas mitochondrial PDC provides cytosolic citrate for lipid synthesis in a coordinated effort to sustain anabolism. In line with the oncogenic function of PDC in prostate cancer, we find that PDHA1 and the PDC activator, Pyruvate dehydrogenase phospatase 1 (PDP1), are frequently amplified and overexpressed at gene and protein level in these tumours.

Conclusions

Taken together, our findings demonstrate that mitochondrial and nuclear PDC sustains prostate tumourigenesis by controlling lipid biosynthesis thereby pointing at this complex as a novel target for cancer therapy.

Legal entity responsible for the study

Molecular Oncology, Institute of Oncology Research

Funding

IBSA foundation

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1683P - Metabolomics in cancer cachexia (ID 4807)

Presentation Number
1683P
Lecture Time
13:15 - 13:15
Speakers
  • J. Lopez-Martin
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Cancer cachexia (CC) is a frequent unmet medical need. CC affects up to 80% of cancer patients, and it is indirectly responsible for at least 20% of cancer deaths. The pathophysiology is characterized by a variable combination of reduced food intake and abnormal metabolism, including systemic inflammation and negative protein and energy balance. Despite its high clinical significance, definite diagnostic criteria of cachexia are lacking. The 'omics' technologies provide a global view of biological systems. Among these, blood-based metabolomics is a promising method for cachexia study.

Methods

This study is part of a pilot, observational, cross-sectional, case-control, hypothesis generating, IRB-reviewed, research project. Objective: discovery and selection of biomarkers of cancer cachexia. Anthropometric, clinical and biochemical data from consenting eligible cancer patients were collected. Plasma proteome was assessed by 2D gel electrophoresis and MALDITOF mass spectrometry. Metabolomics was evaluated by means of a multiplatform non-targeted approach of plasma samples (LC –MS, GC-MS and CE-MS), to increase metabolite coverage. Data were analysed by univariate and multivariate methods, using principal component analyses and error adjustments for multiple comparisons.

Results

from metabolomics study are shown. Subjects: 15 cancer (ca) patients (pts), distributed as follows: Cachexia (CX): 8 pts (male:female 7:1; pancreatic ca: 3, melanoma: 3, biliary duct ca: 2); control (CN): 7 pts (M:F 6:1; colon ca: 1, esophageal ca: 1, gastric ca: 2, pancreatic ca: 1, melanoma: 1, sarcoma:1). Median age: CX 62 y (36-81), CN 64 y (48-80); median body mass index: CX 20 Kg/m2 (17-27), CN 25 Kg/m2 (21-27); median albumin: CX 3.4 mg/dL (2.1-4.1), CN 3.9 mg/dL (3.5-4.8). A total of 89 metabolites (Mbl) were significantly altered in CX pts. The Mbl with highest increase was cortisol (fold change 1.67, p = 0.03). The largest affected group of Mbl was 'amino acids and derivatives', all decreased. Glycerophospholipids, sphingolipids, steroid derivatives, fatty acids, aldehydes, phenylacetamides, carboxilic acids and derivatives, and indoles were also decreased.

Conclusions

These finding suggest that plasma amino acids and lipids profiling has great potential for improving cachexia cancer screening and diagnosis, and understanding disease pathogenesis. Of note, the increased values of cortisol should lead us to revisit the use of glucocorticoids in this setting. Substitutive therapy for some of the observed deficiencies might deserve clinical exploration.

Legal entity responsible for the study

Instituto de Investigación Sanitaria Hospital 12 de Octubre

Funding

ISCIII-FEDER (PI10/2072) and Fundacion Mutua Madrileña.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1684P - Association between the Dietary Inflammatory Index (DII), urinary enterolignans and C-reactive protein in the National Health and Nutrition Examination Survey-2003-2008 (ID 1849)

Presentation Number
1684P
Lecture Time
13:15 - 13:15
Speakers
  • N. Shivappa
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Enterolignans are important biomarkers of microbiome diversity. Higher levels, indicating greater diversity, have been shown to reduce cancer risk. Diet and inflammation have been shown to play a role in maintaining microbiome diversity. This study examined whether inflammatory potential of diet, as measured by the Dietary Inflammatory IndexTM (DII) has an impact on levels of urinary enterolignans in the National Health and Nutrition Examination Survey (NHANES) 2003-2008. We also carried out validation of the DII with C-reactive protein (CRP).

Methods

Data came from NHANES 2003-2008. Enterolignans (enterodiol and enterolactone) and CRP were assayed from urine and serum specimens, respectively. DII scores were calculated from food intakes assessed using 24-hour dietary recalls and expressed per 1,000 calories consumed. Associations were examined using survey-based multivariable linear and logistic regression.

Results

After adjustment, higher DII scores (i.e., relatively more pro inflammatory) were associated with lower levels of creatinine normalized enterodiol (bDIIquartile4vs1 = -1.22; 95% CI = -0.69, -1.74; Ptrend = <0.001) and enterolactone (bDIIquartile4vs1 = -7.80; 95% CI = -5.33, -10.26; Ptrend = <0.001). A positive association also was observed when enterolignans were dichotomized at the 90th percentile value. In this same sample DII scores also were associated with CRP ≥3mg/l (ORDIIcontinuous=1.12; 95% CI = 1.05, 1.19).

Conclusions

In these NHANES data, there was an association between DII and enterolignans. This study also provided a successful construct validation of the DII using CRP in a nationally representative sample. Using enterolignans as a proxy for gut microbiome, these results indicate that diet-associated inflammation modifies gut diversity.

Legal entity responsible for the study

University of South Carolina

Funding

NIH.

Disclosure

N. Shivappa, M. Wirth, J. Hebert: Dr. James R. Hébert owns controlling interest in Connecting Health Innovations LLC (CHI), a company planning to license the right to his invention of the dietary inflammatory index (DII). Drs. Nitin Shivappa and Michael Wirth are employees of CHI. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1685P - Fullerenol/iron nanocomposite modulates doxorubicin-induced cardiotoxicity (ID 4598)

Presentation Number
1685P
Lecture Time
13:15 - 13:15
Speakers
  • M. Seke
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Doxorubicin is a first line cancer chemotherapeutic. Unfortunately, its clinical use is limited by its cardiotoxicity. It is known that iron overload aggravates anthracycline toxicity. Fullerenol is a 1 nm size molecule and in aqueous solutions is in the form of polyanionic nanoparticles, which enables them to serve as a good carrier of positively charged ions such as Fe2+. Fullerenol’s antioxidant activity through scavenging free radicals has already been proved in different biological systems.

Methods

The aim of our study was to investigate the effects of the fullerenol/iron nanocomposite as a pretreatment to doxorubicin on the rat’s heart in comparison to doxorubicin alone. After the 24h-treatment, adult male Wistar rats were sacrificed and hearts were collected for ultrastructural and qRT-PCR analysis. Considering the ability of doxorubicin to induce oxidative stress, and the fullerenol’s capability to mitigate it, we had chosen to monitor gene expression of enzymes involved in antioxidant defense.

Results

Ultrastructural study revealed that in the group pretreated with the nanocomposite prior to doxorubicin application cardiomyocytes were with preserved morphology and the structure of intercalated discs. On the other hand, the heart tissues of animals treated with doxorubicin alone were significantly more damaged. Intensive interstitial edema was observed, as well as vacuolization of cardiomyocytes, hypercontraction of sarcomeres, mitochondria of irregular shapes. qRT-PCR results have shown that neither treatment with doxorubicin alone nor the pretreatment with the nanocomposite did cause significant increase in mRNA levels of catalase and superoxide dismutase.

Conclusions

Our results indicate that the fullerenol/iron nanocomposite applied as pretreatment to doxorubicin induces less damage to the hearth tissue in comparison to doxorubicin alone.

Legal entity responsible for the study

Aleksandar Djordjevic

Funding

Ministry of Education, Science and Technological Development, Republic of Serbia, Grant No. III 45005.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1686P - Ability of TMPRSS2-ERG (TE) expression to predict taxane benefit depending on prior abiraterone or enzalutamide therapy in castration-resistant prostate cancer (ID 4499)

Presentation Number
1686P
Lecture Time
13:15 - 13:15
Speakers
  • M. Marín-Aguilera
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

TMPRSS2-ERG (TE) results in androgen-driven overexpression of ERG, which is involved in resistance to taxanes in preclinical models. In prior work we showed that TE expression in blood correlated with taxanes resistance in metastatic castration-resistant prostate cancer (mCPRC). Here, we studied if the detection of TE in primary tumors predicts taxanes activity in CPRC. We also explored the impact of prior abiraterone or enzalutamide (A/E) in blood TE detection and in TE predictive value.

Methods

mCRPC patients (pts) treated with taxanes in a multicenter biomarker study were included. Formalin-fixed paraffin-embedded (FFPE) tumors and peripheral blood mononuclear cells (PBMCs) fraction were tested for TE presence by RT-qPCR. FFPE were retrospectively obtained. PBMCs were prospectively collected prior to taxane initiation. PSA-PFS was evaluated by Kaplan-Meier analysis using log-rank test. Univariate analysis of TE status (+ vs-) was performed with Cox regression.

Results

124 pts were included: 111 (89.5%) received docetaxel (Dx), 13 (10.5%) cabazitaxel (Cz) and 27 (21.8%) both. Fifty-seven (45.9%) tumors were TE+. Overall, no correlation between tumor TE expression and taxane benefit was observed in the whole population, or in the Dx or Cz group separately. However, in Dx-treated pts without prior A/E (N = 80, 72.1%), tumor TE+ correlated with lower PSA-PFS (median 8.6 vs 13.6 months; HR 1.7, p ≤ 0.05). No differences were observed in Dx treated pts with prior A/E (N = 31, 27.9%) according to tumor TE expression. In 44 pts, matched tumor and PBMC samples were available. Concordance between tumor an blood was 92.8% and 63.3% for pts with and without prior A/E, respectively. TE in blood was + in 1 (7%) pts with prior A/E and in 7 (23.3%) pts without prior A/E. As observed in FFPE samples, in patients without prior A/E to Dx (N = 28; 63.6%), blood TE+ correlated with lower PSA response (0% vs 61.9%, p ≤ 0.01) and reduced median PSA-PFS (3.34 vs 8.2 mM; HR 4.1 p ≤ 0.01).

Conclusions

The predictive value of TE in taxane resistance may be different depending on prior exposure to A/E. This is being tested in a multicenter prospective study.

Legal entity responsible for the study

Hospital Clínic of Barcelona/Institut d'Investigacions Biomèdiques Agust Pi i Sunyer

Funding

None

Disclosure

A. González del Alba: Advisory boards: Sanofi, Janssen, Astellas, Bayer Travel expenses: Astellas, Sanofi, Janssen. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1688P - Identification of patient population with longer survival when treated with S-1 plus cisplatin via predictive enrichment strategy analysis of the FLAGS and DIGEST phase III trial (ID 1434)

Presentation Number
1688P
Lecture Time
13:15 - 13:15
Speakers
  • M. Takeuchi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The FLAGS trial, a randomized phase III trial, compared S-1, an oral fluoropyrimidine, plus cisplatin (SP) with 5-fluorouracil plus cisplatin (FP) in the first-line treatment for advanced gastric cancer (AGC). The results led to the approval of SP by EMA, and it is now marketed in Europe. The purpose of this analysis was to establish a clinical covariate(s) model using the Predictive Enrichment Strategy Analysis (PESA) identifying patients who benefit from SP.

Methods

PESA is a new robust methodology with guidelines by the United States Food and Drug Administration. Consensus-based 15 clinical covariates were selected for PESA and a large cohort with no missing data (FLAGS trial: 889 patients (pts)) was analyzed. The models generated were cross-validated and the results analyzed were validated in the DIGEST trial, a phase III trial comparing SP to FP in diffuse type advanced gastric cancer. From the DIGEST trial, 333 patients and 14 clinical covariates were used in the analysis.

Results

In FLAGS, ECOG Performance status (PS = 1) was the strongest covariate in the enrichment group showing benefit for SP. In the population with PS = 1, the OS in the SP group was significantly longer than the FP group (Hazard Ratio [HR]=0.798, 95%CI = (0.66-0.96) p = 0.0166). Other covariates with high potential to be associated with SP benefit included: diffuse-type histology, positive peritoneal metastases, and the lack of liver metastases. In DIGEST PS = 1 also showed to be most associated with SP benefit. While there was no strong signal from the variables positive peritoneal metastases, and the lack of liver metastases, there appeared to be a signal from the neutrophil variable. In the DIGEST population of diffuse type, patients with PS = 1 and low baseline neutrophil count may benefit from SP.

Conclusions

Presence of PS = 1 was associated with SP benefit in both the FLAGS and DIGEST trial. Although peritoneal and liver metastases resulted in slightly different signals in the trials, further analyses will be done to look at the impact of low baseline neutrophil count on the benefit of SP for PS = 1 and diffuse type histology patients.

Clinical trial identification

FLAGS trial; ClinicalTrials.gov NCT00400179 First received: June 30, 2005 Last updated: March 28, 2012 Last verified: March 2012 DIGEST trial; ClinicalTrials.gov NCT01285557 First received: January 26, 2011 Last updated: October 19, 2016 Last verified: October 2016.

Legal entity responsible for the study

Taiho Pharmaceutical Co,. Ltd.

Funding

Taiho Pharmaceutical Co,. Ltd.

Disclosure

M. Takeuchi: Consulting Taiho Pharmaceutical Co., Ltd. Travel, Accommodations, expenses AnGes MG, Inc. M. Takeuchi: Honoraria: Shionogi & Co., Ltd. Consulting or advisory role: Taiho pharmeceutical Co., Ltd.; Hisamitsu Pharmaceutical Co., Inc.; AstraZeneca K.K.; AbbVie Inc. J.A. Ajani: Honoraria: Bristol-Myers Squibb; Five Prime Therapeutics, Inc.; Medscape, LLC.; Celegene, Taiho pharmaceutical Co., Ltd. Research funding Bristol-Myers Squibb; Merck; Taiho pharmaceutical Co., Ltd.; Delta-Fly Pharma, Inc; Gilead Sciences Inc.

Collapse
Poster display session Poster Display session

1689P - HMGA1 is a new biomarker of liposarcoma progression (ID 2241)

Presentation Number
1689P
Lecture Time
13:15 - 13:15
Speakers
  • R. Loria
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Liposarcoma (LPS) is the most common type of soft-tissue sarcoma that includes a heterogeneous class of tumors classified according to histologic appearances, protein expression pattern and molecular genetics. Molecular subtyping is not only important for accurate diagnosis but may be necessary as a basis for the identification of therapeutic targets. Lipoma is characterized by extensive High Mobility Group A1 (HMGA1) protein aberrations suggesting a role of this protein in the mechanisms of liposarcoma progression as well as previously demonstrated in other tumors.

Methods

Cell lines derived from different liposarcoma subtypes and a cohort of 68 patients were used to analyze in vitro and in vivo the role of HMGA1 in liposarcoma progression.

Results

Our data revealed that HMGA1 is highly expressed in liposarcoma cell lines and that is strongly involved in the mechanism of cell proliferation, mobility and invasion of this subtype of tumor. The in vitro results were confirmed in vivo by the RT-PCR and IHC analyses of 68 specimens of different subtypes of liposarcoma derived from patients surgically treated at Regina Elena National Cancer Institute. The aggressive subtypes de-differentiated and myxoid liposarcoma showed higher HMGA1 levels than well-differentiated liposarcoma. Furthermore, trabectedin, a marine alkaloid isolated from the tunicate Ecteinascidia turbinata, down-regulates HMGA1 and E2F1, as well as its downstream targets Vimentin and ZEB1 in sensitive myxoid liposarcoma cells, suggesting a critical role of the transcriptional complex HMGA1/E2F1 in the regulation of the mesenchymal compartment. These data were further confirmed in vivo by the IHC analysis of myxoid sarcoma specimens derived from patients that received trabectedin therapy before surgery. On the other hand, trabectedin treatment down-regulates the activity of HER3 receptor that in turn inhibits Nf-kB pathway in sensitive myxoid liposarcoma cells but not in resistant counterpart cells demonstrating that the activation of Nf-kB pathway is involved in the mechanisms of drug resistance.

Conclusions

Overall, our data suggest that HMGA1 may represent a new biomarker of liposarcoma progression and that it could be a new potential therapeutic target for the more aggressive liposarcoma subtypes.

Legal entity responsible for the study

Regina Elena National Cancer Institute

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1690P - hsa_circ_0004870 is related to AR-V7 expression and may confer resistance to enzalutamide in castration-resistant prostate cancer (ID 1279)

Presentation Number
1690P
Lecture Time
13:15 - 13:15
Speakers
  • J. Greene
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Androgen deprivation therapy is the mainstay of prostate cancer treatment, however, resistance inevitably develops, resulting in a more aggressive disease, known as castration-resistant prostate cancer (CRPC). While, enzalutamide provides a substantial survival benefit by targeting the AR, it is not curative and many patients develop resistance to therapy. Although not yet fully understood, resistance can develop through multiple mechanisms, such as AR copy number gain or the generation of splice variants such as AR-V7. circular RNAs (circRNA) are a type of non-coding RNA that have an important function in gene regulation and may play a role in drug resistance, through the regulation of miRNA circRNAs are tissue specific, stable and may represent a novel marker of drug resistance in PCa.

Methods

circRNA profiling was performed on an isogenic PCa cell line model consisting of enzalutamide sensitive and resistant subtypes using a high throughput microarray assay. Subsequently, bioinformatic analyses predicted five miRNA binding sites (miRNA Response Elements) for each circRNA and these were stratified based on known associations with PCa. Targets were validated using qPCR.

Results

circRNAs were more often downregulated in resistant cell lines compared to sensitive lines (588 versus 278). hsa_circ_0004870 was significantly downregulated in enzalutamide resistant cells compared with control. RBM39 was determined as the parental gene, which encodes a member of the U2AF65 family of proteins. Previous studies have shown that, U2AF65 results in the expression of AR-V7, by binding to AR pre-mRNA. Expression of all genes were confirmed within our enzalutamide model.

Conclusions

hsa_circ_0004870 is linked to the generation of AR-V7 and may play a key role in the development of enzalutamide resistance in CRPC via a miRNA mediated mechanism.

Legal entity responsible for the study

Trinity College Dublin

Funding

Irish Cancer Society

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1691P - Analysis of DPYD and UGT1A1 genotype in patients with advanced pancreatic cancer treated with modified FOLFIRINOX (ID 3775)

Presentation Number
1691P
Lecture Time
13:15 - 13:15
Speakers
  • C. Vivaldi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Modified FOLFIRINOX (mFOLFIRINOX) is a standard treatment in advanced pancreatic cancer (aPC). Because of the presence of either loss-of-function mutations in DPYD (c.1679T>G, IVS14 + 1G>A, c.2194G>A, c.2846A>T) or UGT1A1*28 variant associated with reduced UGT1A1 expression, deficiency of DPD and UGT may result in drug accumulation and severe toxicities caused by fluoropyrimidines and irinotecan, respectively.

Methods

The present study analyzes the association between DPYD and UGT variants and adverse drug reactions (ADRs) in aPC patients (pts) treated with mFOLFIRINOX. Blood samples were collected from 104 pts, and analyses of DPYD c.1679T>G, IVS14 + 1G>A, c.2194G>A, c.2846A>T and UGT1A1*28 were performed by automatic sequencing. Statistical analysis was performed by chi-square, Mann-Whitney and Spearman's rho tests on SPSS v.23s.

Results

None of the pts was carrier of the c.1679G and c.2846T alleles. Only one IVS14 + 1GA was found and 8 pts had c.2194GA genotype. ADRs grade (G) ≥3 were neutropenia (42.3%), diarrhea (7.7%) and stomatitis (7.7%). The statistical analysis of the IVS14 + 1GA has not been performed due to the extremely low frequency of the mutant allele (0.96%), however IVS14 + 1GA patient experienced G4 hematological and gastrointestinal ADRs after the first cycle. We observed a trend toward significant association between c.2194GA genotype and the risk of thrombocytopenia (p = 0.080) and hand-foot syndrome (HFS) (p = 0.096). The UGT1A1*28 allele was found in 56 (54.4%) pts (*1/*28, n = 38; *28/*28, n = 18) and it was correlated with the risk of developing thrombocytopenia (p = 0.006) and neutropenia (p = 0.044). Moreover, this risk increased as the number of *28 alleles increased (*28/*28 > *1/*28 > *1/*1, p = 0.003). No significant correlation with diarrhea was found.

Conclusions

Our data confirm that DPYD IVS14 + 1A is associated with life-threatening toxicities and that the c.2194A allele could be possibly associated with thrombocytopenia and HFS, but validation in larger cohorts is needed. UGT1A1*28 allele is associated with a higher risk of G3/4 thrombocytopenia and neutropenia, and should be implemented in routine practice to personalize treatment in aPC.

Legal entity responsible for the study

University of Pisa

Funding

Institutional fundings

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1692P - Clinical significance of the expression of membrane receptors of the alternative nuclear factor-kappaB pathway in non-small cell lung cancer (ID 4553)

Presentation Number
1692P
Lecture Time
13:15 - 13:15
Speakers
  • F. Dimitrakopoulos
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

During the last decade, the alternative pathway of Nuclear Factor-kappaB (NF-κB) has gained importance due to its implication in cancer initiation and development where it has been shown to be deregulated. It is mainly activated thgough the membrane receptors Lymphotoxin β Receptor (LTβR), CD40, B-cell activating factor receptor (BAFFR) and Receptor Activator of NF-κB (RANK), having an important role in immune response and multiple cancer cell functions.

Methods

Immunohistochemical analysis of the expression of these 4 receptors was performed on 130 tumour and adjacent non neoplastic formalin fixed and paraffin embedded tissue samples from patients with non-small cell lung cancer (NSCLC).

Results

CD40 and BAFFR expression was higher in neoplastic compared to adjacent non-neoplastic tissue (P = 0.006 and 0.001, respectively) while no such differences were observed for RANK and LTβR. Moreover, CD40 levels in tumour infiltrating lymphocytes (TILs) correlated with development of metastases in adrenals (P = 0.003), liver (P < 0.001) and in brain (P = 0.048), while CD40 levels in stromal cells correlated with liver metastasis (P = 0.013). Cytoplasmic BAFFR expression in cancer cells was associated with T status while BAFFR levels in stromal cells were related to 2-year survival (P = 0.034). Cytoplasmic RANK expression was associated with membrane levels in cancer cells (P < 0.001) but was independent of any clinicopathological characteristics. Finally, nuclear detection of LTβR was related to histological subtypes with squamous cell carcinoma having higher levels cmpared to adenocarcinomas (P = 0.026).

Conclusions

Protein levels of CD40 and BAFFR are altered in NSCLC in agreement with a deregulation of the alternative NF-κB pathway previously shown by our team. CD40, BAFFR and LTβR tissue protein levels appear to constitue biomarkers for specific clinicopathologcal parameters including survival, stage and histological subtype.

Legal entity responsible for the study

Haralabos P Kalofonos.

Funding

Hellenic Society of Medical Oncology (HeSMO).

Disclosure

T. Makatsoris: Travel expenses from Pfizer, Roche and Astellas. Advisory fees from Roche and Boeringer. Honoraria from Roche, Sanofi, Merck and Amgen. H. Kalofonos: Consulting/advisory role: Roche, Novartis, MSD, Genesis, Pfizer, Lilly, Leo, Amgen, Janssen, Merck, Merck-Serono.Research funding Roche Pfizer, Novartis, Amgen, Bayer, Genesis Lilly, MSD, Janssen, Merck-Serono. Travel expenses: Roche, Novartis, Enorasis, Pfizer. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1693P - Development of TP53 signature diagnostic system using multiplex RT-PCR and observational study to confirm the prognostic value of TP53 signature in breast cancer (ID 2633)

Presentation Number
1693P
Lecture Time
13:15 - 13:15
Speakers
  • S. Takahashi
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The structural mutation status of TP53 gene is well known independent prognostic factor of breast cancer. We have reported that status of the TP53 mutation is predictable by expression profile of 33 genes (‘TP53 signature’) in breast cancer. The TP53 signature was reported to be one of the best predictors of prognosis and therapeutic effect by meta-analysis (BMC Cancer, 2015). The aim of this study is to develop a simple diagnostic system for TP53 signature using multiplex RT–PCR and confirm the prognostic value of TP53 signature.

Methods

We made the multiplex RT-PCR system consists of 26 genes, 23 genes from theTP53 predictive genes and 3 internal control genes. TP53 signature status was determined by the ratio of the sum of expression levels of 16 genes that were upregulated in tumors with TP53 mutation to the sum of expression values of 7 genes downregulated in tumors withTP53 mutation. Cutoff value was set at 1.11 to maximize the sensitivity to detect the TP53 mutant signature. Using a 217 breast cancer case cohort, which was prospectively collected from 2007 to 2010, the relationship between the TP53 signature status and clinicopathological features and TP53 structural mutations were analyzed. And we validated the prognostic value of TP53 signature in 191 stage I-II patients.

Results

Of 217 patients, 102 patients were assigned to the TP53 mutant signature. TP53 structural mutation was observed in 35.1% of patients with TP53 mutant signature and 6.3% of patients with TP53 wild-type signature. In 191 stage I-II patients, RFS of the patients withTP53 mutant signature showed significantly shorter than the patients with wild-type signature. Similar result was observed in 164 ER positive patients. In both univariate and multivariate analyses, TP53 signature status showed independent and better correlation to RFS than tumor size, LN status, stage, ER status and TP53 structural mutation status in stage I-II patients.

Conclusions

We developed the diagnostic system to determine TP53 signature status using multiplex RT-PCR. The TP53 status diagnosed by this system could be one of the prognostic biomarker of breast cancer.

Clinical trial identification

UMIN000005172.

Legal entity responsible for the study

Ethics Committee at the Tohoku University Hospital.

Funding

The Grants-in-Aid from the Ministry of Education, Culture, Sports, Science, and Technology of Japan.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1694P - Expression of estrogen receptors and beta-III tubulin in non-small cell lung cancer tissue (ID 4762)

Presentation Number
1694P
Lecture Time
13:15 - 13:15
Speakers
  • E. Ponomarenko
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Estrogen receptors beta (ERβ) are highly expressed in different normal and neoplastic tissues that, until recently, has been considered to be ER-negative based on ERα expression evaluation. Among the genes, regulated via estrogen signaling there is one, coding microtubule protein beta-III tubulin (TUBB3). TUBB3 expression is found in many solid tumors and is linked to poor prognosis and resistance to taxanes. Since it is little known about mechanisms behind TUBB3 expression in non-small cell lung cancer (NSCLC), we decided to find out if there is a correlation between ER and TUBB3 expression in this type of cancer.

Methods

104 surgical samples of NSCLC were converted to single-cell suspension, stained with primary anti-ERα (abSP-1), anti-ERβ (ab14C8), anti-TUBB3 (ab7751) antibodies and secondary fluorescent antibodies. Immunofluorescent estimation was performed using flow cytometry. Expression level was determined as the ratio (%) of specifically fluorescent cells to the number of cells stained with secondary antibodies. Spearman rank correlation was used to test the association between variables.

Results

Both ER were revealed in all NSCLC specimens. Mean expression level of ERβ was significantly higher compared with ERα (46,6 ± 17,0% vs 23,2 ± 14,2%, respectively). Mean TUBB3 expression level was 43,1±15,7%. In all the tumors investigated only weak correlation observed between and ER status and TUBB3 expression level (rs = 0,3 and rs = 0,4 for ERα and ERβ, respectively). In the group of squamous cell cancer specimens (n = 68) the association was strong (rs = 0,5 and rs = 0,5 for ERα and ERβ, respectively). In the group of adenocarcinoma specimens (n = 36) the correlation between ERα and TUBB3 was very weak (rs = 0,3) and there was no correlation between ERβ and TUBB3.

Conclusions

1. Strong correlation between TUBB3 and ER expression was found only in squamous cell cancer tissue. 2. The dominant type of estrogen receptors is ERβ. 3. In clinical terms high ERβ expression means that in case of resistance to standart platinum/taxane duplets, patients with high tumor ERβ expression may benefit from antiestrogen therapy. Supported by RFBR grants (№№15-04-06991-а, 16-34-01049-mol-a) and grant of the President of RF МК-7709.2016.7.

Legal entity responsible for the study

N.N. Blokhin Russian Cancer Research Center.

Funding

Russian Foundation for Basic Research Grants, grant of the President of Russian Federation.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1695P - Deciphering the antitumor efficacy and mechanistic delineation of epigenetic inhibitors in AML using patient tumor derived ex vivo phenotypic assay based platform (ID 4609)

Presentation Number
1695P
Lecture Time
13:15 - 13:15
Speakers
  • P. Majumder
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Epigenetic inhibitors have demonstrated tumor efficacy by modulating genes involved in growth, proliferation, and invasiveness in hematological malignancies like AML. Preclinical evidences suggest therapeutic benefit by combining epigenetic drugs along with other therapeutics like JAK2 inhibitors. However, there is a huge unmet need to understand the disparities in response at the individual patient level.

Methods

We developed a novel functional assay based platform called CANscript™ to predict the efficacy of anticancer drugs in clinic, which mimics patient tumor microenvironment (Majumder B et al., Nature Communications, 2015). Utilizing samples from AML patients we interrogated response to HDAC and DNA MTase inhibitors by assessing tumor viability, proliferation, morphology, and death in this platform. To elucidate the mechanisms of response, we delineated the pharmacodynamic and pathway modulation by immunohistochemistry and mRNA microarray.

Results

Thirty-two AML patients samples were analyzed in this platform. HDAC and DNA MTase blockade resulted antitumor response, which was demonstrated by differential and quantitatively distinct patterns of target engagement. mRNAs and pathway specific protein expression profiling is suggestive of JAK2 pathway deregulation in many of the non-responders. Treatment with JAK2 inhibitor in this cohort led to efficacy in 40% of these non-responders, suggesting the critical role of this pathway. Interestingly, unique JAK2 signatures associated with single agent vs. combination therapy was observed (10%), hinting at functionally distinct mechanisms of antitumor effects at individualized levels.

Conclusions

These findings demonstrate the utility of this ex vivo platform to predict therapeutic response of epigenetic modulators at the individual patient tumor. It also highlights that, within a contextually heterogeneous framework, distinct mechanisms orchestrate response to HDAC and DNA MTase inhibitors as a single agent or in combination with JAK2 inhibitors. Insights gained from these findings can re-shape our strategic thinking of drug selection for the treatment of AML.

Clinical trial identification

Not Applicable

Legal entity responsible for the study

Mitra RxDx

Funding

Mitra RxDx

Disclosure

G. Babu: Independent consultant and full time employee of Kidwai Memorial Institute of Oncology, scientific and clinical advisory board of Mitra Biotech and equity in this organization. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1696P - Proteomics of triple negative breast cancer developing metastases to central nervous system (ID 4799)

Presentation Number
1696P
Lecture Time
13:15 - 13:15
Speakers
  • K. Rojas
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Breast cancer (BC) is the most frequent tumor in woman, representing 20-30% of all malignancies and continues being the first reason of death for cancer in European women. Triple negative (TN) BC present minor survival rates than other BC subtypes. Key reasons for that is the absence of predictive markers of response to current therapy and the absence of targeted therapies. This study aims to identify proteins with predictive value of central Nervous System (CNS) metastases and therapeutic target candidates.

Methods

This is a case-control retrospective study comparing patients (pts) with metastases to CNS vs pts without them after adjuvant treatment. Sample selection included 50 samples. Formalin-fixed, paraffin-embedded samples were retrieved from Hospital 12 de Octubre Biobank. Proteins were quantified by parallel reaction monitoring.

Results

The average age was 55 years (range 25-85). Forty-seven pts (88.67%) had ductal histology and presented high grade tumors (40 pts; 75.47%). Eight women in the case group presented as first distant recurrence CNS (34.80%), local recurrence (3pts, 13.04%), lung (2pt; 8.7%), bone (1pt; 4.34%) and other locations (7pts; 30.38%). In the control group, first distant recurrence occurred locally (6pts; 46.1%), bone (2pts; 15.4%), lung (1pt; 7.7%) and other sites (4pts; 23.1%). Protein expression data was successfully obtained from 50 samples. ISG15 ubiquitin-like modifier, (P05161) was overexpressed in triple negative breast cancer tumors that develop metastases to CNS (p = 0.036) compared to tumors that do not develop these CNS metastases.

Conclusions

TN tumors frequently metastasize to visceral organs, particularly lungs and brain, and are less likely to metastasize to bone. The interferon-stimulated gene 15 ubiquitin-like modifier (ISG15) encodes an IFN-inducible, ubiquitin-like protein. The ISG15 protein is involved in numerous cellular functions, including interferon-induced immune responses and the regulation of cellular protein turnover. Therefore, ISG15 may represent a novel breast tumor marker helpful in selecting pts who will develop CNS metastases. It also should be explored as a therapeutic target in this clinical context.

Legal entity responsible for the study

Biomedica Molecular Medicine SL.

Funding

None.

Disclosure

L. Trilla-Fuertes: Employee of Biomedica Molecular Medicine SL. A. Gámez, J.A. Fresno: Shareholders in Biomedica Molecular Medicine SL. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1697P - Assessing functional Androgen Receptor (AR) pathway activity using a computational model (ID 3490)

Presentation Number
1697P
Lecture Time
13:15 - 13:15
Speakers
  • A. Van Brussel
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Cellular signal transduction research identified 10-15 signaling pathways responsible for driving tumor growth. Defining pathway activity in tumor tissue is necessary to optimize targeted therapy choice. Verhaegh et al (Cancer Research 2014) used a Bayesian network approach to model transcriptional programs of signaling pathways. These pathway models use mRNA expression levels of validated direct pathway target genes to infer a probability of pathway activity in individual patient samples. Here, initial results of the AR model are presented.

Methods

28 bona fide AR target genes were selected and a Bayesian network model for the AR pathway was built and calibrated. The model uses target genes mRNA levels (Affymetrix HG-U133Plus2.0 array) as input to infer probability of AR pathway activity. Evaluation was done using multiple public datasets from clinical studies. The model was also adapted for qPCR data as input, using a subset of most informative target genes.

Results

Biological validation on androgen stimulated LNCaP cultures showed expected AR activity (GSE7868), which was inhibited by the anti-androgen bicalutamide (GSE7708). In cell line xenograft models (GSE21887, GSE33316, GSE966), AR was active in the presence of androgen and inactive in castrated mice. In prostate hyperplasia and 90% of primary prostate cancer (PCa) samples (GSE17951, GSE28403, GSE32982, GSE3325, GSE45016) AR was active; in contrast, AR was inactive in 30-50% of castration resistant or metastatic samples. AR was active in primary PCa samples, but not in samples taken 3 days after surgical castration (GSE32982). In other cancer types AR was mostly inactive, except for a subset of Her2 subtype Breast Cancer (BCa), Luminal BCa (EM-TAB-365, GSE12276, GSE17097, GSE21653), and meningioma samples (GSE16581, GSE9438). Translation to qPCR-RNA measurement as input was successful, underscoring the portability of our approach to other measurement platforms

Conclusions

Our biologically validated computational AR model enables assessing functional AR pathway activity in individual patient tissue samples, based on mRNA microarray or qPCR input from respectively FF or FFPE material. Other pathway models and clinical validation studies are in progress.

Legal entity responsible for the study

Philips Research

Funding

None

Disclosure

A. van Brussel, M.A. Inda, E. Den Biezen, D. van Strijp, J. Wrobel, H. van Ooijen, R. Hoffmann, W. Verhaegh, A. van de Stolpe: Employee of Philips Research.

Collapse
Poster display session Poster Display session

1698P - Evaluation of deamination bias from formalin-fixed tissues of small cell lung cancer with a dual strand targeted amplicon sequence (ID 4417)

Presentation Number
1698P
Lecture Time
13:15 - 13:15
Speakers
  • T. Amano
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Precision medicine is dependent on identifying actionable mutations in tumors. Accurate detection of mutations is often problematic in formalin-fixed paraffin-embedded (FFPE) tissues, as it causes DNA damage such as fragmentation and cytosine deamination. These Sequence artifacts can be difficult to distinguish from true mutations, and are an increasing interpretive issue. Understanding of the characteristics of these sequence artifacts in FFPE tissues is critical to improve the accurate detection of actionable mutations.

Methods

We reviewed the clinical courses of 156 small cell lung cancer (SCLC) patients who had undergone surgery at 17 institutions in Japan between January 2003 and January 2013. In these patients, we obtained the FFPE tissues of 79 cases which were histopathologically confirmed as SCLC and fitted for sequencing analysis with suitable DNA quality. Targeted amplicon sequence was conducted with MiSeq and TruSight panel (Illumina) which is a dual stranded amplicon kit for detecting cytosine deamination. We evaluated the characteristics of deamination bias and the relations with institutions and age of the tissue block.

Results

We could evaluate sequence of 73 samples data from 14 institutions. Target region of the sequencing was 26 genes, total 14686 bp. The total discordant single nucleotide variant (SNV) between forward and reverse strand were 690 cases, 16.4 cases per sample. The highest number of discordant SNV was 132 per sample. The most part of discordant SNV was the deamination change (C>T/G>A), 589 (85.4%) of 690 cases. The highest discordant SNV frequency was 0.25 with read depth 1876 in deamination change pattern, and 0.10 with read depth 4196 in the others. The frequency of the deamination change was different by institutions more than age of the tissue block.

Conclusions

Cytosine deamination from formalin fixation can be a major issue in diagnostic test of genome DNA for cancer samples. Procedures that assess, minimize or remove formalin-induced influences is important in the interpretation of genomic DNA analysis leading to better practice.

Legal entity responsible for the study

Toraji Amano

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1699P - Functional genomic mRNA (FGmRNA) profiling of > 18,000 tumor samples identifies potential new indications for antibody-drug conjugates (ADCs) in a broad range of tumor types (ID 4873)

Presentation Number
1699P
Lecture Time
13:15 - 13:15
Speakers
  • K. Moek
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

ADCs, consisting of an antibody designed against a specific antigen at the cell membrane linked with a cytotoxic agent, are an emerging class of therapeutics. Since ADC targets do not have to be drivers of tumor growth, ADCs are potentially relevant for a wide range of tumor types. Therefore, we aimed to define the landscape of ADC target expression in a broad range of tumor types.

Methods

PubMed and ClinicalTrials.gov were searched for ADCs that are or were evaluated in clinical cancer trials. Gene expression profiles of 18,055 patient derived tumor samples representing 60 tumor (sub)types and ≥ 3,520 samples representing 22 healthy tissue types were collected from the public domain. Next, we applied FGmRNA-profiling (Fehrmann et al. Nat Genet 2015;47:115-25) to predict per tumor type the overexpression rate at the protein level of ADC targets with healthy tissue samples as reference.

Results

We identified 87 ADCs directed against 59 unique targets. 17 ADC targets showed predicted overexpression of ≥ 75% of samples in at least 1 tumor (sub)type, 38 ≥ 50% and 56 ≥ 25%. A predicted overexpression rate of ≥ 10% of samples for multiple ADC targets was observed for high incidence tumors like breast cancer (n = 31 with n = 23 in triple negative breast cancer), colorectal cancer (n = 18), lung adenocarcinoma (n = 18), squamous cell lung cancer (n = 16) and prostate cancer (n = 5). In rare tumor types we identified targets showing high predicted overexpression, for example in uveal melanomas we found 95% predicted overexpression for c-MET.

Conclusions

This study provides a data driven prioritisation of available ADCs for clinical evaluation in 60 tumor (sub)types. This comprehensive ADC target landscape can support clinicians and drug developers in trial design.

Legal entity responsible for the study

UMCG

Funding

European Research Council advanced grant OnQview to E.G.E. de Vries; the Dutch Cancer Society grant, the NWO-VENI grant and a Mandema Stipendium to R.S.N. Fehrmann

Disclosure

E.G.E. de Vries: Advisory board: Medivation, Merck and Synthon: payments to the institution. Research grants: Amgen, Genentech/Roche, Chugai, Servier, Novartis, Synthon, AstraZeneca, Radius Health, CytomX, Nordic Nanovector: payments to the institution. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

TUMOUR BIOLOGY AND PATHOLOGY (ID 5680)

Lecture Time
13:15 - 13:15
Speakers
  • A. Bardelli
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15
Poster display session Poster Display session

1708P - Anti-tumor activity of alectinib in the orthotopic in vivo imaging model with NCOA4-RET fusion positive tumor cells (ID 1477)

Presentation Number
1708P
Lecture Time
13:15 - 13:15
Speakers
  • S. Yano
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The rearranged during transfection (RET) gene was discovered in 1985 as an oncogene produced by recombination during the transfection of NIH 3T3 cells with human lymphoma DNA. RET fusion genes were recently identified in a population of non-small cell lung cancers (NSCLCs). The most common (>80%) fusion partner for RET is KIF5B, followed by CCDC6, NCOA4, TRIM33, CLIP1, and ERC1. Recent clinical trials for RET fusion-positive NSCLC using vandetanib or cabozantinib demonstrated positive clinical response and considerable differential activities for RET inhibitors among fusion partners. Alectinib, an approved ALK inhibitor, is reported to inhibit KIF5B-RET and CCDC6-RET. However, the activity of alectinib with respect to RET with other fusion partners is unknown.

Methods

In the present study, we investigated the effects of alectinib on NCOA4-RET fusion-positive tumor cells (EHMES-10, a mesothelioma cell line) in vitro by MTT assay. We also examined the effect of alectinib utilizing orthotopic implantation model with EHMES-10 cells in the in vivo imaging model.

Results

Alectinib inhibited the viability of NCOA4-RET-positive EHMES-10 cells, as well as CCDC6-RET-positive LC-2/ad and TPC-1 cells. This was achieved via inhibition of the phosphorylation of RET and induction of apoptosis. Moreover, alectinib suppressed the production of thoracic tumors and pleural effusions in an orthotopic intrathoracic inoculation model of EHMES-10 cells. In vivo imaging of an orthotopically inoculated EHMES-10 cell model also revealed that alectinib could rescue pleural carcinomatosis.

Conclusions

These results suggest that alectinib may be a promising RET inhibitor against tumors positive for not only KIF5B-RET and CCDC6-RET, but also NCOA4-RET.

Legal entity responsible for the study

Kanazawa University

Funding

AMED in Japan

Disclosure

S. Yano: Research grants and honoraria from Chugai Pharma. All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1710P - Cathepsin S regulates cell migration and invasion through mediating store-operated calcium entry and the focal adhesion proteins (ID 3402)

Presentation Number
1710P
Lecture Time
13:15 - 13:15
Speakers
  • H. Lin
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Cathepsin S (CTSS), a lysosomal cysteine protease, plays an important role in inflammation, and it has been reported that it is also associated with angiogenesis and extracellular matrix (ECM) degradation promoting cell migration and invasion. Since CTSS is stably overexpressed in the different types of cancer cells, we explored a novel intracellular mechanism other than ECM degradation that regulates cell migration and metastasis.

Methods

Human oral cancer cells, OEC-M1, and breast cancer cells, MDA-MB-231, were used for this study. The expressions of CTSS were knockdown by siRNA transfection and the enzymatic activities were inhibited by highly-selective CTSS inhibitor, 58. The migratory and invasive abilities were determined by wound healing assay and transwell invasion assay, respectively. Microarray data and promoter prediction analysis were used to determine the intra-cellular targets of CTSS. Immunofluorescence assay was executed to evaluate STIM1 puncta formation and calcium influxes from store-operated calcium entry (SOCE) were measured by fura-2 calcium imaging. Western blot analysis was performed to detect the alteration of focal adhesion proteins.

Results

Our data showed that either CTSS knockdown with siRNA or activity inhibition could significantly decrease cell spreading area, and suppress cell migratory and invasive activities in both OEC-M1 and MDA-MB-231 cells. Moreover, inhibition of CTSS enzymatic activity resulted in the suppression of STIM1 aggregation and decreasing calcium influx from SOCE. Furthermore, downregulation of CTSS expression with siRNA could reduce the protein expression of three focal adhesion proteins, including CD29, CD104 and vinculin, which could be restored by CTSS transfection.

Conclusions

These results exhibit a novel intracellular molecular mechanism of CTSS mediating STIM1 aggregation and the calcium influx from SOCE to regulate the focal adhesion proteins, which are crucial for ECM interactions, cell migration and invasion.

Legal entity responsible for the study

Ministry of Science and Technology of Taiwan

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1711P - Type VI collagen (COL6) as part of tumorgenesis: Focus on quantifying specific COL6 protein fragments in serum (ID 4425)

Presentation Number
1711P
Lecture Time
13:15 - 13:15
Speakers
  • A. Wardak
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Type VI collagen (COL6) is emerging as an important component of the tumor microenvironment. The rationale that COL6 derived protein fragments may possess pro-tumorigenic properties has ample precedent (e.g. endothrophin). Little is however known, regarding COL6 degradation fragments as biomarkers for cancer. Here we address the biomarker potential of three specific COL6 degradation fragments measured in serum: Pro-C6 (C-terminal of the α3 chain/endotrophin), C6Ma3 (MMP-generated neo-epitope on the α3 chain), C6M (MMP-generated neo-epitope on the α1 chain).

Methods

Pro-C6, C6Ma3 and C6M were measured by validated competitive ELISAs in serum from patients with various stage solid tumors prior to treatment and healthy controls (table).

Results

C6M and C6Ma3 were significantly elevated (Kruskal-Wallis test) in most cancer types compared to controls, whereas Pro-C6 was not (table). A trend (p = 0.098) toward higher Pro-C6 was seen in the late (3/4) vs early (1/2) stage (Mann-Whitney test), whereas no difference was seen with C6M (p = 0.822) and C6Ma3 (p = 0.458). AUROC was 0.89 (p < 0.0001) and 0.86 (p < 0.0001) and 0.59 (p = 0.216) for C6M, C6Ma3 and Pro-C6, respectively, when comparing all cancer types combined to healthy controls.

1711P

Cancer:BreastColonGastricMelanomaNSCLCOvaryPancreasProstateSCLCHealthy controls
n8677108410721
stage2-32-31-31-31-31-31-31-21-4
C6M1 (p-value2)43.4 (**)56.0 (**)16.5 (ns)31.2 (ns)87.6 (****)47.6 (**)60.5 (*)33.9 (ns)49.5 (**)12.0 -
C6Ma31 (p-value2)2.7 (**)3.1 (**)1.1 (ns)2.3 (ns)3.1 (****)2.4 (ns)2.9 (*)2.3 (ns)2.6 (*)0.99 -
Pro-C61 (p-value2)7.2 (ns)9.8 (ns)7.4 (ns)8.9 (ns)8.4 (ns)20.8 (ns)10.6 (ns)7.1 (ns)9.2 (ns)9.5 -

mean, ng/ml;

vs. healthy controls;

p-value *<0.005, **<0.01, ****<0.0001, ns: not significant

Conclusions

Specific type VI collagen fragments were increased in serum from cancer patients compared to healthy controls, and showed promising clinical accuracy. This clearly support COL-6 remodeling/degradation as an important component in understanding tumorgenesis. Future studies will determine biological and clinical applicability of quantifying various COL-6 fragments in serum in relation to cancer.

Legal entity responsible for the study

Nordic Bioscience

Funding

None

Disclosure

A. Wardak, N. Willumsen, D.J. Mogensen, S.H. Nielsen, C. Jensen, S. Kehlet, M.A. Karsdal: Employed by Nordic Bioscience involved in biomarker development

Collapse
Poster display session Poster Display session

1712P - The combination analysis of tumor infiltration lymphocyte with Neutrophil to lymphocyte ratio may predict prognosis of colorectal cancer in stage I-III (ID 2361)

Presentation Number
1712P
Lecture Time
13:15 - 13:15
Speakers
  • S. Mortazavizadeh
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The tumor infiltration lymphocyte as local inflammation and neutrophil to lymphocyte ratio as systemic inflammation have been known as prognostic factors in colorectal cancer. But little is known about the correlation and impression of the local and systemic inflammation together on the prognosis of colorectal cancer. This study aimed to evaluate the effects of this combination on prognosis.

Methods

In a retrospective study, 206 patients from 2006-2015 with colorectal cancer after curative surgery have been investigated. the patients diagnosed with stage IV or simultaneously had secondary cancers were excluded. The pathological samples after surgery were studied for tumor infiltration lymphocytes (TIL) and other pathological features. Also neutrophil to lymphocyte ratio (NLR>2.38) was calculated from up to 3 days before surgery from peripheral blood. For analysis the combination of these markers, patients were divided to four groups for local inflammation or systemic inflammation predominantly (high TIL/High NLR, high TIL/Low NLR, Low TIL/High NLR and Low TIL/Low NLR) and then the overall survival (OS) and Disease free survival (DFS) for each group were calculated. Then compared with each other.

Results

For these identified patients the number of death events was 73and 133 were alive the median OS was 68 months(Range1 to 122) months .There was significant relationship between local and systemic inflammation (TIL and NLR) (p-value=0.0003), so that, when the local inflammation was predominantly and the systemic inflammation was dramatically low simultaneously, (high TIL/Low NLR) was associated with the best outcomes and improved the overall survival (mean OS = 72.56 month and HR:0.45) also low TIL was significant associated with poor prognosis on OS and DFS (p-value

Conclusions

The analysis of combination of local and systemic inflammation may predict the prognosis of colorectal cancer in patients who underwent the curative surgery. So in future this unexpansive and available methods seem can be used for determining the prognosis for these patients and used as markers.

Legal entity responsible for the study

Seyyed Mohammad Reza Mortazavizadeh

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1713P - Prognostic value of NK and T-lymphocyte markers in operable non-small cell lung cancer (NSCLC) (ID 5345)

Presentation Number
1713P
Lecture Time
13:15 - 13:15
Speakers
  • M. Skrzypski
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

New therapies aimed at activation of T and NK cells expand NSCLC treatments options. It is conceivable that markers designating ‘immune ignorant’, ‘immune excluding’ or ‘inflamed’ tumor phenotypes may influence the effectiveness of specific immune therapies.

Methods

qRT-PCR was used to assess the levels of 48 mRNAs in frozen tumor tissue sections from 115 stage I-IIIA NSCLC patients (33% never-smokers, 75% lung adenocarcinoma) who underwent pulmonary resection, and in matched normal lung parenchyma. mRNA expression (normalized vs. 4 reference genes) was compared between groups that did (45%) and did not relapse.

Results

Low expression of TIGIT (p.adj=0.031) and CTLA4 (p.adj=0.048) was correlated with shorter distant metastasis free survival after correction for multiple comparisons. In the subset of 75 lung adenocarcinoma cases, low expression of TIGIT, NCR3, CXCR3, FASLG, CD96, CTLA4, PD1, FYB and FOXP3 was correlated with shorter distant metastasis free survival after correction for multiple comparisons (p.adj<0.042). Expression of PD-1 (p = 0.016), PDL-2 (p = 0.029) and CTLA4 (p = 0.002) was significantly lower in relapsed vs. non-relapsed NSCLCs, whereas there was no difference for PDL-1. Expression of NK markers: NCR3 (p = 0.006) and CD96 (p = 0.005), but not NCR3-ligand 1 or NKG2D, NKG2C and NKG2A was significantly lower in relapsed vs. not relapsed NSCLCs. Expression of CXCR3 and its ligands: CXCL9 and CXCL10 (chemoattractants for lymphocytes), but not endothelin receptor type B, was significantly lower in relapsed NSCLCs (p < 0.03), which could provide a plausible explanatory mechanism for lower expression of lymphocyte markers in tumours with propensity for metastases. GITR, FOXP3 and CXCL9 expression was significantly higher in tumor samples vs. normal lung parenchyma (p.adj.<0.02). NCR3, CXCR3 and FASLG expression was significantly lower in tumor samples from smokers vs. never-smokers (p.adj<0.02). Samples of normal lung parenchyma from smokers were marked by higher expression of PD-1 and CD96 in reference to never-smokers (p.adj<0.04).

Conclusions

Non-inflamed NSCLC phenotype is associated with higher risk of distant metastases in early stage NSCLC. The non-inflamed phenotype is accompanied by lower expression of chemoattractants for lymphocytes. Expression of immune tolerance markers is increased in NSCLC compared to normal lung tissues.

Legal entity responsible for the study

Medical University of Gdansk

Funding

Medical University of Gdansk

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1714P - KRAS in non-small cell lung cancer (ID 5069)

Presentation Number
1714P
Lecture Time
13:15 - 13:15
Speakers
  • I. Amanam
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Disease heterogeneity with variable molecular mutations is one of the main contributory factors in non-small cell lung cancer (NSCLC). The goal of this study was to better understand the KRAS patients with co-occurring mutations.

Methods

We identified 60 patients with a diagnosis of NSCLC and a KRAS mutation in the COH Cancer Registry from 2009 to 2016. Next generation sequencing was performed.

Results

Of the 60 patients identified, 42 (70%) were Stage IV at diagnosis, 7 (12%) Stage I and 7 (12%) stage II and 4 (6%) Stage III. 47% (78) patients were smokers. Caucasian was the most common 44 (73%) racial group, followed by Asians 9 (15%), African-Americans 3 (5%), other 3(5%) and Pacific Islander 1 (1.7%). The average age at diagnosis was 67 (median 69.5) years; 30 patients (50%) were > 70 years, 23 (38%) patients were 51-69 years, and 7 (12%) 50 years or less. The most common histology was adenocarcinoma 52 (87%), then adenosquamous 3 (5%), large cell 2 (3%) and small cell, squamous cell and carcinosarcoma (1 each, less than 2% each). Majority had metastatic disease 52 (87%) with 20% (12) metastasis to brain, with average 1.6 metastatic sites. An average of 1.97 (range = 0-5) lines of therapy including chemotherapy, biologic agents or immunotherapy were received. 12 (20%) patients received immunotherapy, radiation in 28 (47%) and surgery in 22 (37%) with a median overall survival at 15 months. The most frequent molecular alteration was codon 12 mutation (47, 78%), followed by codon 13 (7, 12%) and codon 61 (6, 10%) mutations. The most common co-occurring mutations in this cohort were TP53 (15, 25%), ATM (9, 15%), LRP1B (9, 15%), ARID1A (8, 13%), STK11 (8, 13%), ARID1B (7, 12%), TERT (7, 12%), EGFR (6, 10%), RBM10 (6, 10%), SPTA1 (6, 10%). We are currently evaluating the relevance of the Circos plot analysis for these mutations, clinical response to immunotherapy and potential biomarkers.

Conclusions

KRAS mutations are among the most common molecular alterations identified in NSCLC. The discovery of effective treatments targeting KRAS mutations has represented a challenge so far. Understanding the significance of co-mutations and their therapeutic implications, especially in response to immunotherapy agents represents an important step to develop better treatment options for KRAS mutated lung cancers.

Legal entity responsible for the study

City of Hope National Medical Center

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1715P - PIK3CA mutation and PDL1 expression in lung squamous cell carcinoma surgically resected (ID 5480)

Presentation Number
1715P
Lecture Time
13:15 - 13:15
Speakers
  • L. Gutierrez Sanz
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Squamous-cell carcinoma (SCC) of the lung is the second most frequent histology in non-small cell lung cancer (NSCLC). Over the last decade new approaches targeting specific pathways in NSCLC have emerged but very few advances were made in its treatment. The PI3K-AKT-mTOR pathway is implicated in multiple cancer processes and PIK3CA mutations are being investigated in SCC as potential therapeutic target. The aim of the study was to evaluate the histological characteristics, mutations in PIK3CA and PD-L1 expression in these tumors.

Methods

Surgically resected lung SCC samples (FFPE) from 100 patients, stage I-III, were included in this study. Clinicopathologic characteristics included tumor size, TNM, smoking status, lymphovascular and pleural invasion, histopathological grade, stromal lymphoplasmacytic reaction and type of tumoral growing. DNA was isolated from 92 samples according to standard procedures and PIK3CA mutation analysis was done using Cobas 4800 platform. PD-L1 expression was analyzed in 74 cases by immunohistochemistry with PDL1 22C3 pharmDX assay. The PD-L1 expression was evaluated by tumor proportion scores (TPS) as IASLC guidelines.

Results

The mean age was 68 years (53-86), 14 females and 86 males. About staging: 53 patients had stage I, 27 stage II, and 19 stage III. 53% and 35% showed vascular and pleural invasion respectively. Low to moderate stromal lymphoplasmacytic reaction was found in 34% and severe in 11%. PI3KCA mutation was found in 9/92 (9,8%) patients: E545X (n = 2), E542K (n = 5), H1047X (n = 1), C420R (n = 1). PD-L1-expression was found in 31 out of 74 cases (42%): 14 cases with TPS between 1-49%; 17 cases with TPS >50%. 5/9 (55%) of PIK3CA mutated cases were PD-L1 positive (2 of them >50%). In PIK3CA non-mutated cases 26 out of 67 (39%) showed PD-L1 expression: 11 cases with TPS between 1-49%; 15 cases with TPS >50%.

Conclusions

PIK3CA mutation was found in 9,8% of SCC of the lung, most of them in exon 9. PD-L1 expression was found in 42% of the SCC in our series. 55% of PIK3CA mutated patients were positive for PD-L1 expression. No correlation has been found between PD-L1 expression and PIK3CA mutation in SCC. It is important to know more about the relation of these factors to select the patients for immunotherapy and new target agents.

Legal entity responsible for the study

IIS

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1716P - TRKA expression and NTRK1 gene copy number across solid tumors (ID 5146)

Presentation Number
1716P
Lecture Time
13:15 - 13:15
Speakers
  • G. Mauri
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Neurotrophic Tropomyosin Kinase Receptor 1 (NTRK1) gene encodes for the protein Tropomyosin-related kinase A (TRKA). A deregulated activity of TRKA has been detected in cancer, leading to oncogenic activity. This can result from translocations, amplifications, deletions and point mutations involving the NTRK1 gene. The incidence of NTRK1 gene copy number gain (GCN) across solid tumors has not been investigated. We present here the results of an immunohistochemistry (IHC) screening for TRKA expression within the phase I ALKA-001 clinical trial. Clinical results of ALKA-001 clinical trial are not presented here.

Methods

Formalin-fixed paraffin-embedded (FFPE) consecutive samples of different solid tumors were tested for TRKA IHC staining. Samples showing TRKA IHC staining in at least 10% of cells were further studied by fluorescence in situ hybridization (FISH) to assess whether NTRK1 gene rearrangements were present and to assess GCN. All patients signed informed consent for molecular screening according to the phase I ALKA-001 clinical trial.

Results

1043 samples were tested; annotation for histology was available in 1023. Most of the samples were colorectal adenocarcinoma (CRC) (n = 550, 53.8%) or lung adenocarcinoma (312, 30.5%). 24 samples (2.3%) were biliary tract carcinoma (BTC). Seventeen (1.6%) samples were characterized by TRKA IHC expression (4 weak, 8 moderate, 5 strong). By FISH, 1/17 (5.9%) displayed NTRK1 gene rearrangement and 15 (88.2%) NTRK1 GCN gain. Among samples harboring NTRK1 GCN gain, 8 (53%) were lung adenocarcinoma, 3 (20.0%) BTC and 2 (13.3%) CRC. Five (33.3%) samples had concomitant ALK and ROS1 GCN gain. None of the lung adenocarcinoma (n = 8) had concomitant EGFR mutations. Both CRC samples (n = 2) harbored KRAS mutation. No correlation was found between grading of TRKA IHC staining and the number of NTRK1 GCN.

Conclusions

NTRK1 GCN gain can be found in 1.6% of solid tumors. In particular, we found GCN gain in 2.6% of lung adenocarcinomas, without EGFR mutations, 0.4% of CRC and 17.6% of BTC even though a limited number of the latter histology was included in the analysis. The prognostic and translational therapeutic impact of this genetic alteration remains to be established.

Clinical trial identification

Not applicable

Legal entity responsible for the study

Salvatore Siena

Funding

The molecular screening was funded by Ignyta within ALKA-372-001 study. Investigators are supported by Fondazione Oncologia Niguarda Onlus - Project: “Terapia molecolare dei tumori” (G.M., S.S. A.S-B.). This project has received funding from the European Union's Horizon 2020 research and innovation programma under grant agreement No 635342

Disclosure

S. Siena: Consultant/advisory board member for Amgen, Bayer, Eli Lilly, Merck, Merrimack, and Roche.All other authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1717P - The correlation between MMR gene expression MSH2/MSH6 and VEGF A/VEGF B in gastro-esophageal cancer (ID 2850)

Presentation Number
1717P
Lecture Time
13:15 - 13:15
Speakers
  • A. Dracea
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

VEGF proteins are key regulators of angiogenesis and targeting VEGF A led to inhibition of new blood vessels formation, with important therapeutic effects in various cancers. The roles of VEGF B are controversial; this peptide expression seems to inhibit apoptosis by suppressing many apoptotic/cell death-related genes and to facilitate metastasis by inducing vascular leakiness, leading to a high degree of tissue hypoxia that consequently activates DNA damage signalling pathways. MSH2/MSH6 is an important complex of proteins in DNA mismatch repair system and their altered expression could represent a response to the rapidly growing number of replication errors in a tissue with a high index of proliferation.

Considering that in certain conditions, DNA damage response products, such as H2AX, promote tumor growth and angiogenesis, in the present study we aimed to identify a common pattern of expression behavior between MMR genes MSH2/MSH6 and VEGF components (VEGF A and VEGF B), in order to use these genes as diagnostic markers in gastro-esophageal cancer.

Methods

mRNA levels of MSH2, MSH6, VEGF A, VEGF B were evaluated in tumoral and peritumoral tissues samples biopsied from 36 patients using qRT-PCR with specific TaqMan gene expression assays.

Results

VEGF A/VEGF B and MSH2/MSH6 mRNAs were expressed in both tumour and peritumour mucosa, with a tendency of tumoral up-regulation for VEGF A and MSH2/MSH6. When comparing the differences between tumoral/peritumoral expression level among the studied genes, we found that MMR and VEGF have a similar pattern of expression as follows: VEGF A gene expression correlates with MSH2 (rho Spearman = 0.4562; p < 0.05) and also, is similar to MSH6 (rho Spearman = 0.5082 p < 0.05); furthermore, VEGF B gene expression is correlated with MSH2 expression (rho Spearman = 0.5350 p < 0.05), with a very strong correlation between MSH6/VEGF B expression (rho Spearman = 0.6730 p < 0.0001).

Conclusions

Our results indicate a possible crosstalk between DNA mismatch repair and VEGF signaling pathways, providing new insight into understanding the potential connection of VEGF B and MSH6 in carcinogenesis.

Legal entity responsible for the study

University of Medicine and Pharmacy of Craiova, Romania

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1718P - Liver-type glutaminase (GAB) suppresses malignant phenotype of glioblastoma cells (ID 3254)

Presentation Number
1718P
Lecture Time
13:15 - 13:15
Speakers
  • E. Majewska
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Glutamine (Gln) plays a pivotal role in the metabolism of tumors of different including glioblastoma (GBM), the most aggressive brain tumor. Glutaminase (GA, EC 3.5.1.2) converts Gln to glutamate (Glu) and ammonia. GA is encoded by two genes: GLS and GLS2, encoding kidney-type isoforms (KGA and GAC) and liver-type isoforms (GAB and LGA), respectively. Kidney-type isoforms promote cell proliferation, while the liver-type isoforms relate to quiescent state of cells. In GBM GLS is highly expressed, while GLS2 is hardly detectable. Transfection of human GBM T98G cell line with a sequence encoding GAB is known to decrease their survival, proliferation index and migration and sensitizes them to damage by hydrogen peroxide. To examine whether the mode of action of GAB extends to other GBM cell lines, the effect of GAB transfection of U87MG, U251MG and LN229 cells with GAB was assessed.

Methods

Mitochondrial activity was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) conversion (details in M. Szeliga et al., Glia, 2009). Cell proliferation was measured by a commercially available ELISA kit based on the detection of BrdU (5-bromo-2'-deoxyuridine) incorporated into the genomic DNA. Migration was analyzed using the scratch assay. The tip scratch of cell monolayer was photographed under Juli Smart cell analyzer and measured after 0 and 24 h. Ability to form colonies was assessed after 14 days of culture following Giemsa staining of fixed cells.

Results

Transfection with GAB: i) decreased mitochondrial activity, proliferation and colony formation ability of U87MG cells ii) inhibited ability of U251MG cells to form colonies iii) decreased mitochondrial activity, proliferation, migration and colony formation ability of LN229 cells. All transfected cells were more sensitive to hydrogen peroxide as compared to the controls.

Conclusions

Suppression of malignant phenotype and their sensitization to hydrogen peroxide damage by GAB transfection appears to be a feature common to all the glioblastoma cell lines so far studied.

Legal entity responsible for the study

Jan Albrecht, PhD

Funding

Ministry of Science and Higher Education of Poland, The Leading National Research Centre (KNOW-MMRC)

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1719P - Alteration of p53 mRNA expression in neuroblastoma and its impact in disease outcome (ID 4505)

Presentation Number
1719P
Lecture Time
13:15 - 13:15
Speakers
  • M. Inomistova
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Neuroblastoma is frequent childhood malignant tumor with high clinical heterogeneity. Despite the rare mutations of TP53 gene, p53-mediated pathway is often inactivated in neuroblastoma. The significance of MDM2, p53 direct antagonist, overexpression in neuroblastoma clinical course and outcome has been already established. But still remain patients with favorable clinical features and poor disease outcome.

Methods

The case group comprised 68 children with neuroblastoma (mean age: 36.7±4.7 months; primary tumors: 88%; MYCN+: 39%; MDM2 overexpressed: 70%). p53 mRNA expression level (EL) was analyzed in tumor samples with qRT-PCR and evaluated by the ΔΔCt method according to control GAPDH mRNA EL.

Results

We established that the value of p53 EL in neuroblastoma cells varied in wide limits. Significantly lower p53 EL was detected in recurrent and metastatic tumor samples comparing to primary tumors (P = 0.001). Insignificant increase of p53 EL in patients with unfavorable clinical and biological features (late occurrence age, IV stage, MYCN amplification) was observed. However, we revealed significant increase of p53 EL in MDM2 overexpressed tumors (P = 0.007). With ROC-analysis we assessed the optimal criterions for distribution of patients according to p53 expression (OC:>1.18 a.u., P = 0.04, AUC:0.69 for high and OC:<0.09 a.u., P = 0.006, AUC:0.84 for low MDM2 expression). We have analyzed 3-year event-free survival (EFS) of patients with neuroblastoma and established 100% EFS survival for patients with low MDM2 and high p53 expressions, while in other groups significant decrease in survival was observed (P < 0.05). EFS rates of patients with low p53/high MDM2 and high p53/low MDM2 expressions were similar (27.7% and 33.3%) and for p53/MDM2 overexpressed tumors it was only 18.2% (P < 0.05).

Conclusions

Regulation of p53-mediated pathway is complex and multicomponent system. Alteration of p53 EL is independent from clinical features marker of neuroblastoma. Analysis of p53/MDM2 co-expression provides the possibility for better neuroblastoma outcome prediction.

Legal entity responsible for the study

National Cancer Institute of Ministry of Public Health of Ukraine

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1720P - Downregulation of BRCA1 protein in clear cell renal cellular carcinoma (ID 4691)

Presentation Number
1720P
Lecture Time
13:15 - 13:15
Speakers
  • E. Sarnowska
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Around 75% of renal cancer in adult kidney is clear cell renal cellular carcinoma (ccRCC). This type of cancer is characterized by lipids overacumulation and mutations in VHL (around 90% cases), BAP1 and PBRM1 genes as well as stabilization of HIF1α transcription factor. Additionally, metabolic switch to aerobic glycolysis and aberration in TCA cycle was observed in ccRCC independently on the stage of the disease. Moreover, the mTOR pathway hyperactivation and downregulation of AMPK pathway featured the ccRCC. This type of cancer is highly resistant to classical chemotherapy. BRCA1 is tumor suppressor gene, mutation in this gene is associated with breast and ovarian cancer. BRCA1 is a protein involved in DNA repair and apoptosis also interacts with BRG1 – core subunit of SWI/SNF chromatin remodeling complex. BRCA1 is transcribed from bidirectional promoter together with NBR2 – lncRNA. Interestingly, NBR2 interacts with AMPK and is downregulated in ccRCC. CTCF is a protein which binds Topologicaly Associated Domains, and CTCF binding site was found in BRCA1/NBR2 promoter region.

Methods

Immunohistochemistry (IHC) on paraffin embedded clinical samples for BRCA1 and BRG1 core subunit of SWI/SNF complex, comparative transcriptomic study, co-immunoprecipitation (Co-IP) and chromatin immunoprecipitation (ChIP) method were used in this work.

Results

In this study we found downregulation of BRCA1 and BRG1 proteins in ccRCC patient samples independently on stage of the disease. Interestingly, downregulation of BRG1 was more severe in samples with strong lymphocyte infiltration. By contrast, downregulation at the transcript level was observed for BRG1 encoding gene but not for BRCA1. BRG1 and CTCF co-precipitated from cancer cells, indicating the existence of co-interaction between CTCF, BRG1 and BRCA1 proteins. Additionally, overexpression of BRG1 caused increased expression of CTCF in human cells. We also found that BRG1 targets both CTCF and BRCA1 genes.

Conclusions

BRCA1, BRG1 and CTCF module is dysregulated in ccRCC cells independently on Fuhrman grade and stage of the disease. This missregulation can have a brought spectrum of changes including 3D chromatin structure, transcription, epigenetic and others.

Legal entity responsible for the study

Elzbieta Sarnowska

Funding

This work was supported by grant from National Science Center No UMO–2013/11/B/NZ2/00132

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1721P - The role of PD-L1 in a high-grade invasive human oral squamous cell carcinoma microenvironment (ID 1946)

Presentation Number
1721P
Lecture Time
13:15 - 13:15
Speakers
  • M. HIRAI
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Blockade of the programmed-death 1 receptor (PD-1)/programmed-death ligand (PD-L1) pathway efficiently reduces tumour growth and improves survival. Durable tumour regression with blockade of the PD-1/PD-L1 checkpoint has been demonstrated in recent clinical studies. Oral squamous cell carcinoma (OSCC) is highly immunosuppressive, and PD-L1 expression has been proposed as a potential mechanism responsible for this phenotype. Despite the fact that anti-PD-1 treatment can produce durable responses, such therapy appears to benefit only a subset of patients. Thus, it is important to understand the mechanisms underlying the regulation of PD-L1 expression in the OSCC microenvironment.

Methods

The subjects were patients with primary OSCC who underwent surgical resection at the Kanazawa University Hospital between 1998 and 2008. And, three human oral squamous cell carcinoma cell lines established from tumor biopsies with different grade of invasive abilities were used: OSC-20, OSC-19 and TSU.

Results

We showed that PD-L1 expression in high-grade invasive OSCC cell lines was lower than that in a low-grade invasive OSCC line and found a close correlation between PD-L1 expression and the epithelial-mesenchymal transition (EMT). PD-L1 expression was upregulated in macrophages and dendritic cells (DCs) in high-grade invasive human OSCC tissues or co-cultured with mesenchymal-phenotype OSCC cells in vitro. TLR4-inhibitory peptide successfully suppressed PD-L1 upregulation on macrophages and DCs co-cultured with mesenchymal-phenotype OSCC cells, suggesting that some EMT-induced tumour antigen is critical for PD-L1 induction on tumour-associated macrophages and DCs.

Conclusions

Further studies are necessary to explore the impact of EMT on the tumour immune microenvironment and to identify potential biomarkers for selecting patients who might preferentially benefit from PD-1/PD-L1 blockade or immunotherapies more broadly.

Legal entity responsible for the study

Kanazawa University

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1722P - Loss of SWI/SNF chromatin remodelling complex is linked to advanced urinary bladder cancer (ID 3856)

Presentation Number
1722P
Lecture Time
13:15 - 13:15
Speakers
  • M. Szymanski
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Bladder cancer originated from urothelium belongs to the top ten among all tumors. Various factors like genetic and molecular defects, appearance of different tumors in the family, previous genitourinary disorders and exposure to chemical compounds are reported as potential causes of this type of cancer. It has been reported that TP53, p21 or Ras mutations and epigenetic alterations of genes coding for these oncogenes are involved in the aetiology of urothelium originating bladder cancer. Additionally, the TCGA study indicated that such important regulatory pathways/machineries like these controlling cell cycle; (PI(3)K)/AKT/mTOR signaling involved in the metabolism control; and chromatin modificators including SWI/SNF chromatin remodeling complex (CRC) are affected in this disease.

Methods

Immunohistochemistry (IHC) on paraffin embedded clinical samples for SWI/SNF core subunits and key enzymes involved in metabolism control, comparative transcriptomic study and confirmatory quantitative real-time PCR (qRT-PCR) were used in this work.

Results

In this study we found a substantial decrease of protein levels of SWI/SNF core subunits in bladder cancer clinical samples. Subsequently, we performed reanalysis of transcriptomic data for clinical samples obtained from GEO database and confirmatory assessment of the transcript level in clinical samples. This analysis showed that the reduced protein level of SWI/SNF core subunits observed in advanced bladder cancer is likely caused by the decreased abundance of corresponding transcripts. We also found that the SWI/SNF complex interacts in human cells with key proteins involved in the control of energy status and glucose metabolism. The IHC analysis indicated altered abundance of these enzymes in cancer cells when compared to normal urothelium consistently to strong metabolic alterations characteristic for this type of cancer.

Conclusions

The down-regulation of SWI/SNF complex on both transcript and protein level, and decreased activity of its partner proteins link the molecular features with metabolic alterations observed in this type of cancer.

Legal entity responsible for the study

Michal Szymanski

Funding

This work was supported by grant from National Science Center No UMO–2014/13/B/NZ2/01187

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1723P - The role of hepatocyte nuclear factor 1 homeobox B (HNF1B) loss in chromophobe RCC (ChRCC) development (ID 4214)

Presentation Number
1723P
Lecture Time
13:15 - 13:15
Speakers
  • E. Jonasch
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

ChRCC is characterized by dramatic chromosomal copy number (CN) changes. Currently, no model is available to precisely elucidate the molecular drivers of this rare tumor. HNF1B is a master regulator of gene expression, and both mutated HNF1B and downregulated HNF1B protein levels have previously been described in ChRCC.

Methods

We queried The Cancer Genome Atlas ChRCC database and analyzed tissue microarray data to determine the relative levels of HNF1B in ChRCC versus other RCC subtypes, and assessed the prognostic impact of dual HNF1B and TP53 loss. We knocked out Hnf1b in proliferating murine embryo fibroblasts (MEFs) and human ACHN cells and measured the effect on gene and protein expression of checkpoint regulatory proteins, spindle integrity, and aneuploidy. We then performed dual knockdown of HNF1B and TP53 and assessed cellular behavior.

Results

We found that HNF1B transcript and HNF1B protein were downregulated in the majority of ChRCC in TCGA, and the magnitude of HNF1B loss is unique to ChRCC. Additionally, we observed a strong correlation between reduction of HNF1B expression and aneuploidy in ChRCC patients. In MEF cells deficient in Hnf1b, we observed the development of aneuploidy. Hnf1b deficiency also reduced spindle checkpoint protein (MAD2L1, BUB1B) and cell cycle checkpoint protein (RB1 and p27) expression, and altered chromatin access of Mad2l1, Bub1b and Rb1 genes. Coordinate loss of Bub1b and Rb1 recapitulated the polynuclearity and larger cell size seen with Hnf1b depletion. TCGA data also showed that TP53 is mutated in 33% of ChRCC whose HNF1B expression was repressed, and the combination of HNF1B loss with TP53 mutation was associated with poor prognosis. The combination of HNF1B loss with TP53 inactivation led to increased cell proliferation and increased aneuploidy, providing evidence that coordinate loss of HNF1B and TP53 may enhance cellular survival and engender an aggressive ChRCC tumor phenotype.

Conclusions

HNF1B deficiency is a major driver of chromosomal instability in ChRCC and lethality is associated with subsequent TP53 loss. Further development of model systems with combined HNF1B/TP53 loss will accelerate the development of treatments specific for ChRCC.

Legal entity responsible for the study

UT MD Anderson Cancer Center

Funding

National Institutes of Health

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1724P - Some mechanisms of increasing malignancy of B16/F10 melanoma in female mice with chronic pain (ID 2216)

Presentation Number
1724P
Lecture Time
13:15 - 13:15
Speakers
  • I. Kotieva
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The impact of chronic pain (CP) on the growth and development of tumors is poorly studied. However, one of the main goals of cancer therapy is the relief of chronic pain, a complex symptom based on combined pathophysiological mechanisms. Our aim was to study the influence of CP on melanoma growth in female mice and to determine levels of the VEGF family members in the tumor (T), its perifocal zone (PZ) and in the skin.

Methods

The study included 64 female C57BL/6 mice; B16/F10 melanoma was transplanted under the skin on the back of animals in the main group 2 weeks after sciatic nerve ligation. Mice with melanoma without CP were used as the controls. Levels of VEGFs: A, C, R1 and R3 were determined by ELISA (BenderMedSystem, Austria).

Results

The life span of mice with melanoma and CP was 1.5 times shorter than the control group. Melanoma in mice with CP was more aggressive, and metastases occurred after 1 week vs. 4 weeks in the controls. The rate of metastasis was higher (100% vs. 60% in the controls); melanoma with CP spread to multiple organs and caused unusual metastases to the heart and uterus. The rapid and specific development of B16/F10 melanoma in mice with CP was accompanied by increased levels of VEGF-A, -C and -R1 in T, PZ and the skin, with their maximal accumulation in T in week 1. The VEGF-A level continued to increase in T and PZ (PZ

Conclusions

CP shortened the life span of female mice with melanoma and enhanced the aggressiveness of B16/F10 melanoma metastases. The activation of angiogenesis in T and PZ can be considered as one of the mechanisms of the neoplastic progression.

Legal entity responsible for the study

Rostov Research Institute of Oncology

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1725P - Impact of global epigenetics machinery on clinical outcome of colorectal cancer patients treated with fluoropyrimidine-based therapy (ID 2741)

Presentation Number
1725P
Lecture Time
13:15 - 13:15
Speakers
  • M. Fouad
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The pathogenesis of colorectal cancer (CRC) is complex and influenced by many factors related to genetic, epigenetics and chronic inflammatory processes.

Methods

This is a prospective study, conducted on 102 Egyptian patients, diagnosed with CRC. Blood samples were collected at baseline and after 3 & 6 months of receiving fluoropyrimidine (FP) based therapy. DNA methylation was measured by LC/MS/MS spectroscopy, acetylated histones (H3) and (H4) were measured by ELISA and RNA expression of FP metabolizing enzymes (TS, TP and DPD), DNA methyl transferases (DNMT3A and B) in addition to inflammatory markers (COX2, IL6, and IL1B) by qRT-PCR.

Results

The median age of the studied patients was 46 years, 47% of them were ≤ 45 years. Forty patients (38.8%) had rectal cancer, they exhibited significant H3 hyperacetylation and upregulation of COX2 and IL1B along with significantly lower median overall survival compared to colonic patients (14.6 versus 23 months respectively). FP therapy produced significant decrease in global methylation, acetylated H3 & H4 levels, downregulation in TP and DNMT3B but significant upregulation in TS and DPD over treatment time. Significant positive correlations were found between global methylation and IL1B (r2= 0.25, P = 0.01), acetylated H3 with DPD and COX2 (r2= 0.28, P = 0.02 and r2= 0.27, P = 0.03 respectively) and 5 methylated cytosine content (5MC) with DNMT3A and IL6 (r2= 0.25, P = 0.04 and r2= 0.34, P = 0.004 respectively). Overexpression of COX2 > 17 had a significant poor prognostic effect on overall and event free survivals (HR = 0.58, P = 0.003 and HR = 0.72, P = 0.008 respectively). Also patients who had global methylation> 30 showed significant reduced event free survival by 39% (P = 0.04).

Conclusions

Global methylation and H3 acetylation regulated COX2, IL6 and IL1B which were not affected by the therapy, however H3 upregulated TS and DPD. Rectal cancer patients showed significant H3 hyperacetylation, upregulation of COX2 and IL1B along with significant lower overall survival.

Legal entity responsible for the study

National Cancer Institute, Cairo University

Funding

National Cancer Institute, Cairo University

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1726P - Comparison of breast cancer subtypes between young and elderly women (ID 4068)

Presentation Number
1726P
Lecture Time
13:15 - 13:15
Speakers
  • I. Kiladze
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Breast cancer(BC) are increasingly recognized as heterogeneous disease based on expression of receptors for estrogen (ER), progesterone (PR), and human epidermal growth factor receptor 2 (HER2). There are four main subtypes of BC with differing tumor characteristics and different risk factors, treatment options and prognoses. Few data exist on the frequency of molecular subtypes in young and older women. The purpose of this study is to compare the distribution of the BC subtypes in young and elderly patients.

Methods

BC during the period 2013 to 2015 including ER/PR and HER2 status, was obtained from the Georgian main histopathology laboratories all over the country. We analyzed 1003 women with BC included 85 women aged 20 to 39 years (YA’s), 118 women aged 40 to 45 years (older premenopausal),665 women aged 46-70 (postmenopausal) and 135women older than 70 years (elderly group) at diagnosis. Incidence rates were calculated by subtype (triple-negative; HR +/HER2 -; HR +/HER2 +; HR -/HER2 +), and differences in subtype characteristics by age groups were evaluated.

Results

The incidence of BC in YA’s was 8,5%. The most common BC subtype was HR +/HER2- among all age groups, and HR -/HER2 + was the least; however, the relative contribution of each subtype varied within age categories. In YA’s HR +/HER2 - was the most commonly diagnosed subtype (62%), followed by HR +/HER2 + (15%), triple-negative (12%) and HR -/HER2 + (11%). Statistically no significant difference of BC subtypes was observed in age groups. HR +/HER2 - subtype was lesser in YA’s than in elder population (62% vs 73%), but statistically non-significant (p = 0.19) and there was not significant difference in prognostically “unfavorable” subtypes (HR-/HER2+ and triple-negative) (23% vs 17%) (p = 0.134; (CI) 95%: 0.09 to 1.71). Surprisingly no difference of Triple-negative BC was observed in YA and elderly groups (12% vs 13%).

Conclusions

The distribution of breast cancer subtypes among young adults didn’t vary from that observed in older women. Our study results seem to be in contradiction with other studies previously reported in literature. Future studies should consider whether distribution of breast cancer subtypes influences long-term survival in young compared with older women.

Clinical trial identification

GYO 02/17

Legal entity responsible for the study

Georgian Group of Young Oncologists

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1727P - Distribution of the PAM50 breast cancer subtypes within each pathology-based group: a combined analysis of 15,339 patients across 29 studies (ID 5055)

Presentation Number
1727P
Lecture Time
13:15 - 13:15
Speakers
  • J. Cejalvo
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Current classification of breast cancer in the clinical setting is based on pathology-based biomarkers such as hormone receptors (HR) and HER2. More recently, identification of the intrinsic molecular subtypes (IS) within each pathology-based group (i.e. HR+/HER2-, HR+/HER2+, HR-/HER2+ and triple-negative [TN]) is revealing clinical value. The objective of this study is to assess the distribution of the IS within each pathology-based group in a large series of breast cancer.

Methods

Twenty-nine studies were identified from the literature (2009–2017) in which IS and pathology-based data were reported. HR was evaluated by immunohistochemistry (IHC) and HER2 by IHC and/or FISH according to standard criteria. Pathology-based groups were divided into 4 groups: HR+/HER2-, HR+/HER2+, HR-/HER2+ and TN. IS (Luminal A [LumA], Luminal B [LumB], HER2-enriched [HER2-E], Basal-like [BL] and Normal-like) were identified using the research-based, or the standardized, PAM50 gene expression-based assay.

Results

PAM50 and pathology data was available in 15,339 patients. The distribution of the PAM50 IS within each IHC-based group is shown in Table 1. Within HR+/HER2- group (n = 9,768), non-luminal subtypes (HER2-E and BL) represented 5.6% and 2.2%, respectively. Within HR+/HER2+ group (n = 1,727), HER2-E and BL represented 29.2% and 2.1%, respectively. Within HR-/HER2+ group (n = 1,332), non-HER2-E subtypes (Luminal A/B and BL) represented 9.3% and 13.8%, respectively. Finally, within TN (n = 2,512), non-BL subtypes (Luminal A/B and HER2-E) represented 5.9% and 11.1%, respectively.

1727P

IHC-BasedPAM50 intrinsic subtype distribution (%)
n = 15,339%LumALumBHER2-EBLNormal
HR+/HER2-63.6854.4734.415.642.243.24
HR+/HER2+11.2634.4530.5729.182.083.71
HR-/HER2+8.686.912.4069.7413.817.13
HR-/HER2-16.383.532.3911.1578.234.70

Conclusions

Our results confirm previous observations that all IS are represented within each pathology-based group. Based on our observations, future clinical trials in unselected breast cancer patient populations should be sufficiently powered to address the prognostic and predictive ability of the IS.

Legal entity responsible for the study

Hospital Clinic Barcelona. August Pi I Sunyer Biomedical Research Institute (IDIBAPS)

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1728P - Genetic association of matrix metalloproteinase MMP- 1, MMP-3 and MMP-9 Genes with HCV-related hepatocellular carcinoma in Egyptian patients (ID 2210)

Presentation Number
1728P
Lecture Time
13:15 - 13:15
Speakers
  • A. Alhanafy
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Hepatocellular Carcinoma (HCC) is one of the most frequent cancers in Egypt where there is high prevalence of infection with the Hepatitis C virus (HCV). Matrix metalloproteinases (MMPs) are multifunctional proteins that play an important role in cell development, differentiation, inflammation and angiogenesis. Polymorphisms in MMPs genes might be involved in development of HCV related HCC. The aim of this study was to explore the relationship of gene polymorphisms in MMP-1,3 & 9 with liver cirrhosis and HCC patients.

Methods

The study included 128 subjects enrolled from Menoufia University Hospital inpatients and outpatients clinics from the Department of Clinical Oncology & Nuclear Medicine and Department of Tropical Medicine in the period between October 2015 and August 2016. Patients were classified into three groups. Group I: 48 patients with HCC in addition to liver cirrhosis, including 26 males and 22 females with a mean age of 58.60±5.29; Group II: 50 patients with liver cirrhosis, including 26 males and 24 females with a mean age of 56.74±5.21; Group III: 30 healthy subjects as controls, including 15 males and 15 females with a mean age of 56.30 ±7.30. Diagnosis of HCC was performed using two imaging methods (abdominal US & triphasic CT). All subjects except controls were positive for serum HCV RNA. Liver function tests, AFP & CHILD score were assessed. Gene polymorphisms were analysed using PCR-RFLP.

Results

HCC patients had higher mutant G2G2 (35.4%) and G2 allele (62.5%) of the MMP-1 gene than patients in both cirrhotic (P < 0.05) and control groups (P < 0.001). In addition, for the MMP-3 gene, HCC patients had the most noteworthy predominance of mutant 5A/5A (22.9%) and 5A allele (52.1%) compared to the cirrhotic (P < 0.05) and control groups (P < 0.001). The results of MMP-9 gene analysis uncovered a higher frequency of the mutant TT genotype and T allele in both HCC (56.3% and 74% respectively) and cirrhotic groups (10% and 35% respectively) compared to the control group. In HCC patients, we detected a significant correlation between heterozygote G1/G2 and G2/G2 of the MMP-1 gene and homozygote TT of MMP-9 with a higher CHILD score, tumor size and stage (P < 0.05). Moreover, MMP-3 5A/6A was associated with a higher CHILD score, portal vein thrombosis and stage (P < 0.05) compared to other genotypes.

Conclusions

Gene mutations in MMP-1,3, 9 may be involved in progression of liver cirrhosis and risk relationship for HCC development.

Legal entity responsible for the study

Menoufia University

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1729P - Heterogenity of epigenetic and EMT marks observed in hepatocellular carcinoma with keratin 19 proficiency (ID 4523)

Presentation Number
1729P
Lecture Time
13:15 - 13:15
Speakers
  • T. Nagasaka
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

The expression of keratin 19 (K19) has been proposed as a novel predictor for poor prognosis in patients with hepatocellular carcinoma (HCC). However, the cell origin of K19-proficient HCC remains unclear. Herein we tried to reveal the cell origin of K19-proficient HCC by tracing epigenetic footprints in cultured cells and clinical materials.

Methods

The KRT19 gene, which encodes K19, has a CpG island in promoter region and therefore implicates DNA methylation as a potential epigenetic process for K19 expression. Firstly, we examined epigenetic alterations in K19-positive HCC cell lines. Next, from a panel of 564 surgically resected HCCs, we clarify the clinicopathological relevance of K19-preficent HCCs by analyzing robust methylation analyses in KRT19 promoter region and LINE-1 in comparison with other cholangiocytic (K7), hepatocytic markers (HepPar-1 and Arginase-1), EMT markers (E-cadherin and vimentin), and a signal pathway associate with biliary differentiation (NOTCH-1).

Results

In vitro, although methylation in KRT19 promoter was associated with K19 deficiency, 5-aza-dC treatment failed to re-expression of K19. From 564 surgically resected HCCs, a cohort of 125 HCC patients was selected and analyzed after exclusion of HCC with recurrence, TNM stage as IIIB or more, preoperative therapy, transplantation, and combined hepatocellular-cholangiocarcinoma. In this cohort, K19 expression was found in 29 HCCs (23.2%), and corresponded with poor survival following surgery (P = 0.025) and extrahepatic recurrence free survival (P = 0.017). Compared with K19-deficient HCCs, the lower methylation level in KRT19 promoter was observed in K19-proficient HCCs (P<.0001). Instead, HCC with genome-wide hypermethylation in LINE-1 was frequently observed in K19-proficient HCCs (P = 0.0079). Additionally, K19 proficiency was associated with K7 proficiency (P = 0.043), and reduced both E-cadherin and HepPar-1 expression (P = 0.043 and<.0001, respectively).

Conclusions

K19-proficient HCC showed the poor prognosis owing to extrahepatic recurrence and the molecular signatures were different from K19-deficeint HCC, providing novel insights of heterogeneity underlying development of HCC with extrahepatic metastasis.

Legal entity responsible for the study

Takeshi Nagasaka

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1731P - Rare malignancy rare site: Extranodal lymphomas (ID 624)

Presentation Number
1731P
Lecture Time
13:15 - 13:15
Speakers
  • S. Gupta
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Extranodal Non Hodgkins Lymphoma (NHL) constitutes one quarter of all NHL cases. Oral cavity and breast represent 2 such uncommon sites.

Methods

Clinical and treatment details of patients with extranodal lymphomas who underwent treatment at our center were collected. Diagnosis was established by excision biopsy or by punch biopsy from the lesion. Histopathological examination (HPE) and immunohistochemistry (IHC) were performed. For staging, patients underwent positron emission tomography (PET) with computed tomography (CT) scan of the whole body.

Results

Case 1: A 32-year old male presented with complaints on the right-side base of tongue and difficulty in swallowing since 20 days. On examination, a nodular swelling was seen over the right side posterior one third of tongue going up to base of tongue. Punch biopsy from the tongue lesion was suggestive of NH. IHC showed CD 3 - positive, CD 30 - positive, ALK 1 - positive and CD 20 - positive with a final report of anaplastic lymphoma kinase (ALK) positive ALCL. Whole body PET-CT showed localized metabolic uptake in the tongue and in the right cervical lymph node - level II. A final diagnosis of ALK positive ALCL - stage IE was made. Case 2: A 22-year old female presented with complaints of lump in the right breast since 10 weeks. On examination there was a lump in the right breast measuring 3 x 2 cm in the lower outer quadrant and no other palpable lymphadenopathy. On evaluation, wide local excision of the lump was suggestive of a round cell tumor. IHC done showed the neoplastic round cells to be positive for CD 20, PAX5, CD 10 with a Ki 67 of 80% and BCL 2, MUM 1 - negative. Bone marrow examination was positive for lymphomatous deposits. Whole body PET-CT showed metabolically active sub-centimetric right axillary lymph node enlargement with diffuse hypermetabolism along axial bone marrow.

Conclusions

Only 11 previous cases of oral ALCL have been reported. Primary breast lymphoma is a rare disease, accounting for only 0.4-0.5% of all breast malignancies, 0.38-0.7% of all NHL. Extranodal NHL can occur at any site and keeping an open mind is the most important pre-requisite for making a diagnosis. Modern diagnostic tools such as IHC is mandatory for diagnosis and management of extranodal NHL.

Clinical trial identification

Not applicable

Legal entity responsible for the study

Haryana Medical Council

Funding

None

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster display session Poster Display session

1732P - Descriptive analysis of families with TP53 mutations: Is there a genotype/phenotype correlation? (ID 5126)

Presentation Number
1732P
Lecture Time
13:15 - 13:15
Speakers
  • J. Silva
Session Name
Location
Hall 8, Madrid, Madrid, Spain
Date
11.09.2017
Time
13:15 - 14:15

Abstract

Background

Li-Fraumeni syndrome (LFS) is a rare and serious hereditary cancer syndrome caused by germline mutations in the TP53 gene. Our objective is to review the molecular and clinical characteristics of our LFS families.

Methods

Retrospective descriptive analysis. Molecular germinal diagnosis was done either by Sanger or NG Sequencing (Trusight Cancer, Illumina); Large Genomic rearrangements were tested by MLPA (MRC Holland).

Results

Among 4952 non-related families registered in our multidisciplinary program, 395 are BRCA1/2 families and 36 haver other molecular diagnoses with 7/36 confirmed TP53 families. Twenty-nine pts were reviewed, including 2 male carriers, (36, 40 years) that haven’t developed cancers yet. Forty-one tumours were registered (median of 1,41 tumours/carrier, 0–4). Female/male was 2:1. Median age for the first tumour was 24 years (1-45) in the index cases and 35 (1,5-61) in relatives (p > 0,05). Breast cancer (BC) (34% of cancers/48% pts) and sarcomas (31% of cancers/44% pts) were the most common malignancies. For BC cases hormone receptor status was confirmed in 8/14 (positive in 6/8, simultaneous HER2 positivity in 2 cases). Median age of death was 40 (34-58). Most mutations were missense (5 - 2 dominant-negative affecting the DNA binding domain: c.743G>A, p.R248Q and c.725G>A, p.C242Y), none was recurrent and the only frequent mutation observed was c.743G>A, p.R248Q. One of the missense and the 2 frameshift mutations have never been described as germinal: c.481G>A, p.A161T; c.86del, p.N29Tfs*15 and c.990del, p.Q331Rfs*14 Chompret criteria were met in 6/7 (85%) of cases and didn't identitfy a breast cancer only family, with 3 consecutive generations affected.

Conclusions

Our data confirms the heterogeneity and complexity of malignancies and mutations in LSF. Breast cancer and sarcomas were the most frequent cancers and missense, non-recurrent mutations were mostly observed. In this study c.481G>A, p.A161T; c.86del, p.N29Tfs*15 and c.990del, p.Q331Rfs*14 are, for the first time, identified as germinal mutations. At this time no genotype-to-phenotype correlation could be confirmed. Chompret’s criteria had only 85% sensitivity for the identification of TP53 families.

Legal entity responsible for the study

Instituto Português de Oncologia de Lisboa Francisco Gentil

Funding

None

Disclosure

J.P. Silva: Travel grants for oncology Congresses: Bristol Meyers-Squibb.

\r\n

All other authors have declared no conflicts of interest.

Disclosure

J.P. Silva: Travel grants for oncology Congresses: Bristol Meyers-Squibb.

All other authors have declared no conflicts of interest.

Collapse