All times are listed in CEST (Central European Summer Time)

Browsing Over 1843 Presentations

Sort By Upcoming First
Poster Display session

BASIC SCIENCE

Date
17.09.2020
Poster Display session

10P - Targeting Angiomotin-p130 reverse resistance to antiangiogenic therapies in breast cancer by inhibition of vasculogenic mimicry

Presentation Number
10P
Speakers
  • Yanwei Shen (Xi'an, China)
Date
17.09.2020

Abstract

Background

Breast cancer (BC) was shown to relapse faster and displayed therapeutic resistance to antiangiogenic therapies (AATs) through an alternative tumor cell-driven mechanism of neovascularization called vascular mimicry (VM). This study aims to ascertain the correlation between Angiomotin-p130 expressing and the formation of VM in antiangiogenic therapy.

Methods

Human BC tissue sections and tissue array were used to ascertain the clinical relevance of Angiomotin-p130 expressing in tumor cells and the formation of VM. We utilized MCF7 and MD-MBA-231 human tumor cells to observe VM in an orthotopic BC model and bevacizumab (anti-VEGF) was used to identify the resistance mechanism of antiangiogenic therapies in breast cancer. Later, tube formation assay was used to explore the efficacy of bevacizumab and anlotinib (anti-VEGFR,PDGFR,FGFR and c-Kit) in the inhibition of VM.

Results

According to human datasets and histological analysis Angiomotin-p130 was identified as a bonafide candidate to regulation VM formation through in vitro and in vivo experiments. Stable overexpression of Angiomotin-p130 in tumor cells led to decreased tumor growth as well as incomplete VM structures in the animal models. We identified highly downregulated Angiomotin-p130 in human breast tumor cells and AAT-treated orthotopic mammary tumors. AAT-treated (bevacizumab and anlotinib) tumors displayed a higher number of Angiomotin-p130-negative tumor cells than endothelial-like phenotypes. Tube formation assay showed that the bevacizumab and anlotinib had no significant difference in inhibition on VM, though anlotinib inhibited the proliferation of tumor cells.

Conclusions

The present study suggests that tumor cell autonomous Angiomotin-p130 pathway may play an important role in AAT-mediated resistance and VM formation in BC. Therefore, targeting Angiomotin-p130 can be combined with standard antiangiogenic therapies to improve the therapeutic outcomes in clinical trials.

Legal entity responsible for the study

Yanwei Shen.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display session

11P - Preclinical evaluation of DS-2087b, a novel and selective inhibitor of EGFR/HER2 exon 20 insertions

Presentation Number
11P
Speakers
  • Yasuhito Nagamoto (Tokyo, Japan)
Date
17.09.2020

Abstract

Background

In-frame insertions in exon 20 of the epidermal growth factor receptor (EGFR) or human epidermal growth factor receptor 2 (HER2) gene are oncogenic drivers in non-small cell lung cancer (NSCLC), breast cancer, and others. However, no pharmacologic treatments are available for EGFR/HER2 exon 20 insertions (ex20ins), and effective treatment should avoid the inhibition of wild-type (WT) EGFR, which causes dose-limiting toxicities, such as rash and diarrhea. Here we describe DS-2087b, a novel oral, potent, and selective inhibitor of EGFR/HER2 ex20ins.

Methods

We evaluated the inhibitory effects of DS-2087b and poziotinib, an investigational tyrosine kinase inhibitor against Ba/F3 cells harboring WT EGFR and against EGFR/HER2 ex20ins mutations, including EGFR V769_D770 insertion ASV (EGFR ins.ASV); EGFR D770_N771 insertion SVD (EGFR ins.SVD); and HER2 A775_G776 insertion YVMA (HER2 ins.YVMA). Moreover, we measured the antitumor activity of DS-2087b in an NSCLC patient-derived xenograft (PDX) model with EGFR ins.ASV.

Results

DS-2087b inhibited the proliferation of Ba/F3 cells expressing EGFR ins.ASV, EGFR ins.SVD, and HER2 ins.YVMA with 50% maximal inhibition of cell proliferation (GI50) values of 23.5, 21.3, and 29.0 nM, respectively, but the GI50 value for Ba/F3 cells expressing EGFR WT was 368.9 nM. Poziotinib showed similar GI50 values between EGFR/HER2 ex20ins and WT EGFR. In Ba/F3 allograft models with EGFR ins.ASV, EGFR ins.SVD, and HER2 ins.YVMA and the PDX model with EGFR ins.ASV, DS-2087b showed remarkable antitumor effects in a dose-dependent manner without body weight loss.

Conclusions

In in vitro and in vivo preclinical assays, DS-2087b is effective against EGFR/HER2 ex20ins and also showed selectivity over WT EGFR. DS-2087b may show clinical utility in patients who harbor EGFR/HER2 ex20ins mutations and reduced toxicities caused by WT EGFR inhibition.

Legal entity responsible for the study

Yuki Abe.

Funding

Daiichi Sankyo Company, Limited.

Disclosure

Y. Nagamoto: Full/Part-time employment: Daiichi Sankyo Company, Limited. M. Miyamoto: Full/Part-time employment: Daiichi Sankyo Company, Limited. N. Togashi: Full/Part-time employment: Daiichi Sankyo Company, Limited. T. Taira: Full/Part-time employment: Daiichi Sankyo Company, Limited. T. Jimbo: Full/Part-time employment: Daiichi Sankyo Company, Limited. T. Isoyama: Full/Part-time employment: Daiichi Sankyo Company, Limited. M. Takahashi: Full/Part-time employment: Daiichi Sankyo Company, Limited. K. Takeuchi: Full/Part-time employment: Daiichi Sankyo Company, Limited. K-I. Yoshida: Full/Part-time employment: Daiichi Sankyo Company, Limited. S. Higuchi: Full/Part-time employment: Daiichi Sankyo Company, Limited. T. Seki: Full/Part-time employment: Daiichi Sankyo Company, Limited. Y. Abe: Full/Part-time employment: Daiichi Sankyo Company, Limited.

Collapse
Poster Display session

12P - Genome mining-aided biosynthesis overcomes Temozolomide-resistant glioblastoma via negatively regulating cIAP-mediated degradation of PACS2 and ER-mitochondria contact sites

Presentation Number
12P
Speakers
  • Lei Chen (Nanjing, China)
Date
17.09.2020

Abstract

Background

Glioblastoma (GBM) is the most aggressive tumour originating from the brain. At present, surgery combined with radiotherapy and chemotherapy is the main therapy modality for GBM. However, the prognosis for patients with GBM still remains poor. It is known that malignant glioblastoma cells are more susceptible to unfolded protein response (UPR) and ERs because of high level of protein synthesis. Therefore, the potential vulnerability of the ER in GBM cells via the disturbance of UPR may hold therapeutic promise for GBM.

Methods

We isolated a class of aspergin derivatives from Eurotium cristatum through polyketide synthase (PKS) and phosphatase (PTase)-dependent genome mining-aided biosynthesis. A methodology was developed to screen anti-glioma compounds in primary culture cells as well as Temozolomide (TMZ)-resistant ERs-sensitive cells. Apoptotic ability of decitabine (DAC) was tested by electron microscopy. Confocal scanning was performed to investigate the DAC-induced UPR and harmful autophagy in ER. Seahorse Mito-Stress test and electron transport chain (ETC) activity assay were used to evaluate the oxygen consumption rate, ATP production, and OXPHOS effect of DAC-treated cells. Xenograft mouse models was established to confirm the anti-glioma effect of DAC on tumour growth.

Results

Firstly, the data indicated that DAC induced dysfunction of ER-mitochondria communication as well as ERs-dependent apoptosis via disrupting the mitochondrion-associated ER membrane (MAM) machinery. Secondly, it was demonstrated that DAC blocked degradation of PACS2 resulting in over-link of MAM networks and destabilization of the ER-mitochondria crosstalk. Thirdly, accumulation of PACS2 promoted translocation of Bid to mitochondria, thereby facilitating disturbances in OXPHOS-dependent energy metabolism. Furthermore, we observed high cIAP1 and low PACS2 expression levels in glioma tissues, Finally, it was confirmed that DAC-induced suppression of PACS2 degradation contributed to anti-glioma activity in vivo.

Conclusions

In conclusion, these findings suggested that DAC might be a candidate for the treatment of ERs-sensitive malignant gliomas through negatively regulating cIAP-mediated degradation of PACS2. Moreover, our study provides new insights that ER-mitochondria contact sites might be therapeutic targets of GBM.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display session

13P - Synergistic activity of PARP inhibitor and ATR inhibitor in melanoma cell lines may depend on BRAF-V600 mutation status

Presentation Number
13P
Speakers
  • Emilio F. Giunta (Napoli, Italy)
Date
17.09.2020

Abstract

Background

Metastatic melanoma, despite recent progresses in its treatment, is still characterized by high mortality. High genomic instability found in human melanoma represents an important pathological feature, explaining high mutational burden and great response to immune checkpoint inhibitors. However, genomic instability could also represent a potential target for drugs inhibiting DNA repair mechanisms. Treatment with PARP inhibitors, alone or in combination with chemotherapy, did not show significant activity in melanoma patients. ATR (ataxia telangiectasia and Rad3 related), a protein involved in DNA repair mechanism in response to double strand breaks, is a new target in cancer therapy. Aim of this work is to evaluate the activity AZD6738, an ATR inhibitor, alone or in combination with olaparib, a PARP inhibitor, in melanoma cell lines.

Methods

Two BRAF-V600 mutant melanoma cell lines (A375 and SAN) and their in-vitro vemurafenib and cobimetinib resistant derivatives (A375-VRCR and SAN-VRCR, respectively), two NRAS mutant melanoma cell lines (SK-MEL-2 and WM1366) and one BRAF/NRAS wild type melanoma cell line (MeWo) have been treated with AZD6738, olaparib and their combination. MTT proliferation assays and colony formation assays have been performed to evaluate in-vitro activity of these drugs.

Results

MTT assays have shown that all four BRAF-V600 mutant cell lines are equally sensitive to AZD6738 (IC50: 1-1,5 μM), independently from acquired in-vitro resistance to vemurafenib and cobimetinib. Combination of AZD6738 and olaparib has shown drug synergism in all these four cell lines. This result has been confirmed in colony formation assays in which, after 240 hours exposure to the combination of AZD6738 (0,4 μM) and olaparib (2 μM), a high mortality (>94%) has been observed in all 4 cell lines, versus a low mortality (1-15%) when treated with single drugs (p<0.01). However, no synergistic activity has been found in the BRAF/NRAS wild type and the two NRAS mutant cell lines.

Conclusions

Our work has highlighted an in-vitro synergistic activity of AZD6738 and olaparib only in BRAF-V600 mutant cell lines, either naïve or with acquired in-vitro resistance to BRAF and MEK inhibitors.

Legal entity responsible for the study

Dipartimento di Medicina di Precisione, Università degli Studi della Campania \"Luigi Vanvitelli\".

Funding

Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli.

Disclosure

P.P. Vitiello: Advisory/Consultancy: Biocartis. E. Martinelli: Advisory/Consultancy: Amgen; Advisory/Consultancy: AstraZeneca; Advisory/Consultancy: Bayer; Advisory/Consultancy: Merck; Advisory/Consultancy: Roche; Advisory/Consultancy: Servier; Advisory/Consultancy: Sanofi. G. Martini: Advisory/Consultancy, Research grant/Funding (institution): Roche; Advisory/Consultancy, Research grant/Funding (institution): Amgen; Advisory/Consultancy, Research grant/Funding (institution): Merck; Advisory/Consultancy: Pfizer; Speaker Bureau/Expert testimony: Sanofi; Advisory/Consultancy, Research grant/Funding (institution): Bayer; Speaker Bureau/Expert testimony: Servier; Advisory/Consultancy: BMS; Advisory/Consultancy: Cellgene; Advisory/Consultancy: Lilly; Research grant/Funding (institution): AstraZeneca; Research grant/Funding (institution): Takeda. T. Troiani: Research grant/Funding (institution): Roche; Research grant/Funding (institution): Merck; Research grant/Funding (institution): Sanofi; Research grant/Funding (institution): Servier; Research grant/Funding (institution): Novartis; Research grant/Funding (institution): Bayer. All other authors have declared no conflicts of interest.

Collapse
Poster Display session

14P - Modulation of MCF-7 cell sensitivity to anticancer agents by targeting glycolysis

Presentation Number
14P
Speakers
  • Alexander M. Scherbakov (Moscow, Russian Federation)
Date
17.09.2020

Abstract

Background

Energy imbalance is one of the tumor cells' key properties in some cases supporting rapid progression and resistance to therapies. Simultaneous effects on glycolysis and other signaling pathways seem to be a promising strategy for anticancer therapy. Objectives: evaluation of the effects of glucose deprivation and glycolysis inhibitor (2-Deoxy-D-Glucose, 2DOG) on the antiproliferative activities of various anticancer agents, searching for promising combinations.

Methods

To simulate glucose deprivation DMEM medium with or without 1, 4.5 g / L glucose was used. Treatment with 2DOG was applied to inhibit glycolysis. Cell viability was estimated by MTT. The activity of the estrogen receptor alpha was assessed by gene reporter analysis. Immunoblotting was used to evaluate protein expression (AMPK, Akt).

Results

Under glucose deprivation, we showed that the effect of antiproliferative agents from various classes on MCF-7 cells almost does not change. No enhanced effect of edelfosin, tamoxifen, apigenin, genistein, doxorubicin, and 1-pentafluorophenylamido-5-sulfonamidoindane (CAIX inhibitor), everolimus, wortmannin was found. Glucose deprivation and 2DOG sensitized MCF-7 cells to two only drugs, metformin and oligomycin A. Glucose deprivation causes a 1000-fold increase in the antiproliferative effects of oligomycin A in hormone-dependent MCF-7 breast cancer cells. Moreover, glucose withdrawal leads to a significant compensatory activation of Akt, one of the main anti-apoptotic proteins, while oligomycin A further enhances Akt activity. AMPK, an energy cell sensor, is activated by 1 nM oligomycin A only under glucose starvation. Oligomycin A and metformin show more pronounced antiestrogenic effects during glucose deprivation.

Conclusions

Glucose starvation and pharmacological inhibition of glycolysis are of interest for harnessing the antitumor action of oligomycin A and metformin in ERα-positive breast cancer, including through improving their antiestrogenic potencies. This project was supported by the Russian Foundation for Basic Research (agreements 18-29-09017, studies of ERα signaling and 19-015-00058, studies of everolimus) and the Russian Science Foundation (agreement 20-13-00402, studies of CAIX inhibitor).

Legal entity responsible for the study

The authors.

Funding

Russian Foundation for Basic Research (agreements 18-29-09017, studies of ERα signaling and 19-015-00058, studies of everolimus) and the Russian Science Foundation (agreement 20-13-00402, studies of CAIX inhibitor).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display session

15P - Study on the mechanism of apatinib reversing tamoxifen resistance in breast cancer

Presentation Number
15P
Speakers
  • Qingxia Li (Shijiazhuang, China)
Date
17.09.2020

Abstract

Background

Tamoxifen treatment revolutionized the management of estrogen receptor (ER) positive breast cancers, however drug resistance compromises its clinical efficacy. We here investigate effect of VEGFR signal pathway inhibitor apatinib on tamoxifen resistant breast cancer cell line and its possible mechanism.

Methods

The morphological differences between parent breast cancer cell line MCF7 and tamoxifen resistant cell line LCC2 were observed. CCK-8 assay was used to determine the proliferation inhibition rate of MCF7 and LCC2 cells treated with different concentrations of 4-hydroxy-tamoxifen (4OHT), and proliferation inhibition rate of LCC2 cells treated with different concentrations of apatinib or 4OHT combined with 10 μM apatinib for 48 hours. The apoptosis rate of LCC2 cells was detected by flow cytometry (FCM). Western blotting was used to detect the expression of ERα, PI3K, Akt and p-Akt pathway proteins in MCF7 and LCC2 cells, and the changes of Bcl-2, Bax apoptosis protein and ER α, PI3K, Akt and p-Akt protein expression in LCC2 cells treated with three different treatments.

Results

The morphology of MCF7 cells is slightly different from LCC2 cells: LCC2 cells are fusiform or triangular with a slightly shorter long axis. CCK-8 assay showed that the inhibition effect on LCC2 cells with the same concentration of tamoxifen was less than that on MCF7 cells. And LCC2 cell inhibition rate increased with the increase of tamoxifen concentration after were treated with 10 μ M Apatinib for 48 hours, which showed a significant synergistic enhancement between them (P < 0.05). FCM showed that the apoptosis rate in the experimental group was higher than control group (P < 0.05), and combination group was higher than tamoxifen group (P < 0.01). Western blotting showed that the expression of ER α, PI3K, Akt and p-Akt protein in the LCC2 was higher than MCF7 (P < 0.05). And in the combination group was less than tamoxifen group, while the expression of Bax protein was increased (P < 0.01).

Conclusions

Apatinib can enhance the re-sensitivity of resistant cells to tamoxifen, which may be related to the up-regulation of Bax protein and down-regulation of Bcl-2 protein and the expression of estrogen receptor pathway proteins ERα, PI3K, Akt and p-Akt.

Legal entity responsible for the study

Qingxia Li.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display session

16P - Development of novel combination of Imatinib and Thymoquinone on colon cancer cell line

Presentation Number
16P
Speakers
  • Mervat M. Omran (Cairo, Egypt)
Date
17.09.2020

Abstract

Background

Imatinib (IM) is a pharmaceutical drug that inhibits tyrosine kinases enzymes which are responsible for the activation of many proteins by signal transduction cascades as c-Abl, c-Kit and the PDGF receptor. Thymoquinone (TQ) is an active constituent of Nigella Sativa seeds. TQ benefits are attributed to its medicinal uses as antioxidant, anticancer and antimicrobial agent. It also possesses anti-inflammatory and hepatoprotective effects. This study aimed to investigate the chemo-modulatory potential of TQ to IM in colon cancer cells (HCT-116).

Methods

Colon cancer cell line (HCT-116) was treated with different concentrations (7.5-120 μM) of IM and/or TQ for 24, 48, 72 h. Cytotoxic and apoptotic potentials of combination therapy were carried out using different techniques. IM-TQ interaction was analyzed using compusyn software.

Results

Our data showed that TQ synergistically augments the cytotoxic activity of IM in HCT-116 cells. The IC50 values for IM were significantly reduced to 7.3, 7 and 5.5 μM after combination with (10 μM) TQ and to 5.8, 5.6 and 4.6 μM after combination with (20 μM) TQ for 24, 48 and 72 h. The CI and DRI values indicate a significant synergism in HCT-116 cells at 24, 48 and 72 h of treatment. The combination of IM with 5 μM TQ showed a marginal effect after 24 and 48 h of treatment, but after 72 h of treatment TQ enhances cytotoxicity of IM on HCT-116 cells. Further investigation by using flow cytometry and real time PCR analysis showed that the TQ sensitized HCT-116 cells to IM therapy. TQ induced apoptotic effect of IM via down regulation of anti-apoptotic BCL-2 (P=0.004) and BCL-XL(P=0.01) and upregulation of tumor suppressor PAR-4 (P=0.002) and BAX(P<0.001).

Conclusions

The current data demonstrates the promising role of Thymoquinone in potentiate chemotherapeutic effects of Imatinib on colon cancer cells.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display session

17P - Engineering and expression of constitutive activators of the PD-1 and LAG-3 signaling pathways

Presentation Number
17P
Speakers
  • Luisa Chocarro (Pamplona, Spain)
Date
17.09.2020

Abstract

Background

The lack of PD-1 or LAG-3 mutants with constitutive inhibitory activities prevents a systematic approach to dissect all the intracellular events regulated by PD-1 and LAG-3 that establish a strong T cell dysfunctionality in patients with intrinsic resistance to PD-1 therapies (Zuazo et al., 2019). It is thought that PD-1 and LAG-3 form a supramolecular complex together with TCR components in order to exert their inhibitory activity over TCR signal transduction. So far, no PD-1 and LAG-3 mutants with constitutive signalling activities have been described. Here we have constructed molecules that provide an initial TCR-dependent signal in T cells that lead to a sustained inhibitory activity of PD-1 and LAG-3.

Methods

To construct molecules with constitutive PD-1 and LAG-3 signaling activities in T cells, we replaced their immunoglobulin domains with a sequence encoding a single chain antibody that binds CD3. In addition, the coding sequence for GFP and mCherry were introduced at their c-terminus to track their expression in T cells. These fusion genes were cloned into the pDUAL lentivector expression system, which co-express antibiotic resistance for selection of cells lines. When expressed in T cells, the engagement with CD3 will trigger a TCR signal in neighbouring T cells, and these molecules will then transmit either PD-1 or LAG-3 signals. To test their expression, human T cells were transduced with lentivectors expressing these constructs singly or in combination, selected with antibiotics and visualized in living cells by fluorescence microscopy.

Results

Both constructs showed strong expression levels, with high accumulation especially at focal points of cell-to-cell contacts as would be expected for CD3-binding molecules. Similar expression patterns were observed in primary human T cells after lentivector modification. The growth of Jurkat cell lines expressing these molecules was evidently delayed compared to control untransduced T cells and to T cells constitutively expressing just the CD3-activating construct.

Conclusions

These results showed that these molecules had functional PD-1 and LAG-3 signaling in T cells leading to reduced cell growth. This will allow to study the reasons behind the intrinsic resistance to PD-1 blockade.

Legal entity responsible for the study

OncoImmunology Research Unit, Navarrabiomed, Instituto de Investigaciones Sanitarias de Navarra (IdISNA), UPNA.

Funding

This research was supported by Asociación Española Contra el Cáncer (AECC, PROYE16001ESCO); Instituto de Salud Carlos III, Spain (FIS project grant PI17/02119); Gobierno de Navarra Biomedicine Project grant (BMED 050-2019); TRANSPOCART (Instituto de Salud Carlos III); “Precipita” Crowdfunding grant (FECYT); Crowdfunding grant from Sociedad Española de Inmunología (SEI); DESCARTHES project grant (Industry department, Government of Navarre).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display session

18P - Prolonged in-use stability of reconstituted atezolizumab in commercial intravenous bags

Presentation Number
18P
Speakers
  • Ada Hui (San Francisco, United States of America)
Date
17.09.2020

Abstract

Background

Atezolizumab (atezo) is administered by intravenous (IV) infusion after dilution in IV infusion bags containing 0.9% sodium chloride (NaCI) solution. The objective of this study was to evaluate the physicochemical stability of diluted atezo in IV infusion bags stored for an extended period of time.

Methods

Two types of IV infusion bags with product-contact surfaces comprised of polyvinyl chloride (PVC) and polyolefin were tested. Polyolefin is a copolymer of polyethylene and polypropylene, so is abbreviated as PO-PE-PP here. Under aseptic conditions, atezo was withdrawn from commercial vials (840 mg or 1200 mg) and added to 100 mL or 250 mL IV infusion bags containing 0.9% NaCl with target concentrations of 2.4, 9.6 and 16.8 mg/mL. Bags containing diluted atezo were stored at 30°C for 24 hours with exposure to ambient light, then at 2–8°C for up to 12 months, protected from light. 10 mL samples were withdrawn at selected time points and evaluated by analytical assays for colour, opalescence and clarity; visible and sub-visible particulates; pH; protein content, by UV spectrophotometric analysis; size exclusion high-performance liquid chromatography (SE-HPLC); ion exchange high-performance liquid chromatography (IE-HPLC); non reduced capillary electrophoresis-sodium dodecyl sulfate (NR-CE-SDS); and potency assays.

Results

There were no substantial changes in atezo product quality by any measure related to any analytical assay listed after storage for 24 hours at 30°C followed by 3 months at 2–8°C. No new peaks were observed on SE-HPLC, IE-HPLC or NR CE-SDS profiles of diluted atezo after the extended storage. Over 12 months of storage at 2–8°C, trends for increases in protein aggregates by SE-HPLC, increases in fragments by NR CE-SDS, and increases in acidic species by IE-HPLC were observed in some diluted samples.

Conclusions

With a comprehensive assessment of multiple atezo concentrations and storage conditions, the study demonstrates that atezo diluted in 0.9% NaCl over a concentration range of 2.4–16.8 mg/mL was physicochemically stable when stored for 24 hours at 30°C followed by 3 months at 2–8°C in both PVC and PO-PE-PP IV infusion bags.

Editorial acknowledgement

Support for third-party writing assistance for this abstract, furnished by Blair Jarvis, MSc, of Health Interactions, was provided by Genentech, Inc., South San Francisco, CA, USA.

Legal entity responsible for the study

Genentech, Inc.

Funding

Genentech, Inc.

Disclosure

A. Hui: Full/Part-time employment: Genentech, Inc.; Non-remunerated activity/ies, F. Hoffmann-La Roche Ltd – Third-party editing assistance: F. Hoffmann-La Roche, Basel, Switzerland. J. Yin: Full/Part-time employment: Genentech, Inc.; Non-remunerated activity/ies, F. Hoffmann-La Roche Ltd – Third-party editing assistance: F. Hoffmann-La Roche, Basel, Switzerland. W. Liu: Full/Part-time employment: Genentech, Inc.; Non-remunerated activity/ies, F. Hoffmann-La Roche Ltd – Third-party editing assistance: F. Hoffmann-La Roche, Basel, Switzerland. K. Zheng: Full/Part-time employment: Genentech, Inc.; Non-remunerated activity/ies, F. Hoffmann-La Roche Ltd – Third-party editing assistance: F. Hoffmann-La Roche, Basel, Switzerland.

Collapse
Poster Display session

19P - Deep eutectic solvent mixture formed from 2-deoxy-D-glucose and metformin targets cancer cell metabolism and induces apoptosis in breast cancer cell line xenografts

Presentation Number
19P
Speakers
  • Kai Reinikainen (Kauhava, Finland)
Date
17.09.2020

Abstract

Background

Cancer cell metabolism has been proposed as a new strategy to fight cancer. Elevated aerobic glycolysis of cancer cells for energy production provides a possibility for therapeutic intervention. 2-deoxy-D-glucose (2DG) and metformin (MET) have been used in combination to inhibit glycolysis and induce cell death in various tumour types. The required doses have exceeded that achievable in human plasma. Deep eutectic solvents (DES) are mixtures of solid compounds that form liquids due to a large depression of the melting point and possess unique properties such as pharmacological activity. DESs have been reported to demonstrate anticancer properties but have not previously been studied in vivo mouse models.

Methods

We investigated the preclinical efficacy of targeting the tumour bioenergetic pathway using conventional and DES mixtures of MET and 2DG in MDA-MB-231 (human breast adenocarcinoma) and UFH-001 (triple-negative breast cancer) cells. We evaluated the in vitro anti-tumour activity of the individual components MET and 2DG and the DES mixture for comparison. In addition, we examined in vivo efficacy using xenograft mouse models.

Results

2DG and MET alone were not sufficient to promote tumour cell death, reflecting the limited efficacy demonstrated in clinical trials. A combined use of 2DG and MET also failed to induce cell death. However, the DES mixture of 2DG and MET led to significant cell death associated with decrease in cellular ATP and sustained autophagy. DES mixtures had the highest impact on tumour cell viability, exceeding the effect of 2DG/MET as single agents or combinations at all clinically relevant concentrations.

Conclusions

We developed a DES mixture from 2DG/MET, which significantly induced apoptosis of cancer cells, exceeding the effect of single components or conventional mixtures. Deprivation of tumour bioenergetics by dual inhibition of energy pathways might be an effective novel therapeutic approach for human breast cancer tumours. The DES does not necessitate the use of toxic components or additional solvents and could be used to develop clinical applications for targeting breast cancer cell metabolism.

Legal entity responsible for the study

Chembrain LTD research organization.

Funding

Chembrain LTD research institution.

Disclosure

S. Vuoti: Full/Part-time employment, currently works at Sanofi Genzyme, however, the presented research is not related to Sanofi but was conducted in 2017-2018 before joining Sanofi: Sanofi Genzyme. All other authors have declared no conflicts of interest.

Collapse
Poster Display session

21P - Urethane-induced lung carcinogenesis in genetically edited C57Bl/6 mice with CHEK2 and GPRC5A heterozygous inactivating mutations

Presentation Number
21P
Speakers
  • Evgeny Imyanitov (Saint-Petersburg, Russian Federation)
Date
17.09.2020

Abstract

Background

Some lung cancer (LC) patients carry germline truncating mutations in DNA repair or other cancer-related genes, however it is unclear whether these allelic variants play a causative role in LC predisposition, or, alternatively, are occasionally present in these subjects due to a chance. Case-control comparison is often not efficient due to rarity of gene-inactivating variants in population, therefore alternative approaches are required. We evaluated whether germ-line inactivation of GPRC5A and CHEK2 genes facilitates LC in mice.

Methods

C57BL/6 mice with heterozygous inactivating mutations in the GPRC5A or CHEK2 genes were created using the CRISPR/Cas9 system. Donor eggs were obtained from the first generation of crossing mice of C57BL/6 (males) and CBA (females) inbred strains. Offspring mice F1 bearing inactivating mutations in GPRC5A and CHEK2 were used in further breeding with C57BL/6 wild-type strain for the study purposes. Altogether, 33 females and 21 males without mutations, 11 females and 19 males with mutations in CHEK2, and 13 females and 14 males with mutations in GPRC5A were analyzed. At the age of 3 months, mice were intraperitoneally injected with urethane (1 g/kg). The experiment was stopped after 45 weeks, all animals were autopsied and the lung tumor nodes were counted.

Results

In the wild-type (control) mice, the number of affected animals was 16/33 (48%) in females and 12/21 (57%) in males (pooled males/females: 28/54 (52%)). The rate of tumor development was not increased in CHEK2-heterozygous mice (females: 6/11 (55%); males: 7/19 (37%); pooled males/females: 13/30 (43%)). However, animals carrying GPRC5A heterozygous inactivating mutations showed a trend towards elevated occurrence of carcinogen-induced lung neoplasms (females: 9/13 (69%); males: 11/14 (79%); pooled males/females: 20/27 (74%); р = 0.06 when compared to controls).

Conclusions

These data support evidences for the involvement of GPRC5A gene in pathogenesis of lung cancer.

Legal entity responsible for the study

The authors.

Funding

This study has been supported by the Russian Science Foundation.

Disclosure

All authors have declared no conflicts of interest.

Collapse