All times are listed in CET (Central European Time).

 

Displaying One Session

Hall Bordeaux Mon, 06.03.2023 17:15 - 18:15
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Location
Hall Bordeaux
Chairs
  • Giuseppe Curigliano (Milan, Italy)
  • Elena Garralda Cabanas (Barcelona, Spain)
  • Ruth Plummer (Newcastle upon Tyne, United Kingdom)
  • Anastasios Stathis (Bellinzona, Switzerland)
  • Juanita S. Lopez (Sutton, United Kingdom)
  • Alison Schram (New York, United States of America)
  • Lillian L. Siu (Toronto, Canada)
Cocktail & Poster Display session Abstract session

Antibody-based therapies

Lecture Time
17:15 - 17:15
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Cocktail & Poster Display session Abstract session

2P - SOT102, a novel CLDN18.2-targeting antibody-drug conjugate for gastric and pancreatic cancer with a wide range of the tumor target expression

Presentation Number
2P
Lecture Time
17:15 - 17:35
Speakers
  • Radek Spisek (Prague, Czech Republic)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Radek Spisek (Prague, Czech Republic)

Abstract

Background

Claudin (CLDN) 18.2, a member of a large family of transmembrane proteins with distinct functions, has been shown to have high prevalence, predominantly in gastric and pancreatic cancer. Ectopic expression of CLDN18.2 was also described in certain proportion of ovarian cancer, NSCLC, hepatocellular cancer and colorectal cancer. Healthy tissue expression is restricted to the stomach epithelium.

Methods

SOT102 represents a novel CLDN18.2 targeting antibody-drug conjugate based on a proprietary monoclonal antibody conjugated to a derivative of PNU-159682 via site-specific sortase mediated conjugation. The CLDN18.2 protein sequence is highly conserved across species with a 100% identity in the targeted extracellular loop among rodents, cynomolgus monkeys and humans.

Results

SOT102 showed an excellent specificity for CLDN18.2 and strong binding to the target followed by an efficient tumor cell killing. Preferential binding to selected patient-derived tumor tissues was observed ex vivo when compared to the healthy stomach tissues from mice and cynomolgus monkeys. Single-agent therapeutic activity of SOT102 was demonstrated in numerous patient-derived xenograft models (gastric, pancreatic, liver, colon and lung adenocarcinomas). Complete responses were observed in all CLDN18.2 positive models, irrespective of the intensity of staining. models, independent of CLDN18.2 expression levels, ranging from low (IHC1+) to high (IHC3+), with minimum effective doses between 0.2 mg/kg and 0.6 mg/kg. An acceptable tolerability profile was observed in the toxicity studies at 10 mg/kg (mouse), 6 mg/kg (rat) and 1 mg/kg (cynomolgus monkey), providing a therapeutic index of approximately 10. SOT102 demonstrated favorable pharmacokinetic properties with the half-live in the range of 8 days and 13 days in cynomolgus monkeys and rats, respectively. SOT102 remains stable without any significant loss of the payload both in vitro and in animal models.

Conclusions

SOT102 represents a novel potent ADC with the potential to treat Claudin 18.2 expressing tumors irrespective of the intensity of expression. The first in human dose escalation trial has been initiated in patients with gastric and pancreatic cancer.

Clinical trial identification

NCT05525286.

Legal entity responsible for the study

Sotio Biotech.

Funding

Sotio Biotech.

Disclosure

R. Spisek: Financial Interests, Personal, Stocks/Shares: Sotio Biotech.

Collapse
Cocktail & Poster Display session Abstract session

3P - Correlation between antibody-drug conjugate (ADC) targetable antigen expression and occurrence of interstitial lung disease (ILD)

Presentation Number
3P
Lecture Time
17:35 - 17:35
Speakers
  • Solange Peters (Lausanne, Switzerland)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Solange Peters (Lausanne, Switzerland)
  • Kaushal Parikh (Rochester, United States of America)
  • Anastasios Dimou (Rochester, United States of America)
  • Aakash Desai (Jacksonville, United States of America)

Abstract

Background

Although approved for multiple tumor types, one of the most limiting toxicities of antibody-drug conjugates (ADC) is development of interstitial lung disease (ILD). With several ADCs currently under development, it is important to identify mechanism of ILD to optimize drug development. We examined the correlation between target antigen expression level in normal lung tissue and the occurrence of ILD for ADCs studied in lung cancer.

Methods

We performed a literature search of ADCs in lung cancer to identify different antigen targets currently under investigation. Data was abstracted for incidence of ILD from publically available trial results of phase I-II clinical trials of ADCs in non-small cell lung cancer (NSCLC) targeting antigens: ERBB2 (trastuzumab deruxetecan), ERBB3 (patritumab deruxetecan), TROP2 (datopotamab deruxetecan), CEACAM5 (tusamitamab ravtansine), and MET (Telisotuzumab Vedotin). We used the highest value of all grade ILD reported at any dose to maximize identification of any correlation with antigen expression. We obtained scRNA-seq expression [(normalized mean transcript protein expression (nMTP)] values in predominant lung cell type from the Human Protein Atlas version 22.0 using enrichment prediction score for each cell type profiled in the lung tissue.

Results

Across different ADCs, the incidence of all grade ILD ranged from 0-26.4%. All ADC target antigens were expressed in lung tissues with highest normalized transcript expression level within lung tissue found in alveolar cells (type 1 or type 2). Across various targets, nMTP expression ranged from 73.7-1549.8. In terms of correlation between target expression and incidence of ILD, no correlation was found between these 2 variables (Pearson correlation coefficient = -0.007).

Conclusions

No correlation was identified between scRNA-seq expression of ADC-targetable antigens on alveolar cells and incidence of ILD. This may imply and further provide support to ILD being a bystander effect rather than true on-target toxicity with ADCs. Further ADC development should focus on reducing bystander effects via selection of linker and payload classes least likely to cause ILD while optimizing dose and drug-to-antibody ratio.

Editorial acknowledgement

N/A

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

S. Peters: Financial Interests, Institutional, Advisory Board: Vaccibody, Takeda, Seattle Genetics, Sanofi, Roche/Genentech, Regeneron, Phosplatin Therapeutics, PharmaMar, Pfizer, Novartis, Mirati, Merck Serono, MSD, Janssen, Incyte, Illumina, IQVIA, GlaxoSmithKline, Gilhead, Genzyme, Foundation Medicine, F-Star, Eli Lilly, Debiopharm, Daiichi Sankyo, Boehringer Ingelheim, Blueprint Medicines, Biocartis, Bio Invent, BeiGene, Bayer, BMS, AstraZeneca, Arcus, Amgen, AbbVie, iTheos, Novocure; Financial Interests, Institutional, Invited Speaker: Takeda, Sanofi, Roche/Genentech, RTP, Pfizer, PRIME, PER, Novartis, Medscape, MSD, Imedex, Illumina, Fishawack, Eli Lilly, Ecancer, Boehringer Ingelheim, BMS, AstraZeneca, OncologyEducation, RMEI, Mirati; Financial Interests, Personal, Other, Associate Editor Annals of Oncology: Elsevier; Financial Interests, Institutional, Invited Speaker, MERMAID-1: AstraZeneca; Financial Interests, Institutional, Invited Speaker, MERMAID-2, POSEIDON, MYSTIC: AstraZeneca; Financial Interests, Institutional, Invited Speaker, Clinical Trial Steering committee CheckMate 743, CheckMate 73L, CheckMate 331 and 451: BMS; Financial Interests, Institutional, Invited Speaker, RELATIVITY 095: BMS; Financial Interests, Institutional, Invited Speaker, BGB-A317-A1217-301/AdvanTIG-301: BeiGene; Financial Interests, Institutional, Invited Speaker, Clinical Trial Chair ZEAL-1: GSK; Financial Interests, Institutional, Invited Speaker, Clinical Trial steering Committee PEARLS, MK-7684A: MSD; Financial Interests, Institutional, Invited Speaker, Clinical Trial Steering Committee SAPPHIRE: Mirati; Financial Interests, Institutional, Invited Speaker, LAGOON: Pharma Mar; Financial Interests, Institutional, Invited Speaker, phase 1/2 trials: Phosplatin Therapeutics; Financial Interests, Institutional, Invited Speaker, Clinical Trial Chair Skyscraper-01; chair ALEX; steering committee BFAST; Steering Committee BEAT-Meso; steering committee ImPower-030, IMforte: Roche/Genentech; Financial Interests, Institutional, Invited Speaker, Phase 2 Inupadenant with chemo: iTeos; Non-Financial Interests, Personal, Officer, ESMO President 2020-2022; Non-Financial Interests, Personal, Officer, Council Member & Scientific Committee Chair: ETOP/IBCSG Partners; Non-Financial Interests, Personal, Officer, Vice-President Lung Group: SAKK; Non-Financial Interests, Personal, Other, Involved in Swiss politics: Swiss Political Activities; Non-Financial Interests, Personal, Officer, President and Council Member: Ballet Béjart Lausanne Foundation; Non-Financial Interests, Personal, Principal Investigator, Involved in academic trials: ETOP / EORTC / SAKK; Non-Financial Interests, Personal, Member: Association of Swiss Physicians FMH (CH), ASCO, AACR, IASLC; Non-Financial Interests, Personal, Leadership Role, ESMO President: ESMO; Non-Financial Interests, Personal, Member, Vice-President Lung Group: SAKK; Non-Financial Interests, Personal, Leadership Role, Vice-President: SAMO; Non-Financial Interests, Personal, Member, Association of Swiss interns and residents: ASMAC/VSAO. A. Dimou: Financial Interests, Personal, Advisory Board: TP Therapeutics, Guardant; Financial Interests, Personal, Advisory Role: Intellisphere Llc. A. Desai: Financial Interests, Personal, Advisory Board: Amgen, Sanofi. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

4P - Optimizing utilization of antibody-drug conjugates in NSCLC by identification of subsets using RNA sequencing

Presentation Number
4P
Lecture Time
17:35 - 17:35
Speakers
  • Edwin Lin (Rochester, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Edwin Lin (Rochester, United States of America)
  • Ying-Chun Lo (Rochester, United States of America)
  • Kaushal Parikh (Rochester, United States of America)
  • Nina Smrecek (Rochester, United States of America)
  • Kayla Goliwas (Birmingham, United States of America)
  • Jessy Deshane (Birmingham, United States of America)
  • Bassel El-Rayes (Birmingham, United States of America)
  • Aakash Desai (Jacksonville, United States of America)

Abstract

Background

Antibody-drug conjugates (ADCs), composed of monoclonal antibodies linked to cytotoxic drugs, are currently approved and standard-of-care for many malignancies including primary lung adenocarcinoma (LUAD). With several ADCs targeting different tumor antigens currently in clinical trials, it is important to characterize ADC targets to optimize drug development for LUAD.

Methods

RNA-sequencing data for 537 primary LUAD and 59 normal lung tissue samples were obtained from The Cancer Genome Atlas. Profiles of ADC-targetable gene expression including ERBB2 (HER2), ERBB3 (HER3), TACSTD2 (Trop2), MET (c-Met), CEACAM5, CD276 (B7-H3) and NECTIN4 within each tumor were assessed by comparison of transcripts per million. Recurrent patterns of ADC-targetable gene expression profiles were identified by hierarchical clustering. Differential gene expression was performed using the DESeq2 software package.

Results

Differential gene expression analysis demonstrated higher expression of CEACAM5 (P=3.72E-53), TACSTD2 (P=9.62E-4) and MET (P=8.06E-10) in tumors compared to normal lung tissue. Interestingly, TACSTD2 (Trop-2) expression was inversely correlated with CEACAM5. On hierarchical clustering, we identified four distinct clusters based on ADC-targetable gene expression profiles: (1) CEACAM5 high/TACSTD2 low, (2) CEACAM5 low/TACSTD2 high, (3) MET high, (4) CEACAM5 mid/TACSTD2 mid. A subset of tumors had high expression of either CD276, ERBB2, or ERBB3; these tumors also had low expression of TACSTD2, CEACAM5, and MET.

Conclusions

In primary LUAD, CEACAM5, TACSTD2 and MET are significantly overexpressed in distinct segregable patterns. Particularly, CEACAM5 and TACSTD2 expression showed an inverse correlation and appeared to be nearly mutually exclusive biomarkers. This emphasizes the need for biomarker-based approaches for optimal selection of ADCs based on expression of target antigens.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

A. Desai: Financial Interests, Personal, Advisory Board: Amgen, Sanofi. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

5P - Tisotumab vedotin (TV) dose schedule optimization in non-cervical populations

Presentation Number
5P
Lecture Time
17:35 - 17:35
Speakers
  • Jenna Voellinger (Bothell, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Jenna Voellinger (Bothell, United States of America)
  • Chaitali Passey (Princeton, United States of America)
  • Yan Summer Feng (Princeton, United States of America)
  • Rudy Gunawan (Bothell, United States of America)
  • Leonid Gibiansky (North Potomac, United States of America)
  • Daniel Polhamus (Tariffville, United States of America)
  • Arnout Gerritsen (Utrecht, Denmark)
  • Christine O'Day (Bothell, United States of America)
  • Leonardo Nicacio (Bothell, United States of America)
  • Ibrahima Soumaoro (Princeton, United States of America)
  • Manish Gupta (Princeton, United States of America)
  • William Hanley (Bothell, United States of America)

Abstract

Background

TV, a tissue factor-directed antibody-drug conjugate, is approved under accelerated approval in the US at a dose of 2.0 mg/kg every 21 days (Q3W) for adult patients (pts) with recurrent or metastatic cervical cancer (r/mCC) who have progressed on or after chemotherapy. Based on TV pharmacokinetics (PK) and exposure-response relationships, we hypothesized that a higher dose intensity and optimized key PK parameters can lead to improvement in efficacy. Here, we report a pharmacometric approach to continuously optimize the TV dosing schedule in non-cervical solid tumors.

Methods

TV is being evaluated for pts with advanced solid tumors in 3 schedules: Q3W; Days 1, 8, and 15 of a 28-day cycle (3Q4W); and Days 1 and 15 of a 28-day cycle (Q2W). A population-PK-tumor growth inhibition (PPK-TGI) model was developed using available longitudinal tumor size data from 711 pts, using a wide range of doses and 3 schedules, to determine the effect of TV exposure on tumor dynamics. A Markov model was developed to characterize the occurrence, severity, and duration of ocular adverse events (OAEs). Alternate TV dosing regimen simulations were performed in non-cervical solid tumor populations.

Results

Compared with the approved 2.0 mg/kg Q3W regimen, the PPK model predicted a 26% increase in AUC12wks at 1.2 mg/kg 3Q4W, but DLTs were observed in pts with ovarian cancer. At a lower dose of 0.9 mg/kg 3Q4W, AUC12wks was predicted to decrease 12% and Cmax to decrease 54%; this regimen had a tolerable safety profile but modest antitumor activity. Alternatively, 1.7 mg/kg Q2W predicted a 24% increase in AUC12wks, 15% decrease in Cmax, and a higher Ctrough. PPK-TGI modeling predicted 1.7 mg/kg Q2W to result in more time above the predicted EC50 and a greater reduction in tumor size, thus potentially better efficacy. Markov modeling predicted a potentially higher rate of Grade ≥2 OAEs for 1.7 mg/kg Q2W compared with 2.0 mg/kg Q3W.

Conclusions

TV demonstrated a favorable benefit-risk profile in r/mCC at the approved 2.0 mg/kg Q3W regimen. Along with available data, our modeling approach suggests that a Q2W schedule may lead to improvements in efficacy and potential risk of a higher rate of Grade ≥2 OAEs. Currently, TV is being evaluated at 1.7 mg/kg Q2W in advanced solid tumors in the enrolling innovaTV 207 study.

Editorial acknowledgement

Writing and editorial assistance was provided by Kristoffer Myczek, PhD, and Stephanie Wamsley of Ashfield MedComms, an Inizio Company, and funded by Seagen, Inc.

Legal entity responsible for the study

Seagen, Inc. and Genmab.

Funding

Seagen, Inc. and Genmab.

Disclosure

J. Voellinger: Financial Interests, Personal, Full or part-time Employment: Seagen, Inc.; Financial Interests, Personal, Stocks/Shares: Seagen, Inc. C. Passey, Y.S. Feng, A. Gerritsen, I. Soumaoro, M. Gupta: Financial Interests, Personal, Full or part-time Employment: Genmab. R. Gunawan, C. O'Day, L. Nicacio, W. Hanley: Financial Interests, Personal, Full or part-time Employment: Seagen, Inc. L. Gibiansky: Financial Interests, Personal, Full or part-time Employment: QuantPharm Llc; Financial Interests, Personal, Other, Paid Consultant: Seagen, Inc., Genmab. D. Polhamus: Financial Interests, Personal, Full or part-time Employment: Metrum Research Group; Financial Interests, Personal, Other, Paid Consultant: Seagen, Inc., Genmab.

Collapse
Cocktail & Poster Display session Abstract session

6P - RC48-ADC for metastatic salivary duct carcinoma with HER2 expression: A single-center retrospective study

Presentation Number
6P
Lecture Time
17:35 - 17:35
Speakers
  • Jun Cao (Hangzhou, China)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Jun Cao (Hangzhou, China)
  • Qing Ji (Hangzhou, China)

Abstract

Background

Agents targeting HER2 arepromising and the best studied therapies in patients with HER2-positive salivary duct carcinoma (SDC). RC48-ADC (Disitamab vedotin) is a novel humanized anti-HER2 antibody-drug conjugate (ADC). But the efficacy and safety of RC48-ADC is still unknown in metastatic SDC with HER2-positive.

Methods

Eligible patients from Zhejiang Cancer Hospital were 35∼76 years old, with confirmed, histologically HER2 expression (IHC1+, 2+, 3+), metastatic SDC. Patients had at least one line of systemic chemotherapy. Patients received RC48-ADC at 1.5 or 2 mg/kg, every two weeks. Clinical efficacy and safety were assessed.

Results

This study enrolled HER2-expressing metastatic SDC patients from June 2022 to Dec 2022. 10 mSDC patients (8 males, 2 females) were enrolled. 90% patients had received ≥2 lines systemic chemotherapy. 80% patients had visceral metastases. As of 05 Jan 2023 (data cutoff), 1 patient achieved CR, 4 patients achieved PR, 4 patients achieved SD, and only 1 patient achieved PD. The overall confirmed DCR was 90%. Most common treatment-related AEs were hypoaesthesia (70%), asthenia (60%), leukopenia (30%), decreased appetite (20%), alopecia (10%). The grade≥3 TRAEs only included hypoaesthesia (30.0%) and neutropenia (10%).

Conclusions

RC48-ADC showed a promising efficacy with a manageable safety profile in HER2-expressing mSDC patients who had failed at least one line of systemic chemotherapy.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

7P - Global ring study determining reproducibility & comparability of CLDN18 testing assays in gastric cancer

Presentation Number
7P
Lecture Time
17:35 - 17:35
Speakers
  • Bharat Jasani (Penarth, United Kingdom)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Bharat Jasani (Penarth, United Kingdom)
  • Hans-Ulrich Schildhaus (Essen, Germany)
  • Andrew Dodson (London, United Kingdom)
  • Suzanne Parry (London, United Kingdom)
  • Scott Clare-Antony (Belfast, United Kingdom)
  • Neil Atkey (Belfast, United Kingdom)
  • Philippe Taniere (Birmingham, United Kingdom)

Abstract

Background

Claudin-18.2 (CLDN18.2) the dominant isoform of CLDN18 in gastric tissue is a highly specific tight junction protein of the gastric mucosa expressed in gastric cancer & has emerged as a promising drug target. Zolbetuximab, a monoclonal antibody (Ab) targeting CLDN18.2 is reported to improve progression free & overall survival in combination with chemotherapy in the first-line treatment of CLDN18.2-positive, HER2-negative, locally advanced unresectable or metastatic gastric (G) & gastroesophageal junction (GEJ) cancers in 2 randomized phase 3 studies. Both studies (SPOTLIGHT, GLOW) met their primary endpoints. Patient selection for CLDN18.2 in these studies used the Ventana CLDN18 (43-14A) assay. It is important in the clinical setting to ensure accurate & reproducible results in the diagnosis of G/GEJ cancer for treatment options. This global ring study was conducted to assess analytical reproducibility & concordance of this assay with 2 other CLDN18 Abs (LSBio LS–B16145-100 & Novus Biologicals NBP2-32002) stained on 3 platforms to confirm possible diagnostic testing methods.

Methods

Tissue microarray (TMA) of 15 gastric cancer samples in triplicate cores was provided to 27 labs across 11 countries. Each lab stained the TMAs using 2-3 CLDN18 Abs with optimized protocols. The TMAs, also stained by the central lab, were used to achieve consensus by expert review. Using agreed scoring algorithms as per the phase 3 studies, IHC scores were generated per core & the results were collated for statistical analysis with the consensus results.

Results

Show high concordance among the 3 Abs across the assessed parameters when performed on each of the 3 platforms, with the highest accuracy & sensitivity observed for Ventana Ab & LSBio Ab across all 3 platforms. Table: 7P

Precision Precision Precision Accuracy Accuracy Accuracy Sensitivity Sensitivity Sensitivity Specificity Specificity Specificity
Antibody Ventana Novus LSBio Ventana Novus LSBio Ventana Novus LSBio Ventana Novus LSBio
Platform
Ventana 0.96 0.88 0.94 0.95 0.88 0.88 0.93 0.67 0.85 0.97 0.96 0.92
Dako - 0.94 0.93 - 0.83 0.94 - 0.72 0.96 - 0.95 0.93
Leica - 0.79 0.97 - 0.94 0.93 - 0.89 0.91 - 0.95 0.96

Conclusions

The study shows multiple Abs applied on multiple platforms give highly reproducible and comparable results for detecting CLDN18.2 in gastric cancer providing a potential for their global adoption for CLDN18 testing.

Legal entity responsible for the study

Diaceutics PLC.

Funding

Astellas Pharma.

Disclosure

B. Jasani, A. Dodson, S. Parry, N. Atkey, P. Taniere: Non-Financial Interests, Institutional, Advisory Role, Performing consulting work on behalf of Astellas Pharma in regards to abstract being submitted’: Astellas Pharma. H. Schildhaus: Financial Interests, Institutional, Advisory Board: MSD, BMS, Novartis; Financial Interests, Institutional, Invited Speaker: AstraZeneca, Roche, Takeda, Agilent, ZytoVision; Financial Interests, Personal, Full or part-time Employment: Targos/DLS Inc.; Financial Interests, Institutional, Research Grant: Novartis; Financial Interests, Institutional, Funding: Blueprint medicines; Other, Personal, Other, Member of the QuIP Board (Quality Assurance Organisation): QuIP GmbH. S. Clare-Antony: Other, Institutional, Advisory Role, Performing consulting work on behalf of Astellas Pharma in regards to abstract being submitted: Astellas Pharma.

Collapse
Cocktail & Poster Display session Abstract session

8P - Targeting antiapoptotic Bcl-2 proteins with highly specific BH3 mimetics in solid tumors

Presentation Number
8P
Lecture Time
17:35 - 17:35
Speakers
  • Benjamin Sobol (Heidelberg, Germany)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Benjamin Sobol (Heidelberg, Germany)
  • Paula K. Hoffmeister (Heidelberg, Germany)
  • Anna-Lena Scherr (Heidelberg, Germany)
  • Annika Kessler (Heidelberg, Germany)
  • Luisa Nader (Heidelberg, Germany)
  • Nathalie Schmitt (Heidelberg, Germany)
  • Dirk Jäger (Heidelberg, Germany)
  • Stefan Welte (Heidelberg, Germany)
  • Katharina Seidensaal (Heidelberg, Germany)
  • Petros Christopoulos (Heidelberg, Germany)
  • Katharina Kriegsmann (Heidelberg, Germany)
  • Stefan Fröhling (Heidelberg, Germany)
  • Mark Kriegsmann (Heidelberg, Germany)
  • Bruno Köhler (Heidelberg, Germany)

Abstract

Background

Evasion of apoptosis is a hallmark of cancer survival and a reason for acquired resistance towards standard treatment, making it one of the challenges in modern cancer therapy. The antiapoptotic proteins Bcl-2, Bcl-xL and Mcl-1, might be key to break such a resistance. In this study we evaluated the effects of highly specific inhibitors for Bcl-xL (WEHI-539), Bcl-2 (ABT-199), and Mcl-1 (S63845) as well as dual inhibition of Bcl-xL/Bcl-2 (ABT-263) in commonly occurring solid tumors.

Methods

Non-Small Cell Lung Cancer (NSCLC), Cholangiocarcinoma (CCA) and Breast Cancer cell lines were either exposed to fractionated photon radiation or chemotherapeutics (Epirubicin) as standard therapy with or without specific Bcl-2 protein inhibition. Protein expression was assessed via Western blots of cell lines. Effects on cell death were detected by flow cytometry measuring apoptosis.

Results

Dual Bcl-xL/Bcl-2 inhibition led to significantly higher cell death induction in combination with radiotherapy in NSCLC and with Epirubicin in triple-negative breast cancer. Sole inhibition of Bcl-xL caused an inferior but notable sensitization in both entities for standard therapy. In CCA, combination of fractionated photon beam radiation and specific Bcl-xL inhibition showed a significant increase of cell death in all four employed cell lines. In addition, the triple-negative breast cancer cell line benefited synergistically from combined therapy with Mcl-1 inhibition and Epirubicin. In NSCLC, no correlation between basal expression of Bcl-2 family proteins and response to therapy was found and proteins were not regulated upon irradiation. Upregulation of Mcl-1 may play a role in promoting radioresistance after specific Bcl-2 and Mcl-1 inhibition. Following Epirubicin treatment, breast cancer cell lines showed a downregulation of antiapoptotic Bcl-2 protein expressions.

Conclusions

Our findings indicate that among antiapoptotic Bcl-2 proteins, targeting Bcl-xL might break resistance to radiation in NSCLC, CCA and breast cancer in vitro. Especially for breast cancer, Mcl-1 could also be a promising target that needs to be further investigated.

Editorial acknowledgement

None.

Legal entity responsible for the study

The authors.

Funding

AbbVie, Brigitte and Dr. Konstanze Wegener foundation, the German Research Foundation (DFG, grant No. KO5205/1-1 and KO5205/3-1) and the German Cancer Aid (DKH, grant No. 70113593).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

9P - An in vivo model for therapeutic antibody efficacy evaluation: The chicken embryo’s CAM-based assay

Presentation Number
9P
Lecture Time
17:35 - 17:35
Speakers
  • Yan Wang (La Tronche, France)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Yan Wang (La Tronche, France)
  • Xavier Rousset (La Tronche, France)
  • Jean Viallet (La Tronche, France)
  • Chloé Prunier (La Tronche, France)

Abstract

Background

Since its introduction, xenografts on the chicken embryo’s ChorioAllantoic Membrane (CAM) has been proven extremely valuable for in vivo studies in cancerology. It is suitable to study tumor development, angiogenesis, malignant cell dissemination, chemotherapy efficacy and toxicity. Here, we demonstrate that this in ovo model, which has an active immune system, is useful for rapidly testing and comparing the efficacy of therapeutic antibodies, like anti-PD1/PD-L1 Abs, Antibody-drug Conjugates (ADCs), etc., on tumor growth and metastatic invasion.

Methods

Tumor cells are grafted on the CAM of chicken egg on day 9 of development. After engraftment, tumors are treated with therapeutic antibodies for 8 days, from a single injection for ADCs, to the treatment every other day for other types of antibodies. Maximal tested doses are scaled to the dose used in murine immune deficient models and in human (in mg/kg). Antibodies validated in ovo are FDA approved antibodies, against antigens expressed by tumor cells or immune cells, like immune checkpoint inhibitors (anti PD1/PD-L1 Abs) or ADCs (T-DM1).

Results

After treatment, the toxicity and the efficacy of tested Abs are analyzed and compared to non-treated control. The table presents the effect of antibodies on tumor growth in the CAM assay. A single antibody leads to different efficacy levels in function of the cancer types. Ab’s efficacy is also checked by measuring metastatic invasion in lower CAM (far from the tumor) and in embryonic tissues, immune cell infiltration or angiogenesis development. Deeper analysis can be performed, like transcriptomic analysis on the tumor to characterize mechanisms of action or more specific and/or fine effects on tumor cell physiology. Table: 9P

Effect of antibody therapies on tumor weight in ovo

Target Antibody Cell Line Cancer Type % Reduction of Tumor Weight
VEGF Bevacizumab MDA-MB-231 Breast -47.85%
VEGF Bevacizumab H460 Lung -28.44%
VEGF Bevacizumab Hep3B Liver -34.25%
PD-1 Pembrolizumab MDA-MB-231 Breast -12.14%
PD-1 Pembrolizumab H460 Lung -19.40%
PD-1 Nivolumab Hep3B Liver -1%
PD-L1 Atezolumab H460 Lung -24.79%
PD-L1 Atezolumab SNU182 Liver -22.48%
PD-L1 Atezolumab OPM-2 Myeloma -41.07%
HER-2 Trastuzumab N87 Gastric -19.17%
HER-2 (+ cytotosic agent) Trastuzumab Emtansine BT-474 Breast -58.94%
CD20 Rituximab Daudi Lymphoma -62.15%

Conclusions

Here we demonstrate that the CAM model is suitable to test therapeutic antibodies, immune therapies and ADCs in vivo. The presence of an active immune system allows to characterize of the effect of antibodies on tumor cell, microenvironment and the entire organism.

Legal entity responsible for the study

Inovotion.

Funding

Inovotion.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

Cell therapies

Lecture Time
17:35 - 17:35
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Cocktail & Poster Display session Abstract session

11P - Successful transfer and prolonged persistence of engineered lymphocytes with T-cell receptor targeting NY-ESO-1

Presentation Number
11P
Lecture Time
17:35 - 17:35
Speakers
  • Johnathan Arnon (Jerusalem, Israel)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Johnathan Arnon (Jerusalem, Israel)
  • Shiri Kein (Jerusalem, Israel)
  • Jonathan Cohen (Jerusalem, Palestinian Territory, Occupied)
  • Aviad Zick (Jerusalem, Israel)
  • Yonaton Zarbiv (Jerusalem, Israel)
  • Mordechai Avner (Jerusalem, Israel)
  • Yafit Halutsi (Jerusalem, Israel)
  • Polina Stepensky (Jerusalem, Israel)
  • Batia Avni (Jerusalem, Israel)
  • Sigal Grisariu (Jerusalem, Israel)
  • Anna Elia (Jerusalem, Israel)
  • Aron Popovtzer (Jerusalem, Israel)
  • Cyrille Cohen (Safed, Israel)
  • Michal Lotem (Jerusalem, Israel)

Abstract

Background

T-cells engineered to express antigen-specific T-cell receptors (TCR) recognize neoantigens derived from intracellular proteins and act against tumors. NY-ESO-1 is a cancer-testis antigen, exclusively re-expressed in cancer cells. Case-series of metastatic cancer patients treated with TCRs targeting NY-ESO-1 show a response rate of 50%. Third generation TCRs have increased stability and affinity in pre-clinical models. We present the results of the first three patients treated with specificity-enhanced third generation TCRs targeting NY-ESO-1, in an initial dose of a phase I-II trial.

Methods

We first demonstrated safety, persistence, and efficacy of the novel TCR in murine models. We then treated three patients: A 73-year-old male with choroidal melanoma (CM); A 43-year-old male with synovial sarcoma (SS); A 40-year-old female with triple-negative breast cancer (TNBC). Patients had suffered disease progression after resction of local disease and standard-of-care treatment for metastatic disease. Patients were ECOG 0-1, carriers of HLA-A*02:01 and had positive immunohistochemistry for NY-ESO-1 from tumor tissue. Between June and September 2022 patients underwent lympho-pheresis and T-cells were ex-vivo retrovirally-transduced to express HLA-A*02:01-restricted TCRs targeting NY-ESO-1. Patients underwent lympho-depletion (Cyclophosphamide 250mg/m2 and Fludarabine 25mg/m2 D1-3) and 5 days later received 1*109 engineered TCRs, followed by 3-days of continues IL-2 (18x106IU/24H).

Results

No severe adverse events were observed. Grade 1-2 fever and chills were resolved shortly after IL-2 cessation. At one week after administration, flow cytometry showed 5% to 44% of CD3+ positive lymphocytes in patients’ blood were engineered TCRs and remaining detectable at 30 to 160 days. Best response was stable disease lasting 3 months for the MC and SS patients and progressive disease for the TNBC patient.

Conclusions

This is an initial dose of a first-in-human investigator-initiated trial, using a specificity-enhanced third generation TCR targeting NY-ESO-1. A durable persistence was shown for the modified lymphocytes associated with short-term stabilization of metastatic disease and with acceptable safety.

Clinical trial identification

HBI-0201-ESO TCRT- NCT05296564.

Legal entity responsible for the study

Michal Lotem, MD, Hadassah Medical Center.

Funding

Dr Miriam and Sheldon G Adelson Medical Research Foundation.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

12P - CRISPR/Cas9-induced knock-out of DGKαζ in TAG-72 CAR-T cells improves function and persistence in ovarian cancer

Presentation Number
12P
Lecture Time
17:35 - 17:35
Speakers
  • Vera Evtimov (Notting Hill, Australia)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Vera Evtimov (Notting Hill, Australia)
  • Maree Hammett (Notting Hill, Australia)
  • Nguyen Nhu-Y (Notting Hill, Australia)
  • Junli Zhuang (Notting Hill, Australia)
  • Ian Nisbet (Notting Hill, Australia)
  • Alan Trounson (Notting Hill, Australia)
  • Richard Boyd (Notting Hill, Australia)
  • Runzhe Shu (Notting Hill, Australia)

Abstract

Background

The efficacy of chimeric antigen receptor (CAR-) T cells against solid tumours to date has been limited. This could be attributed, in part, to the endogenous tumour microenvironment (TME) - a complex milieu of immunosuppressive factors and ligands. To mitigate the impact of the immunosuppressive TME on CAR-T function, we have generated second-generation, tumour associated glycoprotein (TAG)-72 targeting CAR-T cells devoid of diacylglycerol (DAG) kinase α and ζ (DGKαζ). This deletion increases the intracellular level of DAG allowing the cells to be more metabolically active.

Methods

TAG-72 CAR lentivirus transduction was performed on healthy donor T cells followed by CRISPR/Cas9-mediated deletion of DGKαζ to give rise to CTH-004 CAR-T cells; T cells (no CAR) and TAG-72 CAR-T cells were generated in parallel. The phenotype for all groups was characterised by flow cytometry. The cytotoxic effect of CTH-004 CAR-T cells was evaluated in vitro using the real time impedance-based assay, xCELLigence®. In vivo CAR-T cell function was assessed in a NSG mouse model, xenografted with human ovarian cancer cell lines where flank tumours reached >100mm3 before intravenous administration of CAR-T cells. Persistence of CAR-T cells at the tumour site was assessed by immunohistochemistry.

Results

Deletion of DGKαζ did not impact the viability or proliferative potential of CTH-004 CAR-T cells. Further, there were no significant differences in CTH-004 phenotype observed relative to T cell (no CAR) or TAG-72 CAR-T cell controls. CTH-004 CAR-T cells were able to selectively kill TAG-72hi (OV-90) and TAG-72mid (OVCAR-3) but not TAG-72low (MES-OV) expressing ovarian cancer cell lines in vitro. In the OVCAR-3 xenograft model we observed a significant reduction in the size of pre-established tumours with TAG-72 CAR-T cells for 30-40 days after treatment. In contrast, administration of CTH-004 CAR-T cells completely ablated the tumours, with CAR-T cells still observable at the remnants of the tumour site 100 days following treatment.

Conclusions

The combination of TAG-72 CAR-T cells with deletion of DGKαζ provides improved tumour control, probably via either increased T cell killing and / or enhanced longevity.

Legal entity responsible for the study

The authors.

Funding

Cartherics Pty Ltd.

Disclosure

V. Evtimov, M. Hammett, N. Nhu-Y, J. Zhuang, I. Nisbet, A. Trounson, R. Boyd, R. Shu: Financial Interests, Personal, Full or part-time Employment: Cartherics Pty Ltd.

Collapse
Cocktail & Poster Display session Abstract session

13P - Universal prospects of cryopreserved umbilical cord blood CD34+ progenitor cell-derived NK cells: Clinical and preclinical evaluation of non-engineered and genetically engineered candidates

Presentation Number
13P
Lecture Time
17:35 - 17:35
Speakers
  • Anna-Maria Georgoudaki (Oss, Netherlands)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Anna-Maria Georgoudaki (Oss, Netherlands)
  • Nina Lamers-Kok (Oss, Netherlands)
  • Amanda Van Vliet (Oss, Netherlands)
  • Didem Özkazanc (Oss, Netherlands)
  • Denise Vodegel (Oss, Netherlands)
  • Danielle Steenmans (Oss, Netherlands)
  • Monica Raimo (Oss, Netherlands)
  • Adil Doganay Duru (Oss, Netherlands)
  • Jan Spanholtz (Oss, Netherlands)

Abstract

Background

Natural killer cells (NK) are gaining traction as (non-)engineered cell therapy products also used in combination therapies. Glycostem’s ex vivo expansion and differentiation method in a fully closed automated manufacturing platform (uNiKTM), generates GTA002 (oNKord®), an “off-the-shelf” allogeneic cryopreserved NK cell product from umbilical cord blood-derived CD34+ progenitors. Safety and tolerability of a fresh predecessor was shown in a phase I trial in AML and a phase II trial of oNKord® in AML is now on-going. Recent preclinical assessments elucidate the capacity of GTA002 to target hematological and solid malignancies.

Methods

Potency assays in combination with receptor blocking monoclonal antibodies (mAb) for assessment of cytotoxicity and receptor involvement were performed in 2D by flow cytometry and in 3D spheroids by Incucyte. GTA002’s capacity to upregulate CD16a in vivo spurred the exploration of antibody-dependent cellular cytotoxicity ADCC against otherwise non-susceptible targets.

Results

GTA002 showed rapid cytotoxic responses against melanoma cell lines at low effector:target ratios. The involvement of several activating receptors, and specifically death receptor TRAIL, for efficient cytotoxicity was revealed. Multimodal activation for efficient cytotoxicity was verified in both 2D and 3D melanoma models. Pre-clinical results show rapid ADCC against HER2+ and CD19+ tumors within a few hours from target exposure to GTA002. Furthermore, approaches to genetically equip GTA002 with chimeric antigen receptors to generate viveNKTM cells, recently showed efficient preclinical antigen-specific targeting of HER2+ and CD19+ tumors, while preserving innate NK cell cytotoxicity.

Conclusions

Overall, the preclinical innate performance and ADCC of cryopreserved “off-the-shelf” oNKord® cells as well as the cytotoxicity of viveNKTM cells demonstrate the great potential of multimodal targeting against a variety of cancer indications, and open up opportunities for combination therapies with a vast array of established mAb therapeutics and bispecific NK cell engagers.

Legal entity responsible for the study

The authors.

Funding

Glycostem Therapeutics.

Disclosure

A. Georgoudaki, N. Lamers-Kok, A. Van Vliet, D. Özkazanc, D. Vodegel, D. Steenmans, M. Raimo, A.D. Duru, J. Spanholtz: Financial Interests, Personal, Full or part-time Employment: Glycostem Therapeutics.

Collapse
Cocktail & Poster Display session Abstract session

14P - Lentivirally overexpressed c-Myc promoter binding protein (MBP-1) localizes in the cytoplasm of human cutaneous melanoma cell lines increasing cell proliferation and glycolysis rate

Presentation Number
14P
Lecture Time
17:35 - 17:35
Speakers
  • Miriam Hippner-Kunicka (Wroclaw, Poland)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Miriam Hippner-Kunicka (Wroclaw, Poland)
  • Agnieszka Łaszkiewicz (Wroclaw, Poland)
  • Joanna Skrzymowska (Wroclaw, Poland)
  • Piotr Donizy (Wroclaw, Poland)
  • Arkadiusz Miazek (Wroclaw, Poland)

Abstract

Background

C-Myc promoter binding protein (MBP-1) is a product of alternatively translated mRNA encoding alpha-enolase (ENO-1). In contrast to ENO-1, MBP-1 possesses no enzymatic activity but instead is able to bind P2 region of the c-Myc promoter leading to the modulation of its expression. Ectopic overexpression of MBP-1 was shown to reduce cell proliferation and tumorigenicity of numerous tumor cell lines, hence constituting an attractive target for cancer therapy.

Methods

We created lentiviral particles encoding HA-tagged, human MBP-1 protein, its C-terminal deletion mutant (MBP-1ΔC), or control, empty pRLL puro vector. With these tools, we created six stable transfectants derived from A375 and WM9 human melanoma cell lines. Detection of HA-tag by Western blot and immunofluorescence confirmed the overexpression of transfected proteins. We then used qPCR to estimate the effects of MBP-1 overexpression on the c-Myc transcription, Click-it Edu proliferation assay to assess the rate of cell proliferation, lactate detection assay in hypoxia and normoxia to measure the glycolytic rate and in vitro wound-healing assay to evaluate the migration ability of transduced cells.

Results

In our study, we found that overexpressed MBP-1 and MBP-1ΔC predominantly localized in the cytoplasm and only minimally decreased c-Myc mRNA expression, secondly, the proliferation rate of MBP-1- transduced cells increased in comparison to empty vector controls, and thirdly, the rate of glucose metabolism in normoxia and hypoxia increased in MBP-1 and MBP-1ΔC transduced cells. When assessing cell migration, we also found that overexpression of MBP-1 but not MBP-1ΔC led to a substantial decrease in the cell migration capacity of WM9 but not A375.

Conclusions

Our data underline potential pitfalls to avoid when overexpressing MBP-1 by means of lentiviral vectors. We provide evidence suggesting that lentiviral transduction of melanoma cell lines per se strongly affects cell proliferation and glucose metabolism. On the other hand, our research depicted an unexpected tumor-promoting activity of MBP-1 that can be largely dissociated from its nuclear localization and enzymatic activity.

Legal entity responsible for the study

Wroclaw Medical University, Wroclaw University of Environmental and Life Sciences.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

15P - Enhancement Platform for immune Cells (EPiC): invIOs’s innovative cell-therapy platform for creating personalized cancer treatments

Presentation Number
15P
Lecture Time
17:35 - 17:35
Speakers
  • Mario Kuttke (Wien, Austria)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Mario Kuttke (Wien, Austria)
  • Romana Gugenberger (Wien, Austria)
  • Kathrin Thell (Wien, Austria)
  • Sarah Bischof (Wien, Austria)
  • Bernhard Peball (Wien, Austria)
  • Hannes Muehleisen (Wien, Austria)
  • Maria Urban (Wien, Austria)
  • Bianca Gapp (Wien, Austria)
  • Agnes Bugajska-Schretter (Vienna, Austria)
  • Angela Halfmann (Wien, Austria)
  • Peter Morley (Wien, Austria)
  • Alexander Dohnal (Wien, Austria)

Abstract

Background

Immunotherapies have revolutionized the way cancer patients are treated today. While immune checkpoint-blocking antibodies are an established first-line treatment option for patients with various solid tumors, personalized immune cell therapies are still at an early stage of development. Cell-processing platforms have proven indispensable for producing personalized therapies and allowing rapid adjustments to the resulting drug products in response to tumor evolution or escape from immune surveillance.

Methods

invIOs has developed EPiC, a proprietary cell-processing platform for creating personalized immune cell therapies to treat solid and hematological cancers. Our technology allows rapid modification of immune cells via the electroporation of small interfering RNAs in a closed manufacturing process. As such the platform currently enables intracellular silencing of immune checkpoints, including some considered undruggable in different types of immune cells, such as the E3 ubiquitin protein ligase Casitas B-lineage lymphoma-b (Cbl-b).

Results

invIOs currently has two cell-therapy candidates silenced for Cbl-b in clinical and pre-clinical development leveraging the EPiC platform. APN401 is an autologous cell therapy comprising peripheral blood mononuclear cells (PBMCs), while INV441 utilizes tumor infiltrating lymphocytes (TILs). Both cell-therapy candidates showed increased and durable immune responses after transient Cbl-b silencing.

Conclusions

EPiC is a versatile cell-processing platform for developing personalized immune cell therapies for hard-to-treat solid cancers. It enables significantly shorter manufacturing times than currently available approaches. The platform has already generated two candidates using different source materials silenced for Cbl-b using siRNA. The advantage of using an RNAi-based technology is that it allows transient and highly specific modification of immune cells. Beyond silencing, EPiC enables over-expression of proteins and combinations of modalities (e.g. mRNA) to modify multiple immune pathways and create the next generation of personalized immune cell therapies.

Legal entity responsible for the study

invIOs GmbH.

Funding

Has not received any funding.

Disclosure

M. Kuttke, R. Gugenberger, K. Thell, S. Bischof, B. Peball, H. Muehleisen, M. Urban, B. Gapp, A. Bugajska-Schretter, A. Halfmann, P. Morley, A. Dohnal: Financial Interests, Personal, Full or part-time Employment: invIOs GmbH.

Collapse
Cocktail & Poster Display session Abstract session

16P - Melatonin modulates energy metabolism and kinases signaling in ovarian cancer cells

Presentation Number
16P
Lecture Time
17:35 - 17:35
Speakers
  • Luiz Gustavo Chuffa (Botucatu, Brazil)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Luiz Gustavo Chuffa (Botucatu, Brazil)
  • Henrique Spaulonci Silveira (Botucatu, Brazil)
  • Roberta Carvalho Cesário (Botucatu, Brazil)
  • Renan Aparecido Vígaro (Botucatu, Brazil)
  • Fernando Guimarães (Campinas, Brazil)
  • Fábio Rodrigues Seiva (Bandeirantes, Brazil)

Abstract

Background

Ovarian cancer (OC) is the second gynecological malignancy among women and presents high rates of recurrence associated with chemoresistance. Tumor cells adjust their energy metabolism in favor of their rapid progression. Melatonin (Mel), a hormone produced and secreted by the pineal gland during darkness, has obvious anti-tumor activities. Therefore, we investigated the mechanistic role of Mel on energy metabolism in human OC cells (SKOV-3 and CAISMOV24 cells), with special focus on glycolytic metabolism in addition to cell signaling molecules.

Methods

Cell lines were treated with Mel at concentrations of 3.4 μM for SKOV-3 and 7 μM for CAISMOV24 based on the cell cytotoxicity (CC50) for 24 h; a luzindole-treated group was used to test the influence of melatonin receptors, MT1 and MT2. Protein levels of glycolytic enzymes were analyzed by western blot and kinase levels were measured by multiplex assay.

Results

Melatonin levels dropped by half in the OC cells. We observed a significant decline in the levels of HIF-1α, G6PDH, GAPDH, LDH, and PDH in OC cells treated with melatonin regardless of the presence of luzindole, thus proving its receptor-independent actions; conversely, the luzindole-exposed groups had an upregulation of these proteins. In regard to kinases signaling, SKOV-3 cells treated with melatonin had a reduction in the concentration of CREB, JNK, NF-kB, p-38, ERK1/2, Akt, p70s6k, STAT3, and STAT5, all of them associated with tumor growth, metastasis, and recruitment of oncostatic components. In the CAISMOV24 cells, these molecules were reduced after the combination of melatonin with luzindole.

Conclusions

These findings showed that melatonin potentially regulates energy-related processes in OC cells, thereby favoring the reversal of the Warburg effect, in addition to attenuating cell signaling involved with tumor progression.

Legal entity responsible for the study

Institute of Biosciences of Botucatu.

Funding

CAPES, FAPESP (grant number: 2021/12971-7), CNPq (304108/2020-0).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

DNA damage repair

Lecture Time
17:35 - 17:35
Speakers
  • Ruth Plummer (Newcastle upon Tyne, United Kingdom)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Ruth Plummer (Newcastle upon Tyne, United Kingdom)
Cocktail & Poster Display session Abstract session

17P - New therapeutic target in triple-negative breast cancer for enhancing PARP inhibitor efficacy and stimulating the anti-tumour immune response

Presentation Number
17P
Lecture Time
17:35 - 17:35
Speakers
  • Marina Rodriguez-Candela Mateos (A Coruña, Spain)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Marina Rodriguez-Candela Mateos (A Coruña, Spain)
  • Paz Santiago-Freijanes (A Coruña, Spain)
  • Jasmin Röder (Frankfurt am Main, Germany)
  • Pranav Oberoi (Frankfurt am Main, Germany)
  • Noelia Vigo (Vitoria-Gasteiz, Spain)
  • Eloy Almenar (Vitoria-Gasteiz, Spain)
  • Teresa M. Calleja Chucla (A Coruña, Spain)
  • Joaquin Mosquera (A Coruña, Spain)
  • Benigno Acea-Nebril (A Coruña, Spain)
  • Winfried Wels (Frankfurt am Main, Germany)
  • Maria D. Mayán (A Coruña, Spain)

Abstract

Background

Triple negative (TNBC) is the most aggressive subtype of breast cancer, lacking effective targeted therapies. PARP inhibitors (PARPi) like olaparib, in combination with immune checkpoint inhibitors, stand out among the groundbreaking strategies to treat BRCA1/2 mutated TNBC but are often connected to resistance.

Methods

Western blot, coimmunoprecipitation, qPCR, immunofluorescence, proliferation and cytotoxicity assays.

Results

Our preclinical results unprecedentedly show that connexin43 (Cx43) upregulation in Cx43-null BRCA1 mutated TNBC cells, de novo resistant to PARPi olaparib, deeply resensitizes them, reducing their IC50 almost by half as well as their 2D and physiologically-appropriate 3D spheroid proliferation upon treatment with the drug. Consistent with this, olaparib therapy reduces global protein PARylation and induces significantly higher levels of DNA double-strand damage, PARP1 cleavage and caspase 3-mediated apoptosis in Cx43-restored cells, emphasizing their higher susceptibility to PARPi. Similar results were obtained under physiologically-relevant Anoikis conditions and in BRCA1 mutated ovarian cancer cells. Wild type but not Cx43-restituted cells accumulate RAD51 foci after drug treatment, denoting a potential underlying mechanism of resistance. In order to validate our proposal, an innovative extracellular vesicle-based approach was developed as an avant-garde translational strategy to efficiently deliver both Cx43 and PARPi drug to tumour cells. Combination of olaparib with Cx43-enriched vesicles distinctively enhanced olaparib efficacy against de novo resistant BRCA1 mutated TNBC cells versus the drug alone. In addition, in cocultures of patient-derived and EGFR-CAR(chimeric-antigen-receptor)-engineered natural killer (NK) cells with BRCA1 mutated TNBC cells, Cx43 restoration in tumour cells elicited a significantly higher cytotoxic NK antitumour response than wild type cells.

Conclusions

These results, protected by a EU patent, reveal Cx43 as a dual promising novel therapeutic target to increase the efficacy and to resensitize de novo resistant BRCA1 mutated TNBC to PARPi olaparib, as well as to boost NK cytotoxicity against TNBC.

Editorial acknowledgement

N/A

Legal entity responsible for the study

The authors.

Funding

Ministerio de Universidades, European Molecular Biology Organization, Xunta de Galicia.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

18P - PARP1 trapping and hyperactivation by the decoy agonist OX425 induces DNA repair abrogation and a robust anti-tumor immune response

Presentation Number
18P
Lecture Time
17:35 - 17:35
Speakers
  • Vlada Zakharova (Paris, France)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Vlada Zakharova (Paris, France)
  • Claudia Galassi (New York, United States of America)
  • Norma Bloy (New York, United States of America)
  • Chloé Doizelet (Paris, France)
  • Vincent Hayes (Paris, France)
  • Lorenzo Galluzzi (New York, United States of America)
  • Wael Jdey (Paris, France)

Abstract

Background

Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) lead to synthetic lethality when used in cancers with homologous recombination deficiency (HRD). Despite clinical benefits of PARPi, this treatment is associated with the development of resistance of HRD tumors. Here, we describe the anticancer effects of OX425, a first-in-class oligodeoxynucleotide that operates as a PARP1 decoy, resulting in constitutive PARP1 hyperactivation and consequent exhaustion of the DNA damage response.

Methods

OX425-induced PARP trapping, hyperactivation and cell cytotoxicity were examined in vitro in HRD and homologous recombination proficient (HRP) human cancer cells, as well as in non-transformed cell lines. DNA repair efficacy was monitored by analyzing repair protein recruitment to damage sites. RNAseq analysis in HRP/HRD tumor cells treated with OX425 or PARP inhibitors was employed to uncover the molecular mechanisms underlying OX425 effects. The anticancer efficacy of OX425 was assessed in vivo in different HRD and HRP tumor models, including endogenous mammary carcinomas as driven in immunocompetent female mice by MPA plus DMBA.

Results

At odds with conventional PARP inhibitors, OX425 bound to and hyperactivated PARP1 with high affinity in a dose-dependent manner, resulting in elevated cytotoxicity to multiple cancer cells with slight benefits to HRD cancers. Moreover, long-term treatment with OX425 did not show any mutagenicity compared to PARPi. The activity of OX425 was specific to tumor cells, as no significant effect on cell viability was observed for normal cells, at odds with PARP inhibitors. In line with in vitro results, OX425 mediated considerable anticancer effects in vivo as it generated an immunologically active tumor microenvironment. Importantly, OX425 treatment significantly delayed acquired resistance to olaparib in BRCA1 mutated MDA-MB-436 cell-derived xenografts.

Conclusions

Our results provide a safety profile and preclinical rationale for using OX425 in patients bearing HRD tumors, to trigger DNA damage exhaustion and initiate inflammatory responses in the tumor microenvironment.

Legal entity responsible for the study

The authors.

Funding

Onxeo.

Disclosure

V. Zakharova, C. Doizelet, V. Hayes, W. Jdey: Financial Interests, Institutional, Full or part-time Employment: onxeo. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

20P - Mutational signature-based identification of DNA repair deficient gastroesophageal adenocarcinomas for therapeutic targeting

Presentation Number
20P
Lecture Time
17:35 - 17:35
Speakers
  • Pranshu Sahgal (Boston, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Pranshu Sahgal (Boston, United States of America)

Abstract

Background

DNA repair deficiency is a common feature of cancer. Homologous recombination (HR) and nucleotide excision repair (NER) are two most frequently disabled DNA repair pathways in solid tumors. HR deficient breast, ovarian, pancreatic and prostate cancers respond well to platinum chemotherapy and PARP inhibitors. However, frequency of DNA repair pathway deficiency in gastric and esophageal adenocarcinoma (GEA) still lacks diagnostic and functional validation. Furthermore, whether DNA repair deficient GEA have enhanced responsiveness to platinum chemotherapy and sensitivity to PARP inhibitors is not well characterized.

Methods

Using whole exome and genome sequencing data, we measured various HR deficiency-associated mutational signatures in patient specimen of gastric, esophageal and colorectal cancer specimens and gastric cancer cell lines. Gold-standard immunofluorescence assays were used to confirm HR and NER deficiency in cancer cell lines. Relationship between PARP inhibitor treatment and tumor response was evaluated in patients with gastric cancer. Drug sensitivity was determined using standard in vitro cell culture assays. Single-cell RNA-sequencing was done to evaluate gastric cancer response to commonly used chemotherapeutics.

Results

We found that a significant subset of GEA, but very few colorectal tumors, show evidence of HR deficiency by mutational signature analysis (HRD score). Gastric cancer cell lines with high HRD mutational signature scores demonstrated functional HR deficiency by RAD51 assay and increased sensitivity to platinum and PARP inhibitors. A gastric cancer cell line with strong sensitivity to cisplatin showed HR proficiency but exhibited NER deficiency by DDB2 proteo-probe assay. Single-cell RNA-sequencing revealed that, in addition to inducing general apoptosis, cisplatin treatment triggered ferroptosis in a NER-deficient gastric cancer, which may explain the outlier sensitivity.

Conclusions

A subset of upper gastrointestinal tumors have genomic features of HR and NER deficiency and therefore may be more likely to benefit from platinum chemotherapy and PARP inhibition.

Legal entity responsible for the study

The author.

Funding

Research and Technology Innovation Fund (KTIA_NAP_13-2014-0021 and 2017-1.2.1-NKP-2017-00002), Breast Cancer Research Foundation (BCRF-20-159), Kræftens Bekæmpelse (R281-A16566), Degregorio Family Foundation, AGA Augustyn Award in Digestive Diseases.

Disclosure

The author has declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

21P - Cross-resistance between platinum-based chemotherapy (PlCh) and PARP inhibitors (PARPi) in castration-resistant prostate cancer (CRPC)

Presentation Number
21P
Lecture Time
17:35 - 17:35
Speakers
  • Peter H. Slootbeek (Nijmegen, Netherlands)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Peter H. Slootbeek (Nijmegen, Netherlands)
  • Iris Kloots (Nijmegen, Netherlands)
  • Inge M. Van Oort (Nijmegen, Netherlands)
  • Leonie Kroeze (Nijmegen, Netherlands)
  • Jack Schalken (Nijmegen, Netherlands)
  • Haiko Bloemendal (Nijmegen, Netherlands)
  • Niven Mehra (Nijmegen, Netherlands)

Abstract

Background

CRPC patients (pts) with loss-of-function alterations in genes associated with homologous recombination (HR) can derive benefit from both PARPi and PlCh. Cross-resistance between these agents is well recognized in other tumour types and evidence of ‘reversion’ mutations in HR genes is emerging. Yet, optimal treatment sequence and data on cross-resistance in CRPC is lacking.

Methods

In this retrospective pre-planned single-centre study we describe intra-patient responses to PlCh and PARPi, assessed by the order of these HR-deficiency-targeting-agents (HRDtA; PARPi→PlCh or PlCh→PARPi). All pts were treated with both PlCh and PARPi, but agents were not necessarily given directly sequential.

Results

All 28 CRPC pts were metastatic and HR deficient, mostly due to BRCA2 inactivation (79%). Sixteen pts received PARPi→PlCh and 12 PlCh→PARPi. Pts with PlCh→PARPi had a significant shorter time to CRPC (median 10 vs. 18 months, P=0.040) and were significantly more often synchronous metastatic (92% vs. 44%, P=0.016). Progression-free survival (PFS) on the initial HRDtA was longer than the PFS on the subsequent HRDtA (median 5.3 versus 3.4 months, P=0.016). The median PFS on PARPi, given as subsequent HRDtA, was 0.9 months shorter than when administered first. For PlCh the PFS was 3.6 months shorter as subsequent HRDtA than as initial. Of the PARPi→PlCh pts, 6/16 (38%) had a >50% PSA decline to PlCh and 2/8 (25%) evaluable pts had a radiographic response to PlCh. In the PlCh→PARPi group, 6/10 (60%) evaluable pts had a >50% PSA decline to PARPi and 5/9 (56%) a radiographic response to PARPi. In total, 12/26 (46%) had a >50% PSA decline and 7/17 (41%) a radiographic response to a subsequent HRDtA. Overall survival from CRPC did not significantly differ depending on the order of HRDtA (PARPi→PlCh 45 months vs. PlCh→PARPi 33 months, hazard ratio 1.42, P=0.401).

Conclusions

This study suggests cross-resistance between PARPi and PlCh in HR deficient CRPC pts. PlCh appears to induce less cross-resistance to PARPi than vice versa. Still, >40% of the cohort is sensitive to a subsequent HRDtA. Serial assessment of (liquid) biopsies is warranted to unravel the mechanisms defining cross-resistance.

Legal entity responsible for the study

Radboud University Medical Center, Nijmegen, The Netherlands.

Funding

Has not received any funding.

Disclosure

I.M. Van Oort: Financial Interests, Personal and Institutional, Advisory Role: Bayer, Astellas, Janssen, MSD/AstraZeneca; Financial Interests, Institutional, Research Grant: Astellas, Janssen, Bayer. J. Schalken: Financial Interests, Personal, Invited Speaker: Astellas, Bayer. N. Mehra: Financial Interests, Personal, Advisory Board: Pfizer, Roche, MSD, AstraZeneca, Astellas, JNJ; Financial Interests, Institutional, Advisory Board: Janssen; Financial Interests, Institutional, Funding: Astellas, Pfizer; Financial Interests, Personal and Institutional, Funding: Janssen; Financial Interests, Institutional, Invited Speaker: BMS, Janssen; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS; Non-Financial Interests, Personal, Leadership Role, Head of the Prostate Cancer Working Group: Dutch Uro-Oncology Study Group; Non-Financial Interests, Personal, Principal Investigator, co-PI: Prospective Bladder Cancer Infrastructure (Netherlands); Non-Financial Interests, Personal, Leadership Role: Castration-resistant Prostate Cancer Registry. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

22P - Emerging role of histone acetyltransferase CBP in breast cancer cells undergoing DNA damage

Presentation Number
22P
Lecture Time
17:35 - 17:35
Speakers
  • Wafaa S. Ramadan (Sharjah, United Arab Emirates)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Wafaa S. Ramadan (Sharjah, United Arab Emirates)
  • Samrein Ahmed (Sheffield, United Kingdom)
  • Lama Lozon (Sharjah, United Arab Emirates)
  • Soraya Mouffak (Sharjah, United Arab Emirates)
  • Wael Mansour (Hamburg, Germany)
  • Raafat El-Awady (Sharjah, United Arab Emirates)

Abstract

Background

DNA damage response and repair defects resulted from genetic and epigenetic alterations are frequently observed among breast cancer patients and eventually affect their response to chemo- and radiotherapies. Among the epigenetic alterations, the histone acetyltransferase CREB-binding protein (CBP) was found to be dysregulated in breast cancer cells. Despite of many reports that demonstrated the role of CBP in DNA damage repair, the involvement of CBP in the initial events of activating DNA damage response pathways and the potential of targeting CBP in breast cancer therapy remains poorly understood. Here we set out to explore the role of CBP in DNA damage response in breast cancer and normal cells.

Methods

Cancer and normal breast cell lines were used to examine the expression, stability, and activity of CBP and its interaction with other proteins after DNA damage induction by chemotherapeutic agent or radiation. In addition, immunofluorescence, western blot, comet assay, colony formation assay and proximity ligation assay were performed after CBP inhibition or downregulation under DNA damage.

Results

Our results showed that CBP is stabilized and recruited at the sites of DNA double strand breaks. The depletion of CBP impaired the DNA repair capacity and subsequently increased the sensitivity of breast cancer cells to chemo- and radiotherapy, without influencing the behavior of normal cells. Mechanistically, CBP was found to form a stable complex with ATM, a central regulator of DNA damage response, after treatment with DNA damaging agent. Furthermore, CBP depletion or inhibition impaired the autophosphorylation of ATM in breast cancer cells, suggesting it’s pivotal role in ATM activation. The impact of CBP on ATM's kinase activity was further augmented by the observed reduction in the phosphorylation of downstream proteins including Chk2, Chk1 and p53 in CBP-depleted cells. Interestingly, CBP downregulation did not interfere with the activation of ATM in non-cancerous breast cells.

Conclusions

Our data highlights the functional role of CBP in the response of breast cancer cells to DNA damage, particularly in ATM activation. Our results suggest that CBP could be a useful target to modulate the cellular response to DNA damaging agents in breast cancer.

Legal entity responsible for the study

The authors.

Funding

This work is financially supported by grant from the King Hussein Award for Cancer Research (grant number 2021-KHA-001).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

Epigenetics

Lecture Time
17:35 - 17:35
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Cocktail & Poster Display session Abstract session

23P - Synthetic lethality by targeting RHEB in ARID1A-mutated luminal breast cancer

Presentation Number
23P
Lecture Time
17:35 - 17:35
Speakers
  • Deniz Gulfem Ozturk (Istanbul, Turkey)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Deniz Gulfem Ozturk (Istanbul, Turkey)
  • Hande Özkan (Istanbul, Turkey)
  • Ugur Akcan (New York, United States of America)
  • Gözde Korkmaz (Istanbul, Turkey)

Abstract

Background

The heterogeneous nature of luminal breast cancer alters the patients’ response to the treatment. Despite the proven success of endocrine therapy, not all patients respond, or substantially, those who initially responded might develop endocrine resistance. It is necessary to determine novel anti-cancer agents to be used as a combination with the standard endocrine therapy regimens. Mutations in ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, are present at a high frequency in advanced ER+ breast cancer. Developing synthetic lethality (SL)-based therapeutic strategies for ARID1A-mutated breast cancer holds promise.

Methods

Here we first confirmed the effect of ARID1A knockout on endocrine therapeutic response in ER+ breast cancer in vitro. The effect on cell proliferation, viability, apoptosis, and EMT was evaluated. The synthetic lethal partner of ARID1A in breast cancer cell lines was identified using SLIdR (Synthetic Lethal Identification in R), a rank-based statistical method for predicting SL pairs from large-scale perturbation screens, The effect of genetic perturbations of RHEB on cell proliferation, apoptosis, and endocrine therapy response was evaluated in ARID1A-deficient BRCA cells compared to control cells. These results were further confirmed in ARID1A-mutated T47D cells.

Results

In vitro proliferation and viability assays confirmed increased proliferation of ARID1A KO MCF7 cells on endocrine treatment. ARID1A KO cells also promoted EMT. RHEB (Ras Homolog, MTORC1 Binding) was identified as the synthetic lethality pair of ARID1A in breast cancer cell lines. Genetic perturbations of RHEB showed that targeting RHEB selectively inhibit the growth of ARID1A-deficient BRCA cells. Moreover, knockout of RHEB in T47D cells, a breast cancer cell line carrying an ARID1A somatic mutation, significantly impaired cell proliferation, triggers apoptosis and led to susceptibility to endocrine therapy compared to control cells.

Conclusions

This study shows a novel synthetic lethality interaction between ARID1A-RHEB and hypothesizes that the development of small molecule inhibitors of RHEB that selectively inhibits the activation of mTORC1 might represent a novel strategy for treating BRCA with ARID1A loss-of-function mutations.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

24P - Fibroblast growth factor receptor (FGFR) as a target for epigenetic therapies: A case review for a new approach to target tumors with somatic mutations/amplifications of FGFR

Presentation Number
24P
Lecture Time
17:35 - 17:35
Speakers
  • M. A. Nezami (Fresno, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • M. A. Nezami (Fresno, United States of America)

Abstract

Background

Fibroblast growth factor receptors (FGFRs) are aberrantly activated in less than 10 percent of solid tumors, through different mechanisms such as single nucleotide variants, gene fusions and copy number alterations. In some types of cancer, such as urothelial or cholangiocarcinomas this frequency increases to 10–30%. Also, we see increased copy number alterations in breast cancer specifically after using cytotoxic therapies. There have been significant efforts to develop anti-FGFR drugs. Despite initial sensitivity to FGFR inhibition, acquired drug resistance leading to cancer progression develops in most patients.

Methods

Commercial ctDNA assay was provided through Guardant Laboratories. Patients had been informed, consented and treated with multi-targeted epigenetic therapies as on or off label in a phase II clinical trial. The protocol consisted of NP-Q (nanoformulated quercetin) as epigenetic modifier. The detection of ctDNA was correlated with patient’s outcome. All patients were treated with multi-targeted epigenetic therapies on a daily basis per protocol until retested. The retest was performed at least 14 days after the initial testing. Patients did not change their diet or receive any additional therapies during this time.

Results

There was a total of 28 cases identified (7 male and 21 female, ages from 33 to 76), from which 18 cases were tracked and 6 did not desire to be treated and 4 although received therapies, could not be retested. The total responders to the therapy were 14 and non-responders were 4. The response range was between 0.1 mutated allele frequencies to 3.7. Average response was at 2.6. Duration of response was tracked up to 12 months. The range of non-responders increased MAF was not more than 0.2 (3 patients with 0.1 and one patient with 0.2 percent). The response was statistically significant with average of (++) reduction of amplified gene expression manifested by direct inhibition of the FGFR.

Conclusions

To our knowledge this is the first report on longitudinal monitoring of FGFR alteration with liquid biopsy in response to epigenetic therapies. We believe such an approach can be utilized at larger scale to improve patient's response to therapies.

Clinical trial identification

N/A

Legal entity responsible for the study

The author.

Funding

Has not received any funding.

Disclosure

The author has declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

25P - Relationship between aberrant methylation of CpG islands of microRNA gene promoters and changes in their expression in epithelial ovarian cancer

Presentation Number
25P
Lecture Time
17:35 - 17:35
Speakers
  • Irina Pronina (Moscow, Russian Federation)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Irina Pronina (Moscow, Russian Federation)
  • Vitaly Loginov (Moscow, Russian Federation)
  • Alexey Burdennyy (Moscow, Russian Federation)
  • Svetlana Lukina (Moscow, Russian Federation)
  • Elena Filippova (Moscow, Russian Federation)
  • Tatiana Kazubskaya (Moscow, Russian Federation)
  • Eleonora Braga (Moscow, Russian Federation)

Abstract

Background

Epithelial ovarian cancer (EOC) is a disease found mainly in advanced stages and difficult to treat. The study of the expression and methylation of miRNA genes can become the basis for the development of potential new diagnostic and prognostic markers of EOC, as well as the basis for the development of targeted anticancer therapy.

Methods

80 paired samples of EOC (ovarian serous adenocracinoma) were obtained from the National Medical Research Center of Oncology named after N.N. Blokhin. MicroRNA expression was assessed by TaqMan-qPCR with kits from Applied Biosystems, USA. Analysis of methylation of promoter CpG islands of microRNA genes was carried out using bisulfite conversion followed by methyl-specific PCR.

Results

The methylation frequency in the tumor was higher than in paired norm for 10 microRNA genes (MIR-124-1, -124-2, -124-3, -125B-1, -127, -129-2, -132, -137, -193A and -339; p ≤ 5×10-4). ROC analysis showed that a combination of 4 microRNA genes (MIR-124-2, -127, -129-2, -137) can be proposed as a panel of EOC detection markers with sensitivity (Sn) 80%, specificity (Sp) 82 %, AUC=0.86. The combination of miRNA genes (MIR-193A, -129-2, -137) was characterized by Sn=90%, Sp=75% and AUC=0.90. For 5 out of 12 microRNAs studied, a high frequency of reduced expression was observed: miR-125b-5p - in 59% of tumor samples compairing to paired norms, miR-129-5p - in 55%, miR-132-3p - in 55%, miR-137 - in 52% , and miR-193a-5p in 66% (p ≤ 0.05). The correlation of changes in the expression of 12 microRNAs (miR-124-3p, -125b-5p, -127-5p, 129-5p, -132-3p, -137, -148a-3p, -191-5p, -193a-5p, -203a, -339-3p and -375) and changes in methylation of the genes encoding them rs=0.67-0.97 (according to Spearman), p ≤ 10-4.

Conclusions

The role of methylation in the regulation of the expression of 12 microRNAs and the association of the methylation status of 10 microRNA genes with EOC metastasis were revealed. 5 microRNAs downregulated by hypermethylation may find use as potential targets for EOC therapy.

Legal entity responsible for the study

The authors.

Funding

The state task FGFU-2022-0007 of the Ministry of Science and Higher Education of the Russian Federation.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

26P - 3MST: A potential workhorse in H2S signaling trimmed by microRNA-548 in breast cancer

Presentation Number
26P
Lecture Time
17:35 - 17:35
Speakers
  • Alyaa Dawoud (New Cairo, Egypt)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Alyaa Dawoud (New Cairo, Egypt)
  • Rana A. Youness (New Cairo, Egypt)
  • Heba Nafea (New Cairo, Egypt)
  • Tamer Manie (Cairo, Egypt)
  • Reham M. Abdel-Kader (New Cairo, Egypt)
  • Mohamed Gad (New Cairo, Egypt)

Abstract

Background

Gasotransmitters have been established as key players in cancer pathophysiology including breast cancer (BC). Hydrogen sulfide (H2S), being the newest member of gasotransmitters family, was found to be highly elevated in BC tissues. Cystathionine-β-synthase (CBS), cystathionine-γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (3MST) are the endogenous producers of H2S in viable cells. High expression of CBS and CSE in BC tissues was noticed. However, 3MST received less attention. Thus, the aim of this work is to unravel the expression pattern and regulation of 3MST in BC patients and cell lines.

Methods

BC female patients (n=20) were recruited. Patients’ clinical features showed that 65% of patients had lymph node metastasis, 15% were in stage 3-4 of BC, 25% had tumor size ≥5 cm, and 45% were in premenopausal status. Total RNA was extracted using Biazol, reverse transcribed and quantified using qRT-PCR. Western blot analysis was used to quantify the protein. In-silico analysis was used to identify microRNAs (miRNAs) targeting 3MST. MDA-MB-231 cells were cultured and transfected using oligonucleotides. H2S levels were measured by fluorescence probe (AzMc). Functional analysis experiments such as trans-well migration, MTT and colony forming assays were performed in MDA-MB-231 cells following ectopic miR-548 expression.

Results

3MST was found to be significantly up-regulated in BC tissues compared to the non-cancerous tissues. Significant high expression for 3MST was correlated to tumor size ≥5 cm, albeit not to lymph node metastasis nor to menopause. MiR-548 was listed among the top potential regulators of 3MST using 6 different bioinformatics softwares. Ectopic expression of miR-548 in MDA-MB-231 cells resulted in a marked repression of 3MST on both mRNA and protein levels. H2S levels were significantly repressed by 3MST siRNAs and miR-548 mimics. On the functional level, miR-548 markedly reduced cellular viability, migration and colony forming ability of MDA-MB-231 cells.

Conclusions

This study sheds light onto the significant involvement of 3MST in H2S signaling and its positive correlation with BC aggressiveness. Moreover, it highlights its potential target-ability using miR-548 as a novel tumor suppressor therapeutic approach in BC.

Legal entity responsible for the study

The authors.

Funding

Swiss National Science Foundation (SNSF), grant SNF IZSTZ0_198887.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

Immunotherapy

Lecture Time
17:35 - 17:35
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Cocktail & Poster Display session Abstract session

29P - A phase I, open-label, dose escalation and dose expansion study to evaluate the safety, tolerability, pharmacokinetics/pharmacodynamics, antitumor activity of QL1604, a humanized anti-PD-1 mAb, in patients with advanced solid tumors

Presentation Number
29P
Lecture Time
17:35 - 17:35
Speakers
  • Yun Fan (Hangzhou, China)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Yun Fan (Hangzhou, China)
  • Yanjun Xu (Hangzhou, China)
  • Zhiyu Huang (Hangzhou, China)
  • Wei Hong (Hangzhou, China)
  • Lei Gong (Hangzhou, China)
  • Kaiyan Chen (Hangzhou, China)
  • Jing Qin (Hangzhou, China)
  • Fajun Xie (Hangzhou, China)
  • Feng Wang (Zhengzhou, China)
  • Xin Tian (Zhengzhou, China)
  • Xiangrui Meng (Zhengzhou, China)
  • Wenlei Feng (Jinan, China)
  • Lingyan Li (Jinan, China)
  • Baihui Zhang (Jinan, China)
  • Xiaoyan Kang (Jinan, China)

Abstract

Background

PD-1 inhibitor has demonstrated promising efficacy in various solid tumors. Here we report the safety, efficacy, ADA, RO and PK/PD results from a phase I study of QL1604, a humanized anti-PD-1 mAb in pts with advanced solid tumors.

Methods

Pts with histologically or cytologically confirmed advanced solid tumors, had ≥1 target lesion (RECIST 1.1), and failed standard therapy were enrolled. The study included 2 parts: dose escalation and dose expansion. In the dose-escalation part, a titration design combined with 3+3 method was used. Pts received QL1604 at 0.3mg/kg, 1mg/kg, 3mg/kg, and 10mg/kg dose levels (DLs) Q2W in each 4-week treatment cycle until disease progression or other discontinuation events. The observation period for DLT was 4 weeks after the first dose at each DL. In the dose-expansion part, pts received QL1604 at 3 mg/kg Q2W, 10 mg/kg Q2W, 3 mg/kg Q3W, and 200 mg Q3W DLs. The primary endpoints included MTD, safety, the recommended dose for future clinical studies.

Results

As of 16 Dec 2022, 35 pts were enrolled in 2 sites in China. 18 (51.4%) pts had NSCLC. 32 (91.4%) pts had an ECOG PS of 1. 18 (51.4%) pts previously received ≥3 lines of therapy. 29 (82.9%) pts experienced TRAEs. The most common TRAEs (≥20%) were asthenia (37.1%) and anemia (22.9%). 6 (17.1%) pts experienced Gr ≥3 TRAEs. DLTs were observed in 1 pt (thymic cancer) at 3 mg/kg Q2W DL: Gr 3 immune-mediated myositis and myasthenia gravis. The MTD was not reached. 7 pts had PR (1 to 10 mg/kg Q2W or Q3W and 200 mg/kg Q3W; 5 pts with NSCLC and 2 pts with nasopharyngeal carcinoma). The ORR was 20% (7/35) and DCR was 34.3% (12/35). The AUC and Cmax increased in an approximately dose-proportional manner in the dose range 0.3 mg/kg to 10 mg/kg. 3 pts tested ADA positive at baseline and 16 pts had at least one positive result after dosing. RO was >80% on day 15 and day 22 at 3 mg/kg Q2W, 3 mg/kg Q3W, and 200 mg Q3W DLs.

Conclusions

QL 1604 showed good safety profile and efficacy signal in the dose range of 0.3 to 10 mg/kg (Q2W or Q3W) and at a fixed dose of 200 mg Q3W for pts with advanced solid tumors. 3 mg/kg Q3W and 200 mg Q3W were chosen as the recommended doses for future clinical studies.

Clinical trial identification

NCT05649761.

Legal entity responsible for the study

Qilu Pharmaceutical Co., Ltd.

Funding

Qilu Pharmaceutical Co., Ltd.

Disclosure

W. Feng, L. Li, B. Zhang, B. Zhang: Other, Personal, Full or part-time Employment: Qilu Pharmaceutical Co., Ltd. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

30P - Deep learning approach for discovering new predictive histological features of immunotherapy response

Presentation Number
30P
Lecture Time
17:35 - 17:35
Speakers
  • Saima Ben Hadj (Paris, France)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Hippolyte Heuberger (Paris, France)
  • Saima Ben Hadj (Paris, France)
  • Rutger Fick (Paris, France)
  • Alireza Moshayedi (Paris, France)
  • Elton Rexhepaj (Paris, France)

Abstract

Background

Immune checkpoint inhibitors treatments have proven their efficiency against Non-Small Cell Lung Cancer (NSCLC). However a large number of patients (around 45%) don’t respond to immunotherapies and despite the existing biomarkers (PDL1, CD8) and patient selection practices, there is not currently any relevant predictive biomarker of this response. The recent advances in Deep learning image analysis - namely Multiple Instance Learning - allow to train a model to predict survival for patients receiving immunotherapy and to confront visual results to known biomarkers.

Methods

2220 H&E histology images were gathered from 456 NSCLC patients. The 36-month follow-up with vital status is retrospectively documented (68.5% alive, and 31.5% deceased). The slides are first tiled into patches and histology phenotypes are extracted using K-means on VGG-16 feature extractor. The phenotypes are then fed into siamese networks and aggregated into a single patient representation through an Attention layer predicting the vital status (alive / deceased). The Attention map is finally used to extract the most important phenotypes driving the model decision of immunotherapy response. The result of the attention maps is compared with the CD8 and PDL1 biomarker stainings.

Results

We find that a relevant number of histologically phenotypes used for the analysis is 4 and that, based on tiles coming from each of these four phenotypes the survival prediction can be performed with an AUC of 0.75. Qualitative analysis shows that the model decision for predicting response to immunotherapy from H&E stained slide not only rely on PDL1 biomarker but also how immune cells infiltrate the tumoral PDL1 area.

Conclusions

In this study we use a new deep learning approach enabling us to discover new histological phenotypes that predict the response to immunotherapy in NSCLC. Our first results are encouraging and demonstrate the AI capability for discovering new discriminative phenotypes. Our future work will be focused on inspecting more granular phenotypes.

Legal entity responsible for the study

The authors.

Funding

Tribun Health.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

32P - Clinical efficacy and safety of an immune checkpoint inhibitor in combination with regorafenib therapy as second-line regimen for patients with unresectable hepatocellular carcinoma

Presentation Number
32P
Lecture Time
17:35 - 17:35
Speakers
  • Jinpeng Li (Jinan, China)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Jinpeng Li (Jinan, China)
  • Jinlong Song (Jinan, China)

Abstract

Background

This study aimed to evaluate the safety and efficacy of a combination of programmed death-1 (PD-1) inhibitor and regorafenib as second-line treatment for advanced hepatocellular carcinoma (HCC).

Methods

We retrospectively analyzed the data of 38 patients with unresectable HCC who were treated with PD-1 inhibitor in combination with regorafenib as a second-line therapy as well as the data of 32 patients treated with regorafenib only therapy as a control. The clinical data including the baseline data from blood routine test, liver function test, renal function test, tumor staging, tumor imaging features, previous treatment strategies, follow-up imaging results, and adverse events during follow-ups were recorded. The mRECIST were used to evaluate the treatment outcome of intrahepatic lesions, and the Kaplan–Meier method was used to evaluate survival time.

Results

Up to the last follow-up, the rego-PD-1 group had higher objective response rate (39.5% vs. 15.6%, P=0.028), longer progression-free survival (median 5.9 vs. 4.6 months; P=0.044), and better overall survival (OS) (median 14.5 vs. 9.5 months; P=0.041) than the regorafenib only group. Among the 38 patients treated with PD-1 inhibitor and regorafenib combination, 1 patient (2.7%) achieved complete response, 14 patients (36.8%) achieved partial response, 14 patients (36.8%) achieved stable disease, and 9 patients (23.7%) achieved progressive disease. Among the 32 patients treated with regorafenib alone, 5 (15.6%) achieved partial response, 12 (37.5%) achieved stable disease, and 15 (46.9%) achieved progressive disease. Regorafenib alone, Child–Pugh B, and presence of more than 3 tumors > 3 were independent prognostic factors for poor OS. The difference in the incidence of grade 3/4 adverse events between the two groups was not statistically significant (36.8% vs. 28.1%; P=0.439). Grade ≥3 treatment-related adverse events (TRAEs) included hypertension and diarrhea.

Conclusions

PD-1 inhibitor combined with regorafenib is a promising regimen in treating patients with unresectable HCC owing to its safety and effectiveness as well as low incidence of serious adverse events with its use.

Editorial acknowledgement

NO

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

33P - Nomogram to predict survival of patients with unresectable melanoma receiving immune checkpoint inhibitors

Presentation Number
33P
Lecture Time
17:35 - 17:35
Speakers
  • Eftychia Chatziioannou (Tuebingen, Germany)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Eftychia Chatziioannou (Tuebingen, Germany)
  • Lina Maria Serna Higuita (Tübingen, Germany)
  • Tobias Sinnberg (Tuebingen, Germany)
  • Heike Niessner (Tuebingen, Germany)
  • Andrea Forschner (Tuebingen, Germany)
  • Ulrike Leiter-Stoppke (Tuebingen, Germany)
  • Lukas Flatz (Tuebingen, Germany)
  • Teresa M. Amaral (Tübingen, Germany)

Abstract

Background

40-60% of patients with advanced melanoma receiving immune checkpoint inhibitors (ICI) do not derive benefit. Baseline predictive and prognostic biomarkers for ICI are currently missing. Here we aim to identify predictive clinicopathologic features using a random survival forest (RSF) and develop a nomogram for PFS in this collective.

Methods

We included patients with stage IV unresectable melanoma receiving first-line ICI and documented clinicopathologic features and blood markers at the time of therapy start. The variable importance was calculated to screen for predictors of progression-free survival (PFS) using an RSF. We developed a Cox regression prediction model based on the RFS results and established a prognostic nomogram for PFS. Performance of the nomogram was evaluated using calibration plots, Brier score, and discrimination by the Harrel C-Index and AUC. Performance was also verified by internal validation. Patients were divided into four subgroups based on quartiles of the total points (TP) of the nomogram. Kaplan Meier survival analysis was performed.

Results

We included 296 patients with a median PFS of 25 months. Median age was 67 (IQR:57, 77) years. The RSF model identified nine variables associated with PFS [LDH, type of melanoma, neutrophile/lymphocyte rate (NLR), age, S100, number of metastatic organs, presence of liver or brain metastases, and BMI] that were used to construct the prediction model. The C-index was 0.66, and calibration curves showed a good correlation between the predicted and actual progression risks. Patients with total points below 63.88 (below first quartile of TP, n=73) had a median PFS of 63 months (95% CI: 48-76), while the group with points above 121.45 (above third quartile of TP, n=75) had a median PFS of 11 months (95% CI 7-19); HR=3.95 (95% CI:2.63-5.94; p<0.001).

Conclusions

We identified predictors of PFS in advanced unresectable melanoma patients. We also established a nomogram integrating clinicopathologic features and blood markers, with good accuracy for predicting PFS that can be used for risk stratification at the time of therapy start.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

T. Sinnberg, H. Niessner: Financial Interests, Institutional, Funding: Novartis, Pierre Fabre. A. Forschner: Financial Interests, Personal, Advisory Role: Roche, Novartis, MSD, Pierre Fabre; Financial Interests, Personal, Invited Speaker: Roche, Novartis, BMS, MSD, CeGaT; Financial Interests, Institutional, Research Grant: BMS Stiftung Immunonkologie; Financial Interests, Personal, Other, travel support: Roche, Novartis, BMS, Pierre Fabre. U. Leiter-Stoppke: Financial Interests, Personal, Funding: MSD; Financial Interests, Personal, Advisory Role: Sun Pharma, Sun Pharma, Sanofi, Novartis, MSD, Roche, Almirall Hermal; Financial Interests, Personal, Invited Speaker: Sun Pharma, Sanofi, MSD, Novartis, Roche, Almirall Hermal; Financial Interests, Institutional, Invited Speaker: Sanofi, MSD. L. Flatz: Financial Interests, Personal, Research Grant: Hookipa Pharma, SAKK/Immunophotonics, Deutsche Forschungsgemeinschaft, Philogen, Mundipharma; Financial Interests, Personal, Advisory Role: Philogen, Sanofi, Novartis, BMS, Data Safety Board University; Financial Interests, Personal, Stocks/Shares: Hookipa Pharma. T.M.S. Amaral: Financial Interests, Personal, Invited Speaker: CeCaVa, BMS, Novartis, Pierre Fabre; Financial Interests, Institutional, Funding: Novartis, Neracare, Sanofi, Skyline-Dx; Financial Interests, Institutional, Research Grant: Novartis, iFIT; Non-Financial Interests, Personal, Member: Portuguese Society for Medical Oncology, Portuguese Society of Medical Oncology - Young Oncologists Group, ASCO; Other, Personal, Other, Clinical expert: Infarmed. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

34P - Targeting LSD1 rescues MHC-I antigen presentation and overcomes resistance to PD-L1 checkpoint blockade in small cell lung cancer

Presentation Number
34P
Lecture Time
17:35 - 17:35
Speakers
  • Evelyn M. Nguyen (New York, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Evelyn M. Nguyen (New York, United States of America)
  • Hirokazu Taniguchi (Nagasaki, Japan)
  • Joseph Chan (New York, United States of America)
  • Triparna Sen (New York, United States of America)
  • Andrew Chow (New York, United States of America)
  • Charles M. Rudin (New York, United States of America)

Abstract

Background

Small cell lung cancer (SCLC) is a highly aggressive tumor with early primary resistance and modest clinical response to immune checkpoint blockade (ICB). Mutations or transcriptional repression of the major histocompatibility complex class I (MHC-I) represent a key mechanism driving resistance to T cell-based therapies. Lysine-specific demethylase 1 (LSD1) regulates gene expression via demethylating lysines 4 and 9 of histone H3 and has been regarded as a promising therapeutic target in SCLC. Here we investigated the immunomodulatory functions of LSD1 in regulating MHC-I antigen presentation pathway (APP) and resistance to immunotherapy in SCLC.

Methods

To perturb LSD1 function, we employed the pharmacological inhibitor ORY-1001 and RNA interference to assess changes in MHC-I expression in SCLC cell lines by flow cytometry and western blot. We then performed RNA-seq to characterize whole transcriptomic changes in SCLC cells following LSD1 inhibition. To explore effects of targeting LSD1 on T cell cytolysis, we co-cultured SCLC presenting endogenous peptides with pre-activated primary cognate CD8+ T cells. Finally, we treated syngeneic immunocompetent mice bearing genetically engineered mouse model (GEMM)-derived tumors with ORY1001 and/or anti-PD-L1 to evaluate tumor growth and characterize intratumor immune activities.

Results

We discovered a significant and strong negative correlation between expressions of KDM1A and MHC-I/APP genes in SCLC cell lines. We then demonstrated that targeting LSD1 restores MHC-I cell surface expression, transcriptionally activates APP-regulatory genes, and enhances the immunogenic profile of SCLC. Perturbation of LSD1 activates interferon signaling, specifically activating a transactivator of MHC-I genes, and functionally rescues MHC-I-restricted T cell recognition and cytolysis. Concurrent treatment of LSD1 inhibitor and anti-PD-L1 sensitizes refractory SCLC models to ICB via antitumor CD8 T cell activities.

Conclusions

Our data define LSD1 as a potent regulator of antigen presentation in the tumor and provide translational support for combinatory use of LSD1 inhibitors to enhance ICB sensitivity in SCLC and other MHC-I-deficient tumors.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

T. Sen: Financial Interests, Personal, Research Grant: Jazz Therapeutics. C.M. Rudin: Financial Interests, Personal, Advisory Board: Bridge Medicines, Earli, Harpoon Therapeutics. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

36P - Therapeutic vaccination with HPV-16 oncoproteins fused into a checkpoint modifier of early T cell activation protects against HPV-associated tumors in a preclinical model

Presentation Number
36P
Lecture Time
17:35 - 17:35
Speakers
  • Susan L. Currie (Newark, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Susan L. Currie (Newark, United States of America)
  • X Zhou (Philadelphia, United States of America)
  • Z Xiang (Philadelphia, United States of America)
  • W Giles-Davis (Philadelphia, United States of America)
  • Andrew Luber (Newark, United States of America)
  • M Mohammadi (Philadelphia, United States of America)
  • Hildegund C. Ertl (Philadelphia, United States of America)

Abstract

Background

Checkpoint inhibition(CPI) by antibodies against PD-1, CTLA-4 and other immunoinhibitors has revolutionized cancer treatment. However, there are limited data on CPIs that target the activation phase of adaptive immune responses. Signaling through the immunoinhibitory B and T lymphocyte attenuator (BTLA), upon binding to the herpes virus entry mediator (HVEM) on dendritic cells, regulates early steps of CD8+ T cell activation. HSV-1 glycoprotein D (gD) attaches to HVEM and blocks BTLA-HVEM signaling and allows for co-stimulation through LIGHT, which binds to a different domain on HVEM. BTLA blockade, in turn, enhances and broadens CD8+ T cell responses to a target antigen. Here, we report the immunogenicity and efficacy of a chimpanzee adenoviral vector (AdC) vaccine expressing a novel sequence derived from the early proteins 2, 5, 6 and 7 of HPV-16 fused into gD (AdC-gDE7652).

Methods

The frequency of HPV-16-specific CD8+ T-cells was assessed with intracellular cytokine staining in C57/Bl6 or HLA-A2 mice after a single IM vaccination with AdC vectors encoding HPV-16 E7652 oncoproteins expressed within gD or without gD. Efficacy was tested in a standard (5x10ˆ4 cells) dose TC-1 tumor cell challenge model with mice receiving a single IM injection of AdC-gDE7652 or AdC expressing HIV gag fused within gD (AdC-gDgag) 3 days after tumor cell transplantation. Mice were followed for 80 days.

Results

The addition of gD increased HPV-16 -specific CD8+ T-cell frequencies approximately 15-fold. In the standard TC-1 challenge experiment, 100% (n = 10) of the AdC-gDE7652 vaccinated animals experienced regression and complete tumor loss by day 25; this was sustained through day 80. In contrast, 100% (n=10) of the AdC-gDgag vaccinated animals experienced rapid tumor growth and death by day 21.

Conclusions

These preclinical data demonstrated that the addition of gD, an early checkpoint modifier, which acts locally at the site of T cell stimulation, to an HPV-16 vaccine markedly improves the vaccine’s immunogenicity and efficacy. A clinical study evaluating this construct, in HPV-16 induced cancers and precancerous lesions, is in development.

Legal entity responsible for the study

The authors.

Funding

Virion Therapeutics Llc.

Disclosure

S.L. Currie: Financial Interests, Personal, Stocks/Shares: Virion Therapeutics. A. Luber: Financial Interests, Personal, Ownership Interest: Virion Therapeutics. H.C. Ertl: Other, Personal and Institutional, Leadership Role, Co-founder: Virion Therapeutics; Other, Personal, Advisory Role: Biogen, Regenxbio; Other, Personal, Advisory Board: Ring Therapeutics, Canine Rabies Treatment Initiative. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

37P - Dose transition pathways for time-to-event continual reassessment method (TITE-CRM): Will imposing a waiting time result in better performance?

Presentation Number
37P
Lecture Time
17:35 - 17:35
Speakers
  • Zhulin Yin (London, United Kingdom)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Zhulin Yin (London, United Kingdom)
  • Christina Yap (Sutton, United Kingdom)
  • Shing Lee (New York, United States of America)
  • Ken Cheung (New York, United States of America)

Abstract

Background

Novel agents, such as immunotherapies, are likely to cause late-onset toxicities. The inclusion of these toxicities can lead to prolonged trials since traditional phase I designs require full follow-up information for each participant. TITE-CRM, an extension of the CRM design, permits continuous recruitment of participants by incorporating incomplete follow-up data to determine the recommended dose for the next participant. Thus, it can shorten trial duration, but it may also treat participants at suboptimal doses due to the uncertainty of the safety at the current dose. Imposing a waiting window to allow for more safety data to accumulate may assign the next participant to a higher and potentially more effective dose if no DLT is observed. Our aim is to create a practical tool to help trialists “look ahead” to see if imposing a waiting window would make a difference to the model’s dose recommendation.

Methods

We propose the dose transition pathways (DTP) for TITE-CRM design with possible waiting window (DTP-TITE-CRM). The DTP is a practical tool to visualize the dose recommendations for subsequent cohort of participants and was first proposed by Yap et al 2017. In DTP-TITE-CRM, the DTP projects all possible recommendations until complete follow-up is achieved. The user can also specify the maximum time for a participant to wait before being assigned to a dose.

Results

Simulations show that DTP-TITE-CRM has comparable accuracy as TITE-CRM in most scenarios in terms of identifying the MTD. In settings where the MTD is at the highest dose, or all doses are tolerable, DTP-TITE-CRM outperforms TITE-CRM. We illustrate the use of DTP-TITE-CRM in a published trial which used TITE-CRM, where imposing additional waiting time could have led to different dose decisions.

Conclusions

DTP-TITE-CRM looks ahead and enumerates dose recommendations in advance and gives participants and trialists the choice to wait or to treat immediately. This approach is particularly appealing when patient numbers are limited and when the tested agents are expected to have a tolerable toxicity profile, as it allows more participants to be treated at higher doses if safe.

Legal entity responsible for the study

C. Yap.

Funding

National Institute for Health Research (Biomedical Research Centre at The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, London).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

38P - Locoregional radiotherapy improves survival outcomes in de novo metastatic nasopharyngeal carcinoma treated with chemoimmunotherapy

Presentation Number
38P
Lecture Time
17:35 - 17:35
Speakers
  • Yujun Hu (Guangzhou, China)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Yujun Hu (Guangzhou, China)
  • Tianzhu Lu (Nanchang, China)
  • Hao Zhang (Wuhan, China)
  • Min Fang (Nanchang, China)
  • Bijuan Chen (Fuzhou, China)
  • Qiaojuan Guo (Fuzhou, China)
  • Shaojun Lin (Fuzhou, China)
  • Ying Wang (Guangzhou, China)
  • Ping Feng (Guangzhou, China)
  • Xiaochang Gong (Nanchang, China)
  • Jianji Pan (Fuzhou, China)
  • Jingao Li (Nanchang, China)
  • Yunfei Xia (Guangzhou, China)

Abstract

Background

We aimed to investigate the efficacy of locoregional radiotherapy (LRRT) in patients with de novo metastatic nasopharyngeal carcinoma (dmNPC) receiving chemotherapy combined with anti-programmed cell death receptor-1 monoclonal antibodies (anti-PD-1 mAbs) as first-line treatment and to identify optimal LRRT candidates based on Epstein-Barr Virus DNA (EBV DNA).

Methods

We enrolled patients with dmNPC receiving platinum-based palliative chemotherapy and anti-PD-1 mAbs followed or not followed by LRRT from four centers. The endpoints were progression-free survival (PFS), objective response rate (ORR), and overall survival (OS). Additionally, we used the inverse probability of treatment weighting (IPTW) to balance baseline characteristics of the LRRT and no-LRRT groups to minimize selection bias before the comparative analyses. Multivariate analyses were performed using the Cox proportional hazards model.

Results

We included 163 patients with dmNPC (median follow-up: 22 months). The median PFS was 20 months, and the ORR was 69.2%; the median OS was not achieved. After the IPTW adjustments, patients who received LRRT had a significant survival benefit over those who did not receive LRRT (median PFS: 28 months vs. 14 months; ORR: 81.0% vs. 48.3%; all P <0.001). The EBV DNA level after four to six cycles of anti-PD-1 mAbs (weighted hazard ratio [HR]: 2.01, 95% confidence interval [CI]: 1.11−3.65, P = 0.02) and LRRT (weighted HR: 0.58, 95% CI: 0.34−0.99, P = 0.04) were independent prognostic factors. Patients with undetectable EBV DNA levels after four to six cycles of anti-PD-1 mAbs (early EBV DNA clearance) benefitted from LRRT (HR: 0.43, 95% CI: 0.23−0.80, P = 0.007), whereas patients with detectable levels did not (HR: 1.03, 95% CI: 0.47−2.26, P = 0.93).

Conclusions

Palliative chemotherapy combined with anti-PD-1 mAbs followed by LRRT was associated with improved survival outcomes in patients with dmNPC, especially for patients with early EBV DNA clearance.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

40P - Development of radiolabelled plerixafor as a theranostic molecule for targeting CXCR4 receptor expressing cancers: A translational study

Presentation Number
40P
Lecture Time
17:35 - 17:35
Speakers
  • Tamanna Lakhanpal (Chandigarh, India)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Tamanna Lakhanpal (Chandigarh, India)
  • B.R. Mittal (Chandigarh, India)
  • Jaya Shukla (Chandigarh, India)
  • Rajender Kumar (Chandigarh, India)
  • Yogesh Rathore (Chandigarh, India)
  • Pankaj Malhotra (Chandigarh, India)
  • Gaurav Prakash (Chandigarh, India)
  • Alka Khadwal (Chandigarh, India)
  • Amanjit Bal (Chandigarh, India)

Abstract

Background

The emerging science of nuclear medicine emphasizes the use of radionuclides tagged vector molecules delivered to specific targets. The radiolabelled vector, agonist, or antagonist provides a landscape of disease spread and help in targeted radionuclide therapy. Chemokines favor leukocyte infiltration, and tumor metastasis. Chemokines receptor 4 (CXCR4) is over-expressed in more than 70% malignancies. Plerixafor is a CXCR4 antagonist has been explored for radionuclide imaging, and targeted radionuclide therapies.

Methods

Various parameters (temp, pH, time, and reaction volume) for conjugation of Plerixafor with different bifunctional chelating agents (DTPA, NOTA) and conjugation of Plerixafor for radiolabelling with 68Ga and 177Lu for imaging and therapy were optimized. Quality control tests of radiotracers such as radionuclide; radiochemical purity; sterility; pyrogenicity; serum stability were performed. The binding affinity and cytotoxicity were performed in CXCR4 expressing cancer cell lines. In-vivo physiological distribution was conducted in normal rats. After obtaining Institutional Ethical clearance, 68Ga-Plerixafor PET/CT imaging was performed in lymphoma patients and was correlated with 18F-FDG uptake for proof of concept.

Results

DTPA conjugated (1087 Da) and NOTA conjugated (1014 Da) Plerixafor determined mass spectra. Radionuclide and radiochemical purity of 68Ga and 177Lu Plerixafor was ≥ 99%. Synthesized radiotracers were stable, sterile and pyrogen-free, and suitable for intravenous administration. The radioligand binding assay confirmed high target efficacy (Kd= 57.16 nM) of 177Lu-Plerixafor towards CXCR4 expressing cancer cells. Furthermore, the log absolute IC50 concentration of 177Lu-Plerixafor in cytotoxicity studies was 2.628 nM. Nuclear receptor expression was also observed in immunocytochemistry. In-vivo physiological biodistribution of 68Ga-Plerixafor was in the liver (6.36%), spleen (11.56%), and lung (3.57%). Compared to 18F-FDG, concordant uptake in lesions was seen in 68Ga-Plerixafor PET/CT imaging.

Conclusions

High target efficacy of radiolabelled Plerixafor elicits theranostic potential for CXCR4 over-expressing cancers.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

41P - BRCA2 pathogenic variant (PV): A novel agnostic biomarker for immune checkpoint blockers (ICB)?

Presentation Number
41P
Lecture Time
17:35 - 17:35
Speakers
  • Beatriz Alonso de Castro (A Coruña, Spain)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Beatriz Alonso de Castro (A Coruña, Spain)
  • Martin igor Gomez Randulfe (A Coruña, Spain)
  • Kaissa Ouali (Villejuif, France)
  • Kristi Beshiri (Genova, Italy)
  • Javier Gavira Diaz (Barcelona, Spain)
  • Capucine Baldini (Villejuif, Cedex, France)
  • Stephane Champiat (Villejuif, Cedex, France)
  • Jean-Marie Michot (Villejuif, France)
  • Rastislav Bahleda (Villejuif, France)
  • François-Xavier Danlos (Villejuif, France)
  • Anas Gazzah (Villejuif, Cedex, France)
  • Antoine Hollebecque (Villejuif, Cedex, France)
  • Arnaud Bayle (Villejuif, Cedex, France)
  • Yohann Loriot (Villejuif, Cedex, France)
  • Andreea Varga (Villejuif, Cedex, France)
  • Aurélien Marabelle (Villejuif, France)
  • Sophie Postel-Vinay (Villejuif, France)
  • Santiago Ponce Aix (Madrid, Spain)

Abstract

Background

Immunotherapy has revolutionized cancer treatment improving survival rates for many cancer types. Unfortunately, resistances are common and predictive biomarkers are needed. Microsatellite Instability-high (MSI-H) usually predicts response to ICB in all tumour types, whereas we need more studies focusing on tumour mutational burden (TMB) to predict its role as a pan-cancer biomarker. Recent investigations in murine models and cohorts of patients (pts) have shown that BRCA2 PV improved response and overall survival to ICB compared to BRCA1 PV. The aim of this study was to evaluate the response to ICB in BRCA PV and its relation with TMB, MSI-H and prior treatments.

Methods

We conducted an unicohort retrospective study, between May 2020 and November 2022, in metastatic pts carrying BRCA1 or BRCA2 somatic PV and treated with ICB in phase I/II trials at Institute Gustave Roussy (France). Genomic analysis were performed by NGS (liquid biopsy and tumor samples). Data was extracted from electronic medical record and analysed with SPSS software.

Results

A total of 44 pts were enrolled. Median age was 54.6 years [range 29–74 years], 46.7% were female. Lung cancer was the most common tumour (20.5%) in an heterogenic histologic cohort of pts. Median previous lines of treatment was 2.7 [range 0-4]. Tissue analysis revealed 21.1% MSI-H, 36.8% TMB high, 23.8% BRCA1 PV, 38.1% BRCA2 PV. Blood analysis showed 7.3% MSI-H, 52.4% TMB high, 40,5% BRCA1 PV, 52.4% BRCA2 PV. All pts received immunotherapy, 69.8% ICB. The Objective Response Rate (ORR) was 34.1%. Pts with BRCA2 PV presented a better ORR to ICB compared to those with BRCA1 PV (28% vs. 4%, p = 0,030). Mean TMB was lower in BRCA2 PV than in BRCA1 PV pts and median previous lines of treatment was higher in BRCA2 PV than in BRCA1 PV (2.5 vs. 2.0). We didn´t find a significant association between BRCA1/2 PV and MSI status. Duration of response (DoR) to ICB by more than 6 months tended to be higher in pts with BRCA2 PV than those with BRCA1 PV (32% vs. 16%, p = 0,512).

Conclusions

Pts with somatic BRCA2 PV had better ORR to ICB regardless of TMB, MSI and prior lines of treatment than those with BRCA1 PV. Further research should be carried out to confirm BRCA2 as a predictive pan-cancer biomarker to ICB in a larger cohort with BRCA WT pts as comparison.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

K. Beshiri: Financial Interests, Personal, Invited Speaker: Gustave Roussy. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

43P - Prognostic impact of the tumor immune microenvironment in adrenocortical cancer

Presentation Number
43P
Lecture Time
17:35 - 17:35
Speakers
  • Nano Pachuashvili (Moscow, Russian Federation)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Nano Pachuashvili (Moscow, Russian Federation)
  • Liliya Urusova (Moscow, Russian Federation)

Abstract

Background

Adrenocortical carcinoma (ACC) is an uncommon endocrine malignancy, usually characterized by a late detection, aggressive clinical course, and poor outcome. The tumor microenvironment (TME) which includes infiltrating immune cells plays a critical role in tumor growth, survival, and prognosis in cancer patients. The presence of tumor-infiltrating immune cells (TIIC) affect the clinical benefit from novel strategies of immunological checkpoint blockade. Anti-immune pathways like PD-L1 are used by the tumor to overcome immune system and they serve as immunotherapy targets.

Methods

The study included tumor tissue samples from 75 patients with ACC, which treated at the Endocrinology Research Centre (Russia, Moscow) between 2010 and 2022: 47 cases of conventional (62,7%), 18 cases of oncocytic (24%), and 9 cases of myxoid (12%) and 1 case of sarcomatoid (1,3%) variants of ACC. Immunohistochemical analysis of tumor tissue sections was carried out according to the standard technique with a peroxidase detection system with DAB on an automatic Leica BOND III IHC staining system using Leica reagents and protocols. Each of the 75 patients underwent histological diagnostics and a series of immunohistochemical stains for the markers of the main immune cell subsets: CD45, CD3, CD4, CD8, and CD68. The impact of PD-L1 expression and the number of TIIC considering the intratumoral and stromal distribution on pathological characteristics and clinical outcomes were analysed.

Results

The number of СD45+ immune cells in tumor parenchyma and stroma was 189 and 268 cells/mm2, respectively. However, the number of immune cells from all the analyzed populations in tumor parenchyma was higher in oncocytic compared to conventional ACC cases. The analysis of the relationship of survival with the studied factors showed that the overall survival and progression-free survival between conventional and oncocytic histological variants differ significantly. The differences in survival between conventional and oncocytic histological variants of ACC were statistically significant (p-value < 0.05). PD-L1 expression does not affect prognosis.

Conclusions

Rich T-lymphocyte response is a good prognostic factor in ACC. The study of TIIL subpopulations can be used to predict ACC outcomes.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

44P - HPV16-specific CD4 and CD8 T-cell activation and functionality in patients receiving combination PDS0101 immunotherapy

Presentation Number
44P
Lecture Time
17:35 - 17:35
Speakers
  • Lauren V. Wood (Florham Park, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Lauren V. Wood (Florham Park, United States of America)
  • Stephen McCarthy (Dowlestown, United States of America)
  • David Schaaf (Florham Park, United States of America)
  • Nathalie Riebel (Florham Park, United States of America)
  • Sally Jones (Florham Park, United States of America)
  • Adam Cotty (Dowlestown, United States of America)
  • Julie Bick (Dowlestown, United States of America)

Abstract

Background

The novel, investigational HPV-targeted immunotherapy PDS0101 is being studied in combination with pembrolizumab in a phase 2 clinical trial (NCT04260126) in patients with HPV16-positive head and neck cancer. Measurement of antigen-specific activation of endogenous T-cells is critical to understanding drug-induced T-cell based immunity and its association with observed clinical outcomes. In this pilot study, we sought to establish optimal stimulation conditions for in vitro activation with selected HPV16 peptide pools that would enable downstream single cell analysis of functional cytokine profiles of HPV-specific CD4 and CD8 T-cell subpopulations.

Methods

The IsoPlexis CodePlex platform was used to track cytokine profiles generated over time by peptide-activated CD4 and CD8 T-cells. Cryopreserved PBMCs from 2 study subjects collected at 3 timepoints (pre-treatment, and 12 and 36 weeks following 4 and 5 cycles of combination therapy, respectively) were thawed and recovered overnight in the presence of IL-2, before activation with overlapping HPV16 E6 and E7 peptide pools for 1hr, 6hr, 16hrs or 24hrs. Recovered stimulated cells were enriched for CD4 and CD8 populations using magnetic bead separation and the cells plated overnight. The supernatants from each of these populations were recovered and frozen at -80°C until all supernatants were available for analysis. Supernatants were loaded on to CodePlex chips and analyzed using the IsoLight instrument.

Results

Strong CD4 and CD8 T-cell responses were documented after only 1hr of peptide stimulation and at all time course timepoints. Detection of multiple cytokines (background subtracted) was captured by concentration and demonstrated post-treatment increases in granzyme B, IFN-g, TNF-a/b, MCP-1, MIP-1a/1b and perforin reflecting development of HPV-specific CD8 and CD4 T cell reactivity and immune memory. Stimulation between 6-16hrs provided the most multiparametric and robust cytokine signals.

Conclusions

PDS0101 treatment induces polyfunctional CD4 and CD8 T-cell responses across multiple timepoints. Additional studies of cell-specific functional profiles in larger numbers of subjects and correlation with clinical outcomes are planned.

Clinical trial identification

NCT04260126.

Legal entity responsible for the study

PDS Biotechnology.

Funding

PDS Biotechnology.

Disclosure

L.V. Wood, D. Schaaf, N. Riebel, S. Jones: Financial Interests, Personal, Full or part-time Employment: PDS Biotechnology. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

45P - Discovery of CBO-212, a first-in-class drug Fc-conjugate (DFC), targeting CD73 in cancer

Presentation Number
45P
Lecture Time
17:35 - 17:35
Speakers
  • James Levin (San Diego, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • James Levin (San Diego, United States of America)
  • Simon Döhrmann (San Diego, United States of America)
  • Nicholas Dedeic (San Diego, United States of America)
  • Amanda Almaguer (San Diego, United States of America)
  • Doug Zuill (San Diego, United States of America)
  • Elizabeth Abelovski (San Diego, United States of America)
  • Rajvir Grewal (San Diego, United States of America)
  • Joanne Fortier (San Diego, United States of America)
  • Qiping Zhao (San Diego, United States of America)
  • Maria Hernandez (San Diego, United States of America)
  • Karin Amundson (San Diego, United States of America)
  • Madison Moniz (San Diego, United States of America)
  • Hongyuan Chen (San Diego, United States of America)
  • Dhanya Panickar (San Diego, United States of America)
  • Thanh Lam (San Diego, United States of America)
  • Tom Brady (San Diego, United States of America)
  • Allen Borchardt (San Diego, United States of America)
  • Jason N. Cole (San Diego, United States of America)
  • Les Tari (San Diego, United States of America)

Abstract

Background

CD73 contributes to immune evasion in solid tumors by producing immune-suppressive adenosine in the tumor microenvironment. Herein, we describe CBO-212, a first-in-class CD73 targeting DFC, comprising a multi-valent conjugate of a novel small molecule CD73 inhibitor to a proprietary immune-silent hIgG1 Fc. CBO-212 combines the strengths of small molecule inhibitors and monoclonal antibodies targeting CD73 currently in clinical development with potential best-in-class activity.

Methods

Inhibition of CD73 was evaluated in cell-free and cell-based assays. Functional activity was measured in a PBMC rescue assay in the presence of AMP and binding to cancer cells was measured in the presence and absence of AMP by flow cytometry. CD73 internalization was determined using MDA-MB-231 cells. Pharmacokinetic (PK) studies were conducted in Balb/c mice. Plasma concentrations were measured using an anti-hIgG in combination with CD73 capture ELISA. Efficacy of CBO-212 was evaluated in syngeneic mouse models with established CT26.WT tumors.

Results

CBO-212 is a potent, AMP-competitive CD73 inhibitor. In cell-free and cell-based CD73 inhibition assays, CBO-212 IC50s were single digit nM, making them comparable or superior to small molecule inhibitors and anti-CD73 mAbs in clinical development. Unlike most anti-CD73 mAbs, CBO-212 demonstrated complete enzyme inhibition. Furthermore, CBO-212 demonstrated an EC50 in a human PBMC rescue assay of 6 nM, which was equivalent to small molecule inhibitor controls and approximately 100-fold more potent than anti-CD73 mAb controls. Additionally, CBO-212 triggered CD73 internalization, similar to some anti-CD73 mAbs. CBO-212 demonstrates long half-life and stability in mice. In a syngeneic mouse colon cancer model, a single 20 mg/kg dose of CBO-212 resulted in 37.9% tumor growth inhibition (TGI) relative to vehicle (P=0.0152).

Conclusions

CBO-212 demonstrated high potency in CD73 enzyme inhibition and functional PBMC rescue assays. The in vitro potency and favorable PK of CBO-212 translated to significant TGI in a syngeneic model with a single dose. Based on these results CBO-212 is being advanced as a clinical development candidate for the treatment of solid cancers.

Legal entity responsible for the study

Cidara Therapeutics.

Funding

Cidara Therapeutics.

Disclosure

J. Levin, S. Döhrmann, N. Dedeic, A. Almaguer, D. Zuill, E. Abelovski, R. Grewal, J. Fortier, Q. Zhao, M. Hernandez, K. Amundson, M. Moniz, H. Chen, D. Panickar, T. Lam, T. Brady, A. Borchardt: Financial Interests, Personal, Full or part-time Employment, Shareholder: Cidara Therapeutics.

Collapse
Cocktail & Poster Display session Abstract session

Kinase-targeting agents and cell signalling modulators

Lecture Time
17:35 - 17:35
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Cocktail & Poster Display session Abstract session

48P - Efficacy analysis and mechanism exploration of furmonertinib for advanced NSCLC with EGFR exon 20 insertion mutation

Presentation Number
48P
Lecture Time
17:35 - 17:35
Speakers
  • Xiao Zhang (Zhengzhou, China)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Xiao Zhang (Zhengzhou, China)
  • Guoxing Feng (Tianjin, China)
  • Huan Han (Zhengzhou, China)
  • Bosheng Dong (Zhoukou, China)
  • Yanfang Yang (Zhumadian, China)
  • Hui Zhu (Zhengzhou, China)
  • Saijun Fan (Tianjin, China)
  • Hong Tang (Zhengzhou, China)

Abstract

Background

Here, we analyzed the clinical efficacy of furmonertinib, a novel 3rd generation EGFR TKI, in advanced NSCLC patients (pts) who harboring EGFRex20ins and explored mechanism.

Methods

A retrospective single-arm analysis was performed to evaluate the efficacy of 20 NSCLC pts harboring EGFRex20ins receiving furmonertinib treatment from three institutions. Meanwhile, we investigated the clinical efficacy of furmonertinib versus osimertinib as second-line treatment, because pts about furmonertinib as first-line treatment were immature. In addition, the binding activity of different EGFR TKIs to EGFRex20ins were computationally constructed based on the crystal structure of EGFR D770_N771insNPG/V948R (PDB ID: 7LGS) by the Schrödinger software (2021-2 Release, Schrödinger Inc., Portland, Oregon).

Results

Of the 20 pts selected, we found that EGFR S768_D770dup (n=5) variants were more common. Six first-line pts all achieved PR (ORR: 100.0%), five of the eight second-line pts achieved PR (ORR: 62.5%), and three of the six multiple-line pts achieved PR (ORR: 50.0%). We observed 14 pts with PR and six pts with SD as best response to furmonertinib (ORR: 70.0%, DCR: 100%). All pts showed tumor shrinkage in target lesions (median best percent change, -36.43% [-74.78%, -5.56%]). Median PFS was 10.2 (95 % CI, 7.19-13.21) months(mo). Median DOR was 8.5 (95 % CI, 4.97-12.03) mo. Comparative analysis of the efficacy of different groups showed that median PFS was significantly longer in furmonertinib group than in osimertinib (10.2 vs 3.8 mo, p = 0.008). Median OS was numerically longer in furmonertinib group than in osimertinib (18.9 vs 11.7 mo, p = 0.207). No grade 3 or above adverse events were observed. Furthermore, rather than erlotinib (GlideScore: -5.564; MM/GBSA: -52.8044), gefitinib (-7.68; -47.317), and afatinib (-5.075; -44.64), furmonertinib (-11.085; -68.1575) and osimertinib (-10.031; -63.87) revealed favorable binding activity to EGFRex20ins, with furmonertinib being the most significant.

Conclusions

Furmonertinib has positive clinical efficacy to advanced NSCLC pts with EGFRex20ins probably based on its favorable binding activity to EGFRex20ins. Furmonertinib may be the optimal choice for these pts in the future.

Legal entity responsible for the study

H. Tang.

Funding

Natural Science Foundation of Henan Province 212300410400.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

49P - Treatment management in RAS and BRAF mutations in patients with metastatic colorectal cancer

Presentation Number
49P
Lecture Time
17:35 - 17:35
Speakers
  • Merve Özkan (Izmir, Turkey)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Merve Özkan (Izmir, Turkey)
  • Yasar Yildiz (Ankara, Turkey)
  • Utku Oflazoglu (Izmir, Turkey)
  • Zeynep Guc (Izmir, Turkey)
  • Yuksel Kucukzeybek (Ankara, Turkey)
  • Ahmet Alacacioglu (Izmir, Turkey)

Abstract

Background

We aimed to investigate the effects of RAS and BRAF analyses and mutations on survival in patients diagnosed with metastatic colorectal cancer (mCRC), which we followed in our clinic for last 5 years.

Methods

186 patients followed up in our clinic with mCRC were reviewed retrospectively.

Results

The median age of 186 patients followed up with mCRC was 58 (44–76). 118 (63%) were men and 68 (37%) were women. At the end of the mean 40.3(8-122) months follow-up period, 74 (40%) of patients were alive, while 112 (60%) were passed away. Primary tumor localization and metastasis areas (liver, lymph nodes, lung, peritoneal carsinomatosis) of the patients were examined at the time of diagnosis. The overall survival of 47 KRAS mutant patients was 32 months, shorter than that of KRAS wild-type patients (P=0.66). The overall survival of three NRAS mutant patients was 33 months, shorter than that of NRAS wild patients (P=0.68). In accordance with the literature, the survival of BRAF mutant patients was shorter at 17.5 months (P=0.33).

Conclusions

There was no significant difference in survival between RAS/BRAF mutant patients treated with doublet plus anti-EGFR agent or doublet plus anti-VEGF agent. However, one of the BRAF mutant patients was also MSI-high (MSI-H) and treated with doublet plus anti-VEGF therapy; the survival time was significantly longer than that of the other BRAF mutant patients at 28 months (p < 0.05). According to the literature, Ras-wild patients were initially treated with doublets plus anti-EGFR agents. In our study, according to the RAS analysis, no treatment option had a statistically significant effect on survival. The overall survival of 47 KRAS mutant patients was 32 months, shorter than that of KRAS wild-type patients (P=0.66). While the patients in the RAS/BRAF wild group were observed to live longer than the other, no statistical significance was detected (36 months ± 13.7 x 30.5 months ± 10.5, p=0.728). The use of intensive chemotherapy regimens in BRAF mutant and MSI-H patients with a low incidence and poor prognosis of mCRC, the inclusion of anti-VEGF therapy in the first-line treatment, and the selection of new targeted therapies based on new genomic discoveries can increase effectiveness.

Legal entity responsible for the study

M. Özkan.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

50P - KRAS mutation, the molecular landscape of lung adenocarcinoma in the Portuguese population

Presentation Number
50P
Lecture Time
17:35 - 17:35
Speakers
  • Carolina F. Trabulo (Barreiro, Portugal)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Carolina F. Trabulo (Barreiro, Portugal)
  • João Gramaça (Barreiro, Portugal)
  • Nuno Dias Girão (Lisbon, Portugal)
  • José L. Mendes (Lisbon, Portugal)
  • Rita Gameiro Dos Santos (Barreiro, Portugal)
  • Joana C. Gonçalves (Barreiro, Portugal)
  • Isabel C. Gomes Fernandes (Barreiro, Portugal)
  • Sara Alfarroba (Lisbon, Portugal)
  • Patricia Winckler (Lisbon, Portugal)
  • Ines M. Guerreiro (Lisbon, Portugal)
  • Idília Maria Matos Pina (Barreiro, Portugal)
  • Ricardo J. Da Luz (Lisbon, Portugal)

Abstract

Background

Activating mutations in Kirsten rat sarcoma viral oncogene homologue (KRAS) are detected in over 1/3 of non–small-cell lung cancer (NSCLC) subtype, representing the most prevalent genomic driver event. Mostly located in codons 12 and 13, the most frequent is the p.Gly12Cys (c.34G.T) (G12C), representing about 20-25% of KRAS drive mutations in NSCLs, but remains scarce. The impact of them on prognosis is currently subject to debate, as is their impact on the response to chemotherapy and EGFR tyrosine kinase inhibitors. Our goal was to stratify our population, reporting the KRAS mutation frequency in a series of Portuguese NSCLC cases, in order to observe the incidence and impact of change of practice.

Methods

Retrospective study included NSCLC samples submitted for KRAS testing at one Portuguese center in Lisbon, between 1/1/2027 and 31/12/2021. Data cut-off was 1/9/2022. KRAS mutational status was evaluated from tumor biopsy using a new generation sequencing technique on DNA obtained to detect the most common mutations of the KRAS gene. The Oncomine™ Focus Assay panel was used in the IPATIMUP laboratory. Data was obtained from pts' clinical files and analyzed with SPSSv26.0.

Results

101 pts with NSCLC were included in our analysis, median age of 66 years (44-87), 70% male, with good performance status (70% ECOG PS 0-1). Concerning tobacco consumption, 80% were active smokers. Regarding tumor characteristics, all cases were adenocarcinomas, with 46% N1 at diagnosis and 56% of pts M1 ad initium. 10% were relapses. PDL1 status was positive in 40% of pts, and unknown in 2%. Mutation KRAS G12C was the status most frequent, identified in 36,6% of the population including c.34G>T p.Gly12Cys (36 pts) and c.35G>A p.Gly12Cys (1 pt). Followed by c.35G>T p.Gly12Val in 21,7% (22pts), c.35G>A p.Gly12Asp in 13,8% (14pts) and c.35G>C p.Gly12Ala in 6,93% (7). 1,98% (2pts) had 2 KRAS different mutation observed c.34G>T p.Gly12Cys+c.37G>A p.Gly13Ser and c.35G>T p.Gly12Val+c427G>A p.Glu143Lys where the combinations.

Conclusions

Our representative population presented a slight raise in the frequency of the G12C mutation, showing that approximately 36% of Portuguese pts with NSCLC harbor the G12C variant, thus potentially responsive to the new anti-KRAS agents.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

51P - Toxicity profile in early clinical trials with fibroblast growth factor receptor (FGFR) inhibitors (FGFRi): 10-years experience of a drug development unit

Presentation Number
51P
Lecture Time
17:35 - 17:35
Speakers
  • Katerin I. Rojas Laimito (Barcelona, Spain)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Katerin I. Rojas Laimito (Barcelona, Spain)
  • Maria Vieito Villar (Barcelona, Spain)
  • Bartomeu Fullana Grimalt (Terrassa, Spain)
  • Roger Berché (Barcelona, Spain)
  • Cinta Hierro Carbo (Badalona, Spain)
  • Omar Saavedra Santa Gadea (Barcelona, Spain)
  • Irene Braña (Barcelona, Spain)
  • Guzman Alonso Casal (Barcelona, Spain)
  • Vladimir Galvao de Aguiar (Barcelona, Spain)
  • Maria Julia Lostes Bardaji (Barcelona, Spain)
  • Mafalda Oliveira (Barcelona, Spain)
  • Helena Verdaguer (Barcelona, Spain)
  • Rafael Morales Barrera (Barcelona, Spain)
  • Carmen Garcia Duran (Barcelona, Spain)
  • Esther Zamora (Barcelona, Spain)
  • Joan Carles Galceran (Barcelona, Spain)
  • Teresa Macarulla (Barcelona, Spain)
  • Josep Tabernero (Barcelona, Spain)
  • Elena Garralda Cabanas (Barcelona, Spain)

Abstract

Background

FGFRi have been in clinical development for over 10 years, with recent approvals in patients (pts) with cholangiocarcinoma and urothelial tumors with FGFR alterations. Management of toxicity is still challenging with 10-20% of pts experiencing at least one dose reduction, especially hyperphosphatemia. The purpose of our work was to evaluate biomarker inclusion and toxicity profile of pts treated with FGFRi.

Methods

FGFRi-naïve pts included in early clinical trials with FGFRi in VHIO´s Early Drug Development Unit were included in the analysis. Adverse events were evaluated using CTCAE 4.03.

Results

A total of 72 eligible pts were treated between 2012-2022 with 6 different FGFRi. Median age was 56y, 73% female, main tumor types were breast cancer(37%) and colorectal cancer(14%). Most pts(85%) were included based on a biomarker (47% FGFR amplification, 4% FGFR mutation, 3% FGFR fusion and 22% FGFR overexpression/ligand overexpression). Most common alterations occurred in FGFR1(40%), FGFR2(19%), FGFR3(7%), FGFR4(1.7%) and 2.7% with two or more isoforms. Pts treated with panFGFRi (1-4) (76%) had statistically higher % of G2-3 toxicity than pts treated with FGFR1-2-3i(56%)(p<0.01). The commonest ≥G2 toxicity in both groups was hyperphosphatemia (7% G3 and 43% G2), fatigue (17%;G2-3), skin and nail changes (10%;G2-3), ocular/corneal toxicity(8%;G2-3) and diarrhea(6%;G2-3). 40% of pts experiencing G2-3 hyperphosphatemia needed at least 1 dose reduction, and 51.5% had to interrupt treatment. The % of G3 toxicity was statistically superior in pts treated at doses >RP2D than at doses ≤RP2D (p<0.01). Among 16 pts that received doses >RP2D, 68% exhibited G3 toxicity, 75% underwent a dose interruption, and 50% needed dose reduction. Among 56 pts treated at ≤RP2D doses 20% exhibited G3 toxicity, 25% had at least 1 dose interruption and 23% needed dose reduction.

Conclusions

Toxicity management with FGFRi remains challenging with a high proportion of pts requiring dose reductions or interruptions even at the RP2D. FGFRi that spare FGFR4 were associated with lower G3 toxicity. The impact of hyperphosphatemia highlights the potential role of more specific inhibitors.

Editorial acknowledgement

This research has been funded by the Comprehensive Program of Cancer Immunotherapy & Immunology II (CAIMI-II) supported by the BBVA Foundation (grant 53/2021). The research leading to these results has received funding from ”la Caixa” Foundation (LCF/PR/ CE07/50610001). Cellex Foundation for providing research facilities and equipment.

Legal entity responsible for the study

The authors.

Funding

This research has been funded by the Comprehensive Program of Cancer Immunotherapy & Immunology II (CAIMI-II) supported by the BBVA Foundation (grant 53/2021). The research leading to these results has received funding from “la Caixa” Foundation (LCF/PR/ CE07/50610001). Cellex Foundation for providing research facilities and equipment.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

52P - Antitumor efficacy of polypyrrole-polyethyleneimine nanocomplex to target B-cell lymphoma

Presentation Number
52P
Lecture Time
17:35 - 17:35
Speakers
  • Thi Thuy Nguyen (Hue City, Viet Nam)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Thi Thuy Nguyen (Hue City, Viet Nam)
  • Er-Yuan Chuang (Taipei City, Taiwan)
  • Po-Chun Tseng (Taipei City, Taiwan)
  • Ming-Kai Jhan (Taipei City, Taiwan)
  • Chia-Ling Chen (Taipei City, Taiwan)
  • Chiou-Feng Lin (Taipei City, Taiwan)

Abstract

Background

Traditional administration of standard lymphoma treatment can lead to poor pharmacokinetics and poor biological distribution. Recently, the nanomaterial system shows tremendous potential for therapy with a high drug-loading efficiency, good biosafety, improved bioavailability, and active targeting. This study aimed to report novel and intelligent therapeutics with particular and targeted B-cell killing in aggressive B-cell lymphoma based on nanotechnology.

Methods

A synthetic polypyrrole-polyethyleneimine nanocomplex (PPY-PEI NC) was constructed and characterized to provide its interaction with specific target B-cell lymphoma. We further investigated cell apoptosis of PPY-PEI NC in Raji cells in vitro and Raji xenograft mice models and the loss of mitochondrial transmembrane potential by Propidium iodide, Annexin V, and Rhodamine 123 staining, respectively. In addition, protein analysis identified activation of the apoptotic signaling pathway (GSK-3β, Bax, Bcl-2, Mcl-1, PARP, caspase 3) and were examined to clarify their roles.

Results

An earlier engulfment of PPY-PEI NC rapidly targeted B-cell lymphoma in clathrin-dependent endocytosis. PPY-PEI NC effectively caused B-cell lymphoma inhibition in an intrinsic pathway of apoptosis in vitro. PPY-PEI NC decreased anti-apoptotic Bcl-2 family proteins and caused classical caspase substrate activation. PPY-PEI NC induced loss of MTP while stabilizing MTP and inhibiting caspase protected B-cells from mitochondrial apoptosis. In addition, PPY-PEI NC-activated GSK-3β and inhibiting GSK-3β prevented MTP loss and mitochondrial apoptosis. In Raji subcutaneous xenograft mice model, PPY-PEI NC not only significantly inhibited the effect on tumor growth but also demonstrated no noticeable adverse effects on the treated nude mice.

Conclusions

The PPY-PEI nanocomplex strongly showed in vitro and in vivo antitumor activities. These results suggest that PPY-PEI NC has a promising application prospect as an innovative, safe and effective anti-lymphoma agent.

Legal entity responsible for the study

The authors.

Funding

The Ministry of Science and Technology, Taiwan.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

53P - Characterization of the non-ATP competitive PI3Kdelta inhibitor IOA-244 in lymphoma models: From single agent to combination screen and clinical investigation

Presentation Number
53P
Lecture Time
17:35 - 17:35
Speakers
  • Francesco Bertoni (Bellinzona, Switzerland)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Francesco Bertoni (Bellinzona, Switzerland)
  • Chiara Tarantelli (Bellinzona, Switzerland)
  • Filippo Spriano (Bellinzona, Switzerland)
  • Luciano Cascione (Bellinzona, Switzerland)
  • Elisa Civanelli (Bellinzona, Switzerland)
  • Eleonora Cannas (Bellinzona, Switzerland)
  • Afua A. Mensah (Bellinzona, Switzerland)
  • Alberto J. Arribas (Bellinzona, Switzerland)
  • Sara Napoli (Bellinzona, Switzerland)
  • Andrea Rinaldi (Bellinzona, Switzerland)
  • Anastasios Stathis (Bellinzona, Switzerland)
  • Karolina Niewola (Geneva, Switzerland)
  • Giusy Di Conza (Geneva, Switzerland)
  • Michael Lahn (Geneva, Switzerland)
  • Armando Santoro (Rozzano, Italy)
  • Carmelo Carlo-Stella (Milan, Italy)

Abstract

Background

IOA-244 is a highly selective PI3Kδ inhibitor with a differentiated chemical mechanism of action from other PI3K inhibitors. IOA-244 is currently being investigated in patients with solid tumors and lymphoma (NCT04328844). Here, we present preclinical and initial clinical data in lymphomas.

Methods

Anti-proliferative activity by MTT assay at 72h. Transcriptome analysis by total and targeted RNA-Seq on Illumina platform. Follicular lymphoma patients (pts) were treated at 20 mg and 80 mg QD continuous dosing and assessed for safety.

Results

IOA-244 showed moderate dose-dependent anti-proliferative activity, measured as area under the curve, across 66 human B and T cell lymphoma models, with IC50s < 10 mM in 18 cell lines. The activity correlated with PI3Kδ expression as measured by total RNA-Seq (R2=0.18, P=0.0009; 59 B and T cell lymphomas) or by HTG EdgeSeq Oncology Biomarker panel (R2=0.13, P=0.028; 36 B cell lymphomas). In MCL SP53 cells, IOA-244 (5μM; 24, 48, 72h) downregulated BCR, MYD88, NF-κB, MTOR and NOTCH signaling and upregulated cell cycle arrest genes (adj.P<10-10). Changes overlapped with signatures obtained with other PI3K and BTK inhibitors (adj.P <10-10). IOA-244 was then combined with 474 compounds (each given at 5μM) in two cell lines (MCL, SP-53; cutaneous T cell lymphoma, HH) and increased anti-proliferative activity was observed for combinations with inhibitors of other kinases, chemotherapy as well as metabolic and nuclear targets. NHL-FL pts treated with 20mg QD (4/4; 2 female and 2 males) and 80mg QD daily (4/4; 3 female and 1 male) had no DLT. Transient platelet reduction (G3) (N=1), AST/ALT elevation (G2) and Neutropenia (G3) (N=1) were observed in 3/8 pts, which returned to baseline without dose modifications.

Conclusions

Single-agent IOA-244 has moderate activity in vitro in lymphoma, correlated with PI3Kδ expression. Given its favorable monotherapy safety profile, IOA-244 may be used in combination with drugs identified in the present pharmacological screen.

Clinical trial identification

NCT04328844.

Legal entity responsible for the study

IOnctura SA.

Funding

IOnctura SA.

Disclosure

F. Bertoni: Financial Interests, Institutional, Other, consultancy: Helsinn, Menarini; Financial Interests, Institutional, Other, member of the Advisory Board: Phi Pharma; Financial Interests, Personal, Other, co-inventor of patent WO2019185117A1: Fondazione per l'Istituto Oncologico di Ricerca (IOR); Financial Interests, Institutional, Funding: ADC Therapeutics, Bayer AG, Cellestia, Helsinn, ImmunoGen, Menarini Ricerche, NEOMED Therapeutics 1, Nordic Nanovector ASA, PIQUR Therapeutics AG, Oncology Therapeutic Development; Financial Interests, Institutional, Research Grant: Spexis; Non-Financial Interests, Institutional, Product Samples: HTG; Other, Personal, Other, travel grant: Amgen, AstraZeneca, Jazz Pharmaceuticals; Other, Personal, Other, Travel grant: IOnctura SA, Geneva, Switzerland. C. Tarantelli: Other, Institutional, Other: IOnctura. A. Stathis: Financial Interests, Institutional, Expert Testimony: Bayer; Financial Interests, Institutional, Advisory Board: Janssen, Roche, Eli Lilly; Financial Interests, Institutional, Invited Speaker: Pfizer, Merck, Roche, Novartis, ADC Therapeutics, AbbVie, Bayer, Philogen, Cellestia, AstraZeneca. K. Niewola, G. Di Conza: Financial Interests, Personal, Full or part-time Employment: IOnctura SA. M. Lahn: Financial Interests, Personal, Officer: iOnctura; Financial Interests, Personal, Stocks/Shares: iOnctura. A. Santoro, C. Carlo-Stella: Financial Interests, Institutional, Sponsor/Funding: IOnctura SA. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

54P - Inhibition of ATM vs ATR in combination with radiotherapy affects cellular toxicity and expression of immune checkpoint molecules differently in HNSCC

Presentation Number
54P
Lecture Time
17:35 - 17:35
Speakers
  • Tina Jost (Erlangen, Germany)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Tina Jost (Erlangen, Germany)

Abstract

Background

Treatment of human papilloma virus (HPV)-negative head and neck squamous cell carcinoma (HNSCC) is still challenging, regarding its radio resistance and sparing of healthy surrounding tissue when applying radiotherapy (RT). Small molecules kinase inhibitors (smKI), targeting components of the DNA damage repair (DDR) pathway such as ATM and ATR, in combination with RT are promising to overcome these challenges. We hypothesized that inhibition von ATM vs. ATR concomitant to RT increases cellular toxicity and leads to diverse immune surface marker expression on HNSCC cells.

Methods

The effect of smKIs AZD0156 (ATMi) and VE-822 (ATRi) in combination with RT on HPV-pos. and HPV-neg. human HNSCC was analyzed. Colony formation (Co, smKI, RT, smKI+RT), immunogenic and non-immunogenic cell death (necrosis, apoptosis) were measured using Annexin/PI staining (flow cytometry). Immune-stimulating (ICOS-L, OX40-L, TNFSFR9, CD70) and immune-suppressive (PD-L1, PD-L2, HVEM) surface-marker were measured after 48h of treatment of HNSCC cells (HSC4, Cal33, UM-SCC-47, UD-SCC-2).

Results

Colony forming was significantly inhibited by smKI+RT in cancer cells, while sparing toxicity in healthy fibroblasts. Effects were more prominent in HPV-pos. compared to HPV-neg. HNSCC cells. Furthermore, ATRi demonstrated stronger cellular toxicity by inducing cell death at 0.1 μM compared to 1 μM ATMi. In contrast, ATMi only in combination with RT led to significant increase of apoptosis. After treatment with ATRi, upregulation of immune-suppressive checkpoint molecules on the cell surfaces was observed predominantly, but less influence on immune-stimulatory surface marker was found. Of note, ATMi treatment w/o RT led to even increased expression of both suppressive and stimulatory immune checkpoint molecules.

Conclusions

Inhibition of ATR shows greater toxicity, but ATM inhibition has stronger influence on the expression of immune checkpoint molecules. Taken together, combined treatment has the potential to be a therapeutic option that could improve tumor control without increasing toxicity in HNSCC.

Legal entity responsible for the study

Strahlenklinik Erlangen.

Funding

Bundesministerium für Bildung und Forschung (GREWIS-alpha, 02NUK050E).

Disclosure

The author has declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

55P - The SOS inhibitor BAY293 contributes to amplified vertical inhibition of the MAP kinase pathway in human melanoma cells

Presentation Number
55P
Lecture Time
17:35 - 17:35
Speakers
  • Martin Hohenegger (Vienna, Austria)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Martin Hohenegger (Vienna, Austria)
  • Razia Mirzahi (Vienna, Austria)
  • Nadine Szorger (Vienna, Austria)
  • Monica Ambrosio (Vienna, Austria)
  • Gerhard Hamilton (Vienna, Austria)

Abstract

Background

The driver mutation BrafV600E in human melanoma has been successfully targeted by vemuarfenib and successors. Since then vertical inhibition of the mitogen-activated protein (MAP)-kinase pathway by dual inhibition of B-Raf and MAP-kinase kinase (MEK)1/2 has been introduced in the therapy of advanced melanoma. Here we investigated in vitro inhibition of the guanine nucleotide exchange factor Son of Sevenless (SOS) by BAY293 in order to identify possible synergism with Braf or/and MEK1/2 inhibitors in human melanoma cell lines.

Methods

The 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide (MTT) assays and wound healing assays were used to monitor viability, proliferation and migration in human melanoma cell lines from the early (WM35, WM278, and WM793b) and the metastatic growth phase (A375 and 518a2), all harbouring the driver mutation BrafV600E, but wild type NRAS. Apoptosis and kinase signalling were investigated by caspase assays and Western blot analyses.

Results

The SOS inhibitor BAY293 inhibited basal ERK1/2 phosphorylation in a dose dependent manner in human metastatic A375 and 518a2 melanoma cells. Importantly, viability was not affected in these cells following BAY293 exposure times below 24h. Longer incubation times enhanced cell death and reduced gap closure in a wound healing assay. Co-application of BAY293 with inhibitors of B-Raf and/or MEK1/2 augmented these effects significantly. Concomitantly, ERK1/2 phosphorylation was significantly reduced by these drug combinations. Similar effects were seen in early stage melanoma cells so that no stage dependent differences were observed.

Conclusions

While the SOS inhibitor BAY293 is not capable to substantially inhibit viability of melanoma cell lines, co-administration with vemuarfenib and trametinib augment cytotoxicity in an additive manner. Taken together these results are indicative for additivity in vertical inhibition of the MAP kinse pathway using BAY293 also in wild type RAS melanoma.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

56P - Inhibition of HIF-2α-dependent transcription with small molecule inhibitors may provide therapeutic benefit beyond renal cell carcinoma

Presentation Number
56P
Lecture Time
17:35 - 17:35
Speakers
  • Kelsey S. Gauthier (Hayward, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Kelsey S. Gauthier (Hayward, United States of America)
  • Dana Piovesan (Hayward, United States of America)
  • Sadeesh Ramakirshnan (Pittsburgh, United States of America)
  • Soonweng Cho (Hayward, United States of America)
  • Kenneth V. Lawson (Hayward, United States of America)
  • Kai Liao (Hayward, United States of America)
  • Paul Foster (Hayward, United States of America)
  • Tzuling Cheng (Hayward, United States of America)
  • Yatrik Shah (Ann Arbor, United States of America)
  • Matthew Walters (Hayward, United States of America)

Abstract

Background

Inhibition of HIF-2α has had a substantial impact on the treatment paradigm for cancer patients with germline mutations in VHL. Preclinically, selective HIF-2α inhibitors demonstrate dramatic anti-tumor effects in xenograft models in which HIF-2α is a molecularly defined tumorigenic driver. However, therapeutic application of HIF-2α inhibitors beyond VHL-mutated clear cell renal cell carcinoma remains to be seen. In contrast to HIF-1α, HIF-2α has a more selective expression profile accompanying tailored regulons that may guide indications of interest.

Methods

The potential therapeutic benefit of HIF-2α inhibition in various oncology indications was investigated using bioinformatic, in vitro, and in vivo approaches. In mice, the role of HIF-2α in hepatocellular carcinoma (HCC) was investigated using an autochthonous high-fat diet carcinogen-induced tumor model.

Results

Consistent with the mechanism of action, inhibition of HIF-2α in various cell lines did not attenuate cell growth in 2D culture. However, significant alterations in the hypoxic gene signatures were detected, with substantial variation in HIF-2α-dependence. HIF-2α-specific gene signatures derived using pharmacological inhibition or genetic deletion applied to available datasets predicted poorer survival in multiple cancer indications, including renal (as validation) and liver. In an HCC mouse model, mice with deletion of HIF-2α in hepatocytes had significantly reduced number and size of liver nodules. These effects were apparent upon deletion of HIF-2α from birth or after tumors were established. Finally, mice treated with a HIF-2α inhibitor after tumor formation also had significantly decreased tumor burden.

Conclusions

HIF-2α plays a distinct and differential role in the transcriptional response to hypoxia in different tissue types and cell lines. In vivo data suggest that HIF-2α may contribute to the formation and progression of HCC, supporting clinical investigation of HIF-2α inhibition beyond ccRCC. AB521 is a potent and selective inhibitor that has been evaluated in healthy volunteers and is progressing through a phase Ib clinical trial in cancer patients.

Legal entity responsible for the study

Arcus Biosciences, Inc.

Funding

Arcus Biosciences, Inc.

Disclosure

K.S. Gauthier, D. Piovesan, S. Cho,K.V. Lawson, K. Liao, P. Foster, T. Cheng, M. Walters: Financial Interests, Institutional, Stocks/Shares: Arcus Biosciences; Financial Interests, Institutional, Full or part-time Employment: Arcus Biosciences. Y. Shah: Financial Interests, Institutional, Funding: Arcus Biosciences. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

57P - Deciphering the role of E2F transcription factor-1 in glutamine metabolism

Presentation Number
57P
Lecture Time
17:35 - 17:35
Speakers
  • Katharina Huber (Graz, Austria)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Katharina Huber (Graz, Austria)
  • René Dreos (Lausanne, Switzerland)
  • Sarah Geller (Lausanne, Switzerland)
  • Valentin Barquissau (Lausanne, Switzerland)
  • Dorian Ziegler (Lausanne, Switzerland)
  • Daniele Tavernari (Lausanne, Switzerland)
  • Albert Giralt (Lausanne, Switzerland)
  • Hector Gallart-Ayala (Lausanne, Switzerland)
  • Giovanni Ciriello (Lausanne, Switzerland)
  • Julijana Ivanisevic (Lausanne, Switzerland)
  • Martin Pichler (Graz, Austria)
  • Lluis Fajas (Lausanne, Switzerland)

Abstract

Background

Metabolic reprogramming is considered as a hallmark of cancer and is clinically exploited as a target for therapy. The E2F transcription factor-1 (E2F1) regulates metabolic pathways and operates as an oncogene or tumor suppressor as evident by the observation that E2f1-knockout mice develop spontaneous tumors. This discrepancy warrants a detailed investigation how E2F1’s metabolic functions control cellular proliferation and tumor growth.

Methods

Taking advantage of mouse embryonic fibroblasts (MEFs), isolated from E2f1-wild-type (E2f1+/+) and E2f1-knock-out (E2f1-/-) mice as well as transcriptomic profiles (TCGA Pan-cancer study), we investigated the biological outcomes of altered E2F1 expression on glutamine metabolism.

Results

Our data indicate that E2F1 binds the promoter of several glutamine metabolic genes. Interestingly, the gene expression levels of genes in the glutamine metabolic pathway are strongly increased in mouse embryonic fibroblasts lacking E2F1 compared to E2f1+/+ MEFs. In addition, we confirm that E2f1-/- MEFs are more efficient in metabolizing glutamine and producing precursors for proliferation. Mechanistically, we observe a co-occupancy of E2F1 and MYC on glutamine metabolic promoters, increased MYC binding after E2F1 depletion and silencing of MYC decreases the expression of glutamine metabolic genes in E2f1-/- MEFs, suggesting that MYC contributes to increased glutamine metabolic gene expression in E2f1-/- MEFs. These features have also been observed in human patient samples in a screen of 21 different cancer types and showed high relevance in uterine carcinosarcoma and soft tissue sarcomas.

Conclusions

These results confirm increased proliferation upon E2F1 loss, which is dependent on glutamine metabolism. Altogether, our results suggest that E2F1 is a regulator of glutamine metabolism and highlights potentially new targets for cancer interventions.

Legal entity responsible for the study

M. Pichler and L. Fajas.

Funding

Austrian Science Fund (J4597-B), Swiss National Science Foundation grant (31003A_143369).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

58P - Neratinib could be effective as monotherapy or in combination with trastuzumab in HER2-low-expressing breast cancer cells and organoid mode

Presentation Number
58P
Lecture Time
17:35 - 17:35
Speakers
  • Maryam Arshad (London, United Kingdom)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Maryam Arshad (London, United Kingdom)

Abstract

Background

Several studies suggested that a subset of the HER2-low breast tumours may respond to anti-HER2 treatments. The aim of this study is to understand the mechanisms of resistance to trastuzumab monotherapy and the effects of HER2 targeting treatments in HER2-low breast cancer cells.

Methods

The HER2 expression of a panel of eight breast cancer lines was assessed by immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) and their responses to trastuzumab were investigated. We further investigated the effect of different anti-HER2 treatments and/or ADAM10/17 inhibitor in two HER2 lower expressing cells.

Results

We established a panel of patient-derived organoids (PDOs) from the cancer tissues obtained from breast cancer patients to assess the effects of these drugs. Compared to sensitive high (IHC3+) HER2 expressing breast cancer cells, moderately low HER2 expressing (IHC 2+) MDA-MB-361 and MDA-MB-453 cells showed an intermediate response to trastuzumab. Trastuzumab treatment induced upregulation of HER ligand release, resulting in activation of HER receptors, which could account for trastuzumab insensitivity in these cells. Adding a dual ADAM10/17 inhibitor to inhibit the shedding of HER ligands in combination with trastuzumab only showed a modest decrease in the cell viability in HER2-low breast cancer cells and PDOs. Nevertheless, a pan-HER inhibitor, neratinib monotherapy or in the combination with trastuzumab was effective in decreasing the activation of HER family receptors in HER2-low breast cancer cells. Furthermore, neratinib was effective in reducing cell viability in PDOs, although a greater effect was seen in the combination treatment.

Conclusions

This study highlighted that neratinib in combination with trastuzumab may be effective in a small subset of moderate to low HER2 expressing breast cancers and will require further validation in a larger panel of PDOs and in future clinical studies.

Legal entity responsible for the study

M. Arshad.

Funding

PUMA Biotechnology.

Disclosure

The author has declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

59P - The influence of the ABCB1, ABCG2 and OATP1 transporters and the CYP3A enzyme on the bioavailability and tissue distribution of TPX-0131

Presentation Number
59P
Lecture Time
17:35 - 17:35
Speakers
  • Jamie Rijmers (Amsterdam, Netherlands)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Jamie Rijmers (Amsterdam, Netherlands)
  • Rolf Sparidans (Utrecht, Netherlands)
  • Nancy Loos (Amsterdam, Netherlands)
  • Maria Lebre (Amsterdam, Netherlands)
  • Alfred Schinkel (Amsterdam, Netherlands)
  • Jos Beijnen (Utrecht, Netherlands)

Abstract

Background

TPX-0131 is an ALK inhibitor developed for non-small lung cancer (NSCLC) patients that became resistant to first-line treatment. At the moment, pharmacokinetic data for TPX-0131 are still sparse. Therefore, we are interested to gain more insight into the pharmacokinetics of TPX-0131. Based on its structure and previously performed experiments with similar ALK inhibitors, TPX-0131 might be a substrate for the ABCB1, ABCG2, and OATP1 transporters and the CYP3A enzyme. Patients suffering from NSCLC often develop brain metastases. Therefore, the penetration of TPX-0131 into the brain is important to evaluate. Our main interest is therefore gaining more insight into the plasma pharmacokinetics (oral exposure) and the extent of brain penetration of TPX-0131.

Methods

An in vitro transwell experiment and in vivo experiments using genetically modified mouse models will be performed. With the use of a transmembrane in vitro transport assay, the potency of human ABCB1, human ABCG2 and murine Abcg2 to transport TPX-0131 will be evaluated. In vivo, the effect of ABCB1, ABCG2, OATP1 and CYP3A on the bioavailability and tissue distribution of TPX-0131 will be assessed. Different knockout and transgenic mouse models will be used for this purpose. The plasma concentration and tissue-to-plasma ratios of the genetically modified mouse models and wild-type mice will be compared after oral administration of 10 mg/kg TPX-0131.

Results

The in vivo data show that the brain concentration and brain-to-plasma ratio of TPX-0131 were significantly increased in the Abcb1a/b;Abcg2-/-compared to the wild-type mice. The plasma levels did not show any significant differences between these strains. Secondly, no significant differences could be observed between the Oatp1a/b-/- and wild-type mice. Furthermore, plasma levels appeared to be similar between Cyp3a-/-, Cyp3AXAV (expressing transgenic human CYP3A4 in liver and intestine in Cyp3a-/- background) and wild-type mice.

Conclusions

TPX-0131 might be a substrate for the ABCB1 and ABCG2 transporters. However, TPX-0131 is probably not a substrate for the OATP1 transporter(s) or the CYP3A enzyme.

Legal entity responsible for the study

Schinkel Group.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

60P - MAPKAP1/SIN1: A promising therapeutic target in resistant BRAF-mutated melanoma

Presentation Number
60P
Lecture Time
17:35 - 17:35
Speakers
  • Emilien Ezine (Caen, France)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Emilien Ezine (Caen, France)
  • Celeste Lebbe (Paris, France)
  • Nicolas Dumaz (Paris, France)

Abstract

Background

The PI3K/AKT/mTOR pathway plays a major role in melanoma pathogenesis and is also involved in resistance to targeted therapies (TT). The mTORC2 complex plays an important role in this pathway allowing activation of AKT and contributes to the development of BRAF-mutated (BRAFm) melanomas and their resistance to treatments. Once activated, AKT phosphorylates several effector proteins, thus regulating multiple key cellular processes, including proliferation, survival, motility, metabolism and angiogenesis.The goal of our project is to study this complex in melanoma in order to target it specifically. For this purpose, we are focusing more particularly on one principal protein of this complex: SIN1 (also called MAPKAP1) involved in the stabilizing of mTORC2 complex, in its substrate’s specificity, and a major player in the total activation of AKT.

Methods

We are analysing SIN1 expression, by quantitative RT-PCR and IHC and its interaction with other proteins, by Proximity Ligation Assay (PLA), in situ in melanoma biopsies of the French national MELBASE cohort. We will correlate these results with clinico-biological data from MELBASE to determine if there is a diagnostic or prognostic value of these analyses. In a second step, we are mapping the minimal domain of SIN1/NRAS interaction by Peptide Array to develop cell-permeable peptides to inhibit NRAS/mTORC2 interaction. Furthermore, we are evaluating whether SIN1 has also functions independent of the mTORC2 complex in melanoma, by searching for other partners of SIN1 by PLA in melanoma lines.

Results

Our preliminary results show that mTORC2 interacts with NRAS in BRAFm melanoma and that inhibiting the NRAS/mTORC2 interaction reduces the proliferation of melanoma cell lines and in particular those resistant to TT. This leads us to develop inhibitors to specifically target resistant melanomas.

Conclusions

The results of this project will provide us a better understanding of the mTORC2 signalling pathway in the development of BRAFm melanoma and its response to TT. Given the lack of a specific mTORC2 inhibitor at this time, SIN1 could be an ideal therapeutic target to aim for mTORC2.

Legal entity responsible for the study

The authors.

Funding

Fondation ARC.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

61P - The cysteine-rich protein 61 (Cyr61) contributes to tumor proliferation and invasion via HGF-mediated NF-kB signaling pathway in human hepatocellular carcinoma

Presentation Number
61P
Lecture Time
17:35 - 17:35
Speakers
  • Jiyoon Jung (Daegu, Korea, Republic of)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Jiyoon Jung (Daegu, Korea, Republic of)
  • SUNGAE Koh (Daegu, Korea, Republic of)
  • Kyunghee Lee (Daegu, Korea, Republic of)

Abstract

Background

CCN growth factor family regulates a variety of biological events such as cell proliferation, cell invasion, cell differentiation, apoptosis, and growth inhibition. Cysteine-rich protein 61 (Cyr61) is one of six secreted proteins in the CCNs. It has been shown that Cyr61 plays an important role in tumorigenesis and carcinogenesis in glioma, breast cancer, and gastric cancer, and others. However, Cyr61 has not yet been elucidated on the tumorigenesis, proliferation, and invasion of hepatocellular carcinoma, and needs to be researched and discovered. In this study, the HGF-mediated association of Cyr61, interleukin-8 (IL-8), and NF-κB (Nuclear factor kappa-light-chain-enhancer of activated B cells) and cancer cell proliferation and invasion were investigated in two types of hepatoma cell lines.

Methods

In this study, cell culture, cDNA microarray analysis, western blotting, Real-time Polymerase chain reaction, Zymography, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, Cyr61 knock-down with short hairpin RNA (shRNA), chromatin immunoprecipitation assay, Standard two chamber invasion assay.

Results

First, we confirmed that the expression level of Cyr61 was up-regulated by HGF (hepatocyte growth factor) in hepatoma cells. To identify associated pathway of HGF-induced Cyr61 about IL-8, NF-κB expression, the cells were treated with PI3K (Phosphoinositide 3-kinase) inhibitor (LY294002) and then analyzed by Western blotting. The HGF-mediated IL-8 and NF-κB levels were decreased with LY294002. The role for Cyr61 associated with IL-8 and NF-κB was determined by knock down cell of Cyr61. Cyr61-sh RNA cells showed a decreased level of IL-8 and NF-κB. HGF-mediated cell proliferation and invasion were decreased in Cyr61 knock down cell.

Conclusions

These results suggest that Cry61 plays an important role in cell proliferation and metastasis in hepatocellular carcinoma, and Cry61 may be a novel target for the prevention of progression and treatment of hepatocellular carcinoma.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

62P - Anti-malarial drug quinacrine: A potential molecule for repurposing in targeting human non-small cell lung cancer cells (NSCLC)

Presentation Number
62P
Lecture Time
17:35 - 17:35
Speakers
  • ANGSHUMAN SARKAR (Zuarinagar, India)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • ANGSHUMAN SARKAR (Zuarinagar, India)

Abstract

Background

Lung cancer is one of the leading contributors of cancer related mortalities worldwide. Out of all the lung cancer cases non-small cell lung cancer (NSCLC) accounts for approximately 80% of overall cases diagnosed. Quinacrine (QC), a synthetic drug belonging to the 9-aminoacridine another name mepacrine, repurposing quinacrine as an anticancer agent appears to be a promising strategy based on its ability to target multiple pathways.

Methods

Human lung cell lines A549 and NCI H520 were cultured for this study. Cytotoxicity of QC was assessed using resazurin dye. Cell cycle analysis was carried out using FACS. Mitochondrial membrane potential activity was analysed using JC-1 activity kit. RT-PCR analysis was carried out to evaluate the profile of various genes at their m-RNA level. Western blotting was performed using various antibodies. Microscopic analysis was carried out to understand the nuclear architecture, along with ROS estimation. GST assay kit was used for analysing GST activity.

Results

NSCLC cells adapt to the chemotherapeutics. through altering numerous cellular pathways along with enhanced activity of enzymes such as GST, MTs including altering various signalling cascades. Through our study we have discovered novel binding of quinacrine with GSTA1 and inhibiting its catalytic activity. This finding has been accompanied with detailed study of the downstream effects of this molecule and novel interaction on viability of cancer cells, cell cycle progression and apoptotic signalling cascade among two non-small cell lung cancer cell lines namely A549 and NCI H520. We have shown that quinacrine causes generation of reactive oxygen species (ROS) leading to ER stress and mitochondria mediated cell death.

Conclusions

Through a detailed study we have finally established that QC inhibited the activity of GSTA1 which inhibits cell survival and promotes apoptosis. QC possesses the advantage of targeting multiple signalling pathways via activation of apoptotic signalling cascades. Our findings add promising value to its expanding horizon of antineoplastic potential, which could be further utilized in designing combinational therapies for better and targeted destruction of NSCLC with fewer side effects.

Legal entity responsible for the study

The author.

Funding

This study was funded by the Department of Biotechnology, Govt. of India grant no.-6242-P59/RGCB/PMD/DBT/ANSR/2015 along with a supporting grant from ‘Goa Cancer Society’, Goa, India.

Disclosure

The author has declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

63P - Role of EGFR-targeted therapy in the treatment of advanced and metastatic cervical cancers

Presentation Number
63P
Lecture Time
17:35 - 17:35
Speakers
  • Abhishek Krishna (Mangalore, India)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Abhishek Krishna (Mangalore, India)
  • Athiyamaan Ms (Mangalore, India)
  • Sourjya Banerjee (Mangalore, India)
  • Johan Sunny Kilikunnel (Mangalore, India)
  • Challapalli Srinivas (Mangalore, India)
  • Dilson Lobo (Mangalore, India)

Abstract

Background

The treatment of choice for locally advanced cervical cancer is radiation therapy combined with concomitant chemotherapy using platinum complexes. 30 to 50% of patients with locally advanced stage will have a recurrence. The overall outcome for such recurrent cervical cancer is bleak, and treating such individuals remains a challenge. Due to chemotherapy's limited effectiveness and toxicity, targeted therapy agents have been tried to improve the outcomes. There are only a handful of studies wherein Gefitinib has been used to treat carcinoma of the cervix. This research looked at gefitinib’s role in treating recurrent and metastatic cervical cancer.

Methods

Patients diagnosed with metastatic cervical cancer or those who developed any recurrence post-chemoradiation or distant metastases either upfront or post-chemoradiation were eligible to be enrolled in the study. The patients were administered with gefitinib at 250 mg per day after verification of their blood parameters. Gefitinib was continued until the patient withdrew consent or the disease progressed. RECIST criteria was used to evaluate the patient's response to therapy. Toxicities were assessed with CTCAE criteria.

Results

Thirty patients with a median age of 58.5 years were included. Majority of the patients had FIGO stage IIIB disease at their initial presentation. Twenty-seven patients received salvage chemotherapy prior to Gefitinib. The median follow-up time was seen to be 6 months (3 -15 months). Two patients (7%) had a complete clinical response, 7 patients in the study (23%) was seen to have a partial response, 5 patients out of the 30 patients (17%) showed a stable disease whereas 16 patients were seen to have progressive disease (53%). The disease control rate, signified by patients with complete, partial response, and stable disease was 47%. The median PFS was noted to be 4.5 months and the 1-year PFS was 20%. None of the individuals experienced toxicity of grade 3 or higher. All toxicities were managed conservatively.

Conclusions

The EGF receptor, expressed by different types of cancers has been discovered as a promising anti-cancer target. Given the positive results and low toxicity, gefitinib could be a promising treatment approach for individuals with advanced cervical cancers.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

64P - Isothermal chemical KRAS denaturation assay for monitoring stability and inhibitors interactions

Presentation Number
64P
Lecture Time
17:35 - 17:35
Speakers
  • Randa Mahran (Turku, Finland)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Randa Mahran (Turku, Finland)
  • Niklas Vello (Turku, Finland)
  • Anita Komulainen (Turku, Finland)
  • Harri Härmä (Turku, Finland)
  • Kari Kopra (Turku, Finland)

Abstract

Background

KRAS is a frequent oncogene in human cancer and is an interesting therapeutic target due to its critical function in many malignancies. Although RAS was previously considered undruggable, recent successful identification of covalent inhibitors has shed light on the potential for cancer treatment. In drug research, RAS enzymatic activity is often measured, but methods based on RAS thermal stability (TSA) are also found useful. In these assays, RAS binding ligands stabilizes the structure over the native conformation and shifts its denaturation temperature. Among TSAs, differential scanning fluorimetry (DSF) has gained the most attention due to method’s good throughput. However, DSF lacks the needed sensitivity and results are markedly TSA dye-dependent. In addition, ligand behavior might vary when measured over the physiological temperature. In some cases, binding affinities obtained using isothermal chemical denaturation (ICD) have been found more reliable. Commonly used chemical denaturants like urea and guanidium chloride can be used to provide reliable information on ligand binding in a concentration dependent manner. However, these denaturants often require long incubation time and may affect the affinity of ionic compounds at high concentrations.

Methods

To overcome the difficulties in TSA and ICD techniques, we have developed a modification of the Protein-Probe (PP) technique, to measure isothermal protein stability at mild ICD conditions over time. The principle of this method is based on designed dual-labeled peptide-probe, and time-resolved Förster resonance energy transfer (TR-FRET) monitoring.

Results

As a model KRAS mutation, we studied KRAS(G12C) (50 nM) interaction with the most promising covalent inhibitors. Inhibitor induced stabilization of KRAS(G12C) can be monitored directly from the signals in a dose dependent manner in less than 60 min. The same effect was not seen with G12V.

Conclusions

ICD combined with the novel FRET-probe is a prominent option for TSAs, as no special equipment are needed. Isothermal concept can also potentially provide more understanding of how these proteins behave in vivo.

Legal entity responsible for the study

The authors.

Funding

Academy of Finland and Otto A. Malm Foundation.

Disclosure

H. Härmä, K. Kopra: Financial Interests, Personal, Ownership Interest: QRET Technologies Oy. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

65P - Influence of efflux and uptake transporters on the pharmacokinetics of the SYK inhibitors entospletinib and lanraplenib

Presentation Number
65P
Lecture Time
17:35 - 17:35
Speakers
  • Nancy Loos (Amsterdam, Netherlands)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Nancy Loos (Amsterdam, Netherlands)
  • Rolf Sparidans (Utrecht, Netherlands)
  • Viet Bui (Amsterdam, Netherlands)
  • Maria Lebre (Amsterdam, Netherlands)
  • Jos Beijnen (Utrecht, Netherlands)
  • Alfred Schinkel (Amsterdam, Netherlands)

Abstract

Background

Spleen tyrosine kinase (SYK) is a cytoplasmic nonreceptor tyrosine kinase important for signaling through multiple immunoreceptors across different immune cell types, including B cell receptor (BCR). Inhibition of SYK significantly inhibits the functioning of immunity. The SYK inhibitors entospletinib (IC50 = 7.6 nM) and lanraplenib (IC50 = 120 nM) are highly selective and disrupt kinase activity. Entospletinib (800 mg twice daily) could be beneficial for the treatment of a variety of B cell malignancies, whereas lanraplenib (30 mg once daily) could be used for systemic lupus erythematosus (SLE) and lupus nephritis (LN). Pharmacokinetic data are still limited for entospletinib and lanraplenib. Therefore, we are interested to gain insights into the impact of the efflux transporters P-glycoprotein (P-gp/ABCB1), breast cancer resistance protein (BCRP/ABCG2) and influx transporters organic anion transporting polypeptides (OATP’s) on the pharmacokinetics of these SYK inhibitors.

Methods

We used wild-type, single and combined Abcb1a/1b and/or Abcg2, and Oatp1a/1b deficient mouse strains. Entospletinib (10 mg/kg) and lanraplenib (10 mg/kg) were administered simultaneously to the mice by oral gavage. At several time points, blood samples were collected via the tail vein and the experiment was terminated using cardiac puncture followed by cervical dislocation.

Results

A slight increase in plasma exposure of entospletinib was observed in Abcb1a/1b;Abcg2-/- compared to wild-type, but not for lanraplenib. Brain exposure of both drugs was clearly restricted by ABC transporters, with a 3.6-fold increase in brain penetration for entospletinib and 12.1-fold for lanraplenib comparing the knockout strain with wild-type. We observed only a slight influence of OATP transporters on the tissue distribution of lanraplenib, but not of entospletinib.

Conclusions

ABC transporters play an important role in the brain disposition of entospletinib and lanraplenib, although they have only a slight impact on the plasma exposure of entospletinib. The role for the OATP uptake transporters seems to be less prominent for both drugs, but they might be involved in the tissue disposition of lanraplenib, but not of entospletinib.

Legal entity responsible for the study

Schinkel Group.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

66P - Targeting allosteric sites on PDK-1 and PLK-1 with bioactive compounds from Daucus carota as a potential therapy for triple-negative breast cancer

Presentation Number
66P
Lecture Time
17:35 - 17:35
Speakers
  • Kayode Y. Raheem (Islamabad, Pakistan)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Kayode Y. Raheem (Islamabad, Pakistan)
  • Jairus Nandwa (Islamabad, Pakistan)

Abstract

Background

Triple-negative breast cancer (TNBC) is a devastating subtype of breast cancer that lacks expression of estrogen receptors, progesterone receptors, and HER2. Targeting TNBC has been challenging due to the lack of specific therapeutic targets. However, recent studies have identified allosteric sites on the enzymes PDK-1 and PLK-1 as potential drug targets for TNBC. New medications for TNBC have considerable adverse effects, highlighting the need for more targeted and effective therapy.

Methods

In this study, we used computer-aided drug design (CADD) and theoretical chemistry techniques, including molecular docking, MM-GBSA calculation, molecular dynamic (MD) simulation, and a pharmacokinetic study, to identify bioactive compounds from Daucus carota that could bind to these allosteric sites and inhibit the activity of PDK-1 and PLK-1. DFT calculations confirmed the binding affinity and stability of the molecules. Machine-learning predictive models(QSAR) were used to find potential compounds against the two targets.

Results

Ten (10) lead compounds from Daucus carota's bioactive compounds are discovered; eight have more potential as PDK-1 and PLK-1 inhibitors than docetaxel and doxorubicin. Astragalin and Scolimoside showed substantial binding affinity and persistent interaction in the allosteric region of the two proteins. These compounds are stable and fluctuate little after 50ns of MD simulation. DFT analysis confirms enhanced bioactivity and chemical reactivity of hit compounds with favourable intramolecular charge transfer between electron-donor and electron-acceptor groups. The predicted QSAR model showed the efficiency of certain compounds as PDK-1 and PLK-1 inhibitors (pIC50).

Conclusions

This study provides evidence that targeting PDK-1 and PLK-1 with bioactive compounds from Daucus carota may be an effective method for the treatment of TNBC.

Editorial acknowledgement

Associate Professor Muhammad Muddassar COMSATS University Islamabad.

Legal entity responsible for the study

K.Y. Raheem.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

67P - Molecular testing and treatment of patients with advanced solid tumors harboring an NTRK gene fusion: Interim results of the REALTRK registry

Presentation Number
67P
Lecture Time
17:35 - 17:35
Speakers
  • Corinne Vannier (Freiburg im Breisgau, Germany)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Corinne Vannier (Freiburg im Breisgau, Germany)
  • Uwe M. Martens (Heilbronn, Germany)
  • Annalen Bleckmann (Muenster, Germany)
  • Matthias Zaiss (Freiburg im Breisgau, Germany)
  • Nina Burkhart (Hamburg, Germany)
  • Thomas Decker (Ravensburg, Germany)
  • Stefan Kaldune (Rosenheim, Germany)
  • Christian Langer (Kempten, Germany)
  • Marion Looß (Freiburg im Breisgau, Germany)
  • Larissa Hillebrand (Freiburg im Breisgau, Germany)
  • Ott Vahtrik (Freiburg im Breisgau, Germany)
  • Melanie Frank (Freiburg im Breisgau, Germany)
  • Beate Niemeier (Freiburg im Breisgau, Germany)
  • Karin M. Potthoff (Heidelberg, Germany)
  • Benjamin Kasenda (Basel, Switzerland)

Abstract

Background

Neurotrophic Tyrosine Receptor Kinase gene fusions involving either NTRK1, NTRK2 or NTRK3 (encoding the tropomyosin receptor kinases TRKA, TRKB and TRKC, respectively) are oncogenic drivers. The overall prevalence of NTRK fusion-positive tumors is ∼0.30%, though frequencies vary by tumor type, with >90% reported for some rare tumor types such as secretory carcinoma of the breast and salivary glands. In Europe, the TRK inhibitors (TRKi) entrectinib and larotrectinib, both showing high clinical activity and favorable safety profiles, have received tumor-agnostic approval based on pooled data of non-comparative phase I/II clinical trials with limited patient numbers.

Methods

REALTRK is a retro- and prospective, observational, intersectoral, multicenter cohort study in Germany and Switzerland (NCT04557813). Enrollment of 120 patients (pts) with advanced solid tumors harboring an NTRK fusion diagnosed with a validated assay according to ESMO recommendations is planned. Pts with detailed information on NTRK testing qualify for complete documentation, including demographic and clinical characteristics, details on NTRK testing, treatment, outcome, and safety of TRKi. Furthermore, physician-reported factors on NTRK testing and treatment decision making as well as patient-reported outcomes on quality of life (EORTC QLQ-C30) are assessed.

Results

At data cut-off (Sept 30th, 2022), 18 pts have been enrolled at 13 sites in Germany and Switzerland. Of those, 12 pts were eligible for complete documentation. Median age at diagnosis of NTRK fusion was 60.7 years and median time from NTRK fusion diagnosis to start of TRKi therapy, received by 11 pts, was 0.9 months. Tumor entities and data on NTRK fusions are presented in the table.

Conclusions

REALTRK provides valuable data on NTRK testing and treatment reality of pts with NTRK fusion-positive solid tumors and generates clinically relevant real-world evidence. Table: 67P

Tumor entity NTRK gene Fusion partner gene
CNS tumor (including glioma) NTRK1 PRCC
NTRK2 BCR
NTRK3 ARHGEF7
Colorectal cancer NTRK1 LMNA
TPM3
TP53
NTRK3 EML4
Lung cancer (NSCLC/SCLC) NTRK1 KIF5B
TPM3
Soft tissue sarcoma NTRK3 ETV6
Thyroid cancer NTRK3 ETV6

Legal entity responsible for the study

iOMEDICO AG.

Funding

Roche GmbH.

Disclosure

A. Bleckmann: Financial Interests, Personal, Advisory Role: Alexion, Gilead, Novartis, Bristol-Myers Squibb, Bayer, Servier, Roche, AstraZeneca, Takeda, Merck, BeiGene, MSD, Lilly, ArtTempi, Jannsen-Cilag, Amgen, Boehringer Ingelheim. M. Zaiss: Financial Interests, Personal, Advisory Board: AbbVie, AstraZeneca, Bristol-Myers Squibb, Celgene, Esai, Gilead, Hexal, Jannsen, Lilly, Novartis, Pierre-Fabre, Pfizer, Roche, Vifor. T. Decker: Financial Interests, Personal, Advisory Board: Novartis, iOMEDICO. B. Kasenda: Financial Interests, Personal, Advisory Board: Astellas, Roche. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

68P - Investigating CDK4/6 palbociclib resistance mechanisms in MCF7 breast cancer cell line

Presentation Number
68P
Lecture Time
17:35 - 17:35
Speakers
  • Heloise Beutier (Rushden, United Kingdom)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Heloise Beutier (Rushden, United Kingdom)

Abstract

Background

Deregulation of the cyclin D-CDK4/6-INK4-RB pathway leading to uncontrolled increased cell proliferation, is observed in various cancer types including breast cancer. Palbociclib is one of the selective CDK4/6 inhibitors approved for hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative advanced or metastatic breast cancer. Despite initial response to Palbociclib, intrinsic or acquired resistance emerges eventually. Understanding resistance mechanisms to CDK4/6 inhibitors enables us to design drug combination regimen to overcome or delay resistance onset, to identify biomarkers to predict therapy outcome that can be utilized to stratify patients who benefit from the treatment and to identify novel druggable targets in the CDK4/6 drug resistance milieu.

Methods

To investigate proteome alterations leading to Palbociclib resistance, we established resistant sublines of MCF7 breast cancer cell line by culturing the cells in a) under constant pressure of 1uM Palbociclib and b) in drug holiday after 1uM Palbociclib for several cycles. We then performed RPPA (Reverse Phase Protein Array) technology to analyse 384 protein targets in the two groups compared to parental Palbociclib sensitive MCF7 cells as well as comparing the two resistant sublines.

Results

Pathways analyses in sensitive vs. resistance cells showed significant changes in cell cycle dependent and independent pathways including EGFR pathway, p53 pathway, and the JAK-STAT signalling pathways.

Conclusions

These pathways are closely related to the occurrence, development, and metastasis of cancer. Drugs that target these pathways may provide new strategies for combination therapies in Palbociclib-resistant patients. Also, to validate the proteome alterations, we will perform RNAseq analysis on the cell line pairs to investigate whether the proteome alteration is observed at transcriptome level.

Legal entity responsible for the study

Pharmaron.

Funding

Pharmaron.

Disclosure

The author has declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

69P - Osimertinib is selective against NSCLC cells and modulates the multidrug-resistant phenotype in patient-derived cell cultures and co-cultures of NSCLC cells and fibroblasts

Presentation Number
69P
Lecture Time
17:35 - 17:35
Speakers
  • Sofija Jovanović Stojanov (Belgrade, Serbia)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Sofija Jovanović Stojanov (Belgrade, Serbia)
  • Ana Podolski-Renic (Belgrade, Serbia)
  • Jelena Dinić (Belgrade, Serbia)
  • Miodrag Dragoj (Belgrade, Serbia)
  • Mirna Jovanović (Belgrade, Serbia)
  • Ana Stepanović (Belgrade, Serbia)
  • Ema Lupšić (Belgrade, Serbia)
  • Radovan Bajović (Belgrade, Serbia)
  • Sofija Glumac (Belgrade, Serbia)
  • Dragana Marić (Belgrade, Serbia)
  • Maja Ercegovac (Belgrade, Serbia)
  • Milica Pešić (Belgrade, Serbia)

Abstract

Background

Osimertinib belongs to the third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) approved for metastatic EGFR-mutant non-small-cell lung carcinoma (NSCLC) patients. Herein, we studied osimertinib selectivity towards NSCLC cells, its efficacy dependence on the EGFR mutation status, and its ability to evade the classical mechanism of multidrug-resistance (MDR) mirrored in the increased expression of main ATP Binding Cassette (ABC) transporters (ABCB1, ABCC1, and ABCG2).

Methods

Primary patient-derived cultures were established from the NSCLC resections. After short-term culturing (2-3 weeks), a mixed population of cancer and non-cancer cells (around a ratio of 1:1) and two co-cultures of NSCLC cell lines (sensitive NCI-H460 and MDR NCI-H460/R) with lung fibroblasts MRC-5 were treated with 8 chemotherapeutics (cisplatin, carboplatin, paclitaxel, docetaxel, etoposide, vinorelbine, gemcitabine, and pemetrexed) as well as osimertinib. The maximum concentration reached in human plasma to which the patient is exposed during therapy (Cmax) was set as an upper limit and four lower concentrations were also applied during the study. Immunofluorescence assay enabling discrimination of epithelial cancer cells positive to a cocktail of antibodies against cytokeratin 8/18 vs. negative mesenchymal non-cancer cells was conducted using high-content imager ImageXpress Pico (Molecular Devices) with CellReporterXpress 2.9 software. Within the same immunoassay, MDR markers (ABCB1, ABCC1, and ABCG2) were analyzed by corresponding antibodies.

Results

Osimertinib showed selectivity against NSCLC cells, particularly in the patient-derived cell culture without EGFR mutations. Other chemotherapeutics were not selective towards cancer cells, on contrary, they showed higher cytotoxicity in non-cancer cells. Osimertinib did not change the expression of ABCB1 in cancer cells, but it significantly decreased the expression of ABCC1 and ABCG2 transporters in cancer and non-cancer cells.

Conclusions

Osimertinib can be valuable as a selective anticancer drug and an MDR modulator even in NSCLC without EGFR mutations.

Legal entity responsible for the study

The authors.

Funding

Science Fund of the Republic of Serbia - TargetedResponse - 7739737.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

Molecular profiling applications

Lecture Time
17:35 - 17:35
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Cocktail & Poster Display session Abstract session

71P - Molecular Tumor Board at the European Institute of Oncology: An early Italian precision oncology experience

Presentation Number
71P
Lecture Time
17:35 - 17:35
Speakers
  • Edoardo Crimini (Milan, Italy)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Edoardo Crimini (Milan, Italy)
  • Matteo Repetto (Milan, Italy)
  • Luca Boscolo Bielo (Milan, Italy)
  • Elena Guerini-Rocco (Milan, Italy)
  • Liliana Ascione (Milan, Italy)
  • Cristina Zanzottera (Milan, Italy)
  • Luca Mazzarella (Milan, Italy)
  • Alberto Ranghiero (Milan, Italy)
  • Carmen Belli (Milan, Italy)
  • Carmen Criscitiello (Milan, Italy)
  • Angela Esposito (Bristol, United Kingdom)
  • Massimo Barberis (Milan, Italy)
  • Giuseppe Curigliano (Milan, Italy)

Abstract

Background

Precision oncology personalizes treatments for cancer patients, aiming to improve their clinical outcomes. ESMO Scale for Clinical Actionability of Molecular Targets (ESCAT) allows evidence-based evaluation of genomic findings. Molecular tumor boards (MTBs) multi-disciplinary expertise enables ESCAT implementation and personalized treatment choice.

Methods

We retrospectively reviewed outcomes of patients discussed at European Institute of Oncology MTB between June 2019 and June 2022. We performed descriptive statistics and survival analysis comparing patients that received molecularly-matched treatments (MMT) and non-matched treatments (nMMT).

Results

MTB discussed 251 patients with cancer encompassing 26 different primary cancer types. Breast cancer accounted for 25.5% of the total. 188 (74.6%) patients had at least one actionable alteration. After MTB discussion, 76 patients received MMT while 76 received nMMT. Patients receiving MMT displayed higher overall response rate (37.3% vs 12.9%), median progression-free-survival (PFS 5.8 months, 95% CI 4.1–7.5 vs 3.6 months, 95% CI 2.5–4.8, p=0.041; HR 0.679, 95% CI 0.467–0.987) and median overall-survival (mOS 35.1 months, 95% CI not evaluable vs 8.5 months, 95% CI 3.8 – 13.2; HR 0.431, 95% CI 0.250 – 0.744, p=0.002). Superiority in OS and PFS persisted in multivariable models. Among 61 pretreated patients receiving MMT, 37.5% had superior PFS compared to the prior line (PFS2/PFS1 ratio ≥ 1.3). Higher OS (p=0.001) and PFS (p=0.049) were observed for patients with higher actionability evidence (ESCAT tier I) alterations, while no difference was observed in lower evidence levels. Table: 71P
Most frequent actionable genes

Alteration Total number Treated patients Drug administered ESCAT at discussion ORR
PIK3CA 51 4 Alpelisib IA 1/4 (25%)
BRCA2 32 19 PARP-i IA 7/19 (37%)
BRAF V600 20 2 BRAF + MEK-i IIIA 1/2 (50%)
ERBB2 17 1 Trastuzumab + pertuzumab IIIA 1/1 (100%)
TMB-HIGH 16 4 Anti-PD(L)-1 IC 2/4 (50%)
ATM 15 2 PARP-i X 0/2 (0%)
MSI-HIGH 14 8 Anti-PD(L)-1 IC 4/8 (50%)
BRCA1 11 9 PARP-i IA 2/9 (22%)
RET 10 10 RET-i IIIA 7/10 (70%)

Conclusions

We report real-world data of the first Italian experience of MTB application in clinical practice. Our work shows that MTBs can yield valuable clinical benefit in terms of OS and PFS. Biomarkers with lower actionability ESCAT level appear to be linked to lower clinical benefit.

Clinical trial identification

This study is approved by IEO Ethical Committee with the reference number UID 3572.

Editorial acknowledgement

None

Legal entity responsible for the study

E. Crimini, M. Repetto.

Funding

Has not received any funding.

Disclosure

E. Guerini-Rocco: Financial Interests, Personal, Advisory Role: AstraZeneca, GSK, Roche, Termo Fisher; Financial Interests, Personal, Advisory Board: Novartis. C. Criscitiello: Financial Interests, Personal, Invited Speaker: Pfizer, Novartis, Eli Lilly, Roche, Gilead; Financial Interests, Personal, Advisory Board: MSD, Seagen, AstraZeneca, Daiichi Sankyo. A. Esposito: Financial Interests, Personal, Speaker’s Bureau: Novartis. G. Curigliano: Financial Interests, Personal, Invited Speaker: Roche, AstraZeneca, Daiichi Sankyo, Novartis, Pfizer; Financial Interests, Personal, Advisory Board: Ellipsis, Roche, AstraZeneca, Daiichi Sankyo, Lilly, Pfizer, Veracyte, BMS, Merck, Exact Sciences, Celcuity; Financial Interests, Institutional, Research Grant, Investigator Initiated Trial: Merck; Financial Interests, Institutional, Funding, Phase I studies: BMS, Novartis, AstraZeneca, Daiichi Sankyo, Roche, Blueprint Medicine, Kymab, Astellas, Sanofi, Philogen; Financial Interests, Institutional, Invited Speaker, Phase I clinical basket trial: Relay Therapeutics; Non-Financial Interests, Personal, Officer, Italian National Health Council as Advisor for Ministry of Health: Consiglio Superiore di Sanità; Non-Financial Interests, Personal, Advisory Role, Member of the Scientific Council. Patient Advocacy Association: Europa Donna; Non-Financial Interests, Personal, Advisory Role, Cancer Research Foundation: Fondazione Beretta; Non-Financial Interests, Personal, Invited Speaker, No compensation for this role. This a public national company for cancer prevention: Lega Italiana Lotta ai Tumori; Non-Financial Interests, Personal, Officer, Member of the Advisory Council: EUSOMA. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

72P - MDM alterations in patients with advanced or metastatic cancers

Presentation Number
72P
Lecture Time
17:35 - 17:35
Speakers
  • Iwona Lugowska (Warsaw, Poland)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Iwona Lugowska (Warsaw, Poland)
  • Hanna M. Kosela Paterczyk (Warsaw, Poland)
  • Mateusz Iwanski (Rzeszow, Poland)
  • Monika Chelstowska (Warsaw, Poland)
  • Anna Dawidowska (Warsaw, Poland)
  • Sylwia Jaczewska (Warsaw, Poland)
  • Agnieszka Napora (Warsaw, Poland)
  • Marcin Witczak (Warsaw, Poland)
  • Andrzej Tysarowski (Warsaw, Poland)
  • Piotr Rutkowski (Warsaw, Poland)

Abstract

Background

Oncogenic alterations in MDM represent important therapeutic targets in various type of cancer. We aimed to assess the prevalence of its incidence together with p53 status in advanced solid tumors based on commercially available NGS panel and to monitor patients’ clinical pathway care.

Methods

Between 2019-2022, 740 patients were included for molecular testing in the Center of Excellence for Precision Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland. The population with MDM alterations was extracted for this analysis. We analyzed their therapeutic options over the years, access to clinical trials and impact on patients’ survival.

Results

In our cohort, 48 patients (6.5%) had been identified with MDM alterations as amplification (38), rearrangements (5), and SNPs (5). The most common histology were sarcomas (22), cholangiocarcinoma (3), gastric cancer (3), colorectal cancer (3), breast cancer (3), ovarian cancer (2), salivary gland cancer (2), melanoma (2), urothelial cell carcinoma (2), other (6). In sarcoma, 20/22 had amplification of MDM2, 6/22 rearrangements (MDM2-PPFIA2 rearrangement, MDM2-CSMD1 rearrangement, MDM2-EYS rearrangement) and SNPs (I388V and R332P). The predominant histology types were liposarcomas 14 (64%), sporadically MDM changes were found in rhabdomyosarcoma, osteosarcoma, angiosarcoma, leiomyosarcoma, undifferentiated spindle cell sarcoma and malignant peripheral nerve sheath tumour.

Conclusions

The detection of MDM alterations allows for access to targeted therapies, especially in sarcomas, which may be an important therapeutic option in these rare cancers.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

I. Lugowska: Financial Interests, Personal, Invited Speaker, The reports of clinical trials: Roche, BMS, Macrogenics, Amgen; Financial Interests, Institutional, Other, Research grants: Roche; Financial Interests, Institutional, Other, Research grant: Agenus; Financial Interests, Personal and Institutional, Invited Speaker: Agenus, Roche, BMS, Janssen, Astra, Incyte, Macrogenics, Checkpoint Inhibitors, Celon, Pfizer, MSD, DeBio; Non-Financial Interests, Personal, Project Lead: MSCI; Non-Financial Interests, Personal, Advisory Role, Board Member: EORTC. H.M. Kosela Paterczyk: Financial Interests, Personal, Research Grant: Roche, ABM; Financial Interests, Personal, Invited Speaker: BMS, MSD, Pierre Fabre; Financial Interests, Personal, Principal Investigator: Rain. M. Chelstowska: Financial Interests, Personal, Research Grant: Roche, ABM, MSD, BMS, Pfizer, Janssen. A. Dawidowska: Financial Interests, Personal, Research Grant: Roche, MSD, ABM, BMS, Pfizer, Janssen. S. Jaczewska: Financial Interests, Personal, Research Grant: Roche, BMS, ABM, Pfizer; Financial Interests, Personal, Invited Speaker: Janssen. A. Napora, A. Tysarowski: Financial Interests, Personal, Research Grant: Roche. P. Rutkowski: Financial Interests, Personal, Invited Speaker, Honoraria for lectures: MSD, BMS, Pierre Fabre; Financial Interests, Personal, Advisory Board: MSD, BMS, Pierre Fabre, Merck, Sanofi, Blueprint Medicines, Philogen; Financial Interests, Personal, Invited Speaker: Merck, Sanofi, Novartis; Financial Interests, Institutional, Research Grant for ISS: Pfizer; Financial Interests, Institutional, Funding, Research Grant: BMS; Non-Financial Interests, Personal, Invited Speaker: Polish Society of Surgical Oncology; Non-Financial Interests, Personal, Officer: ASCO; Non-Financial Interests, Personal, Invited Speaker, President Elect: Polish Oncological Society. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

73P - Automated detection of typical and atypical mitotic figures for improving survival prediction in breast cancer

Presentation Number
73P
Lecture Time
17:35 - 17:35
Speakers
  • Saima Ben Hadj (Paris, France)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Saima Ben Hadj (Paris, France)
  • David Wallis (Paris, France)
  • Marc Aubreville (Ingolstadt, Germany)
  • Christof Bertram (Vienna, Austria)
  • Rutger Fick (Paris, France)

Abstract

Background

The number of typical and atypical (defined as mitoses with any morphological appearance other than the typical forms) mitotic figures (MFs) and a high atypical-to-typical mitosis ratio are strongly associated with tumour aggressivity, survival rates, and a predictor of poor response to chemotherapy in breast cancer. Manual detection is time consuming, especially on whole slide images (WSIs). An automated approach is therefore necessary to investigate these aspects on a larger scale. We demonstrate that deep learning can be used to automate this detection, improving on the performance of pathologists.

Methods

All MFs in the mammary carcinoma dataset (21 hematoxylin and eosin (H&E)-stained WSIs with ∼14 000 MFs and ∼36 000 hard negatives) were labelled as typical or atypical. These slides (originally scanned on a Leica scanner) were then rescanned on six other scanners (2x Hammamatsu, 2x 3DHISTECH, Philips, Olympus), and the annotations were registered. This gave a large, multi-scanner dataset, which was used to train a YOLOv6 deep learning object detection model. For testing, all MFs in the (human) TUPAC16 and MIDOG21 datasets were labelled by two pathologists as either typical or atypical. In cases of disagreement, a third reader gave a consensus. We used the alternative version of the TUPAC16 dataset provided by the same authors as the MIDOG21 dataset to reduce potential label bias. We then ran our model on these images and compared the mean average precision (mAP) vs the consensus to the mAPs of the two individual pathologists vs the consensus.

Results

The mAP of our model (0.80) was higher than the average mAP of the two pathologists (0.75, p<0.05), showing that the model can successfully automate the process of MF detection. There was considerable disagreement in the labelling by the two pathologists (14% of cases). By automating the process we reduce this variability, meaning we can more consistently predict clinical outcomes (e.g. survival rates) from our results.

Conclusions

The numbers of both typical and atypical MFs are indicators of patient survival and response to treatment. We have demonstrated an automated deep learning model that can accurately detect these figures and could thus be used for patient survival prediction.

Legal entity responsible for the study

The authors.

Funding

Tribun.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

74P - Tumor agnostic comparison of immunohistochemistry and next-generation sequencing in detecting ALK fusions and assessment of ALK tyrosine kinase inhibitor efficacy

Presentation Number
74P
Lecture Time
17:35 - 17:35
Speakers
  • Monica F. Chen (New York, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Monica F. Chen (New York, United States of America)
  • Soo Ryum Yang (New York, United States of America)
  • Jason Chang (New York, United States of America)
  • Mark G. Kris (New York, United States of America)
  • Alexander Drilon (New York, United States of America)

Abstract

Background

Reliable methods to identify anaplastic lymphoma kinase (ALK) fusions are critical to matching patients to ALK tyrosine kinase inhibitors (TKIs) therapy, on or off trial. Various methods including FISH have been used, but immunohistochemistry (IHC) and next-generation sequencing (NGS) are most commonly employed. Evaluating the concordance of IHC and NGS is key, particularly in non-lung cancers where data is sparse.

Methods

NGS+ (MSK-IMPACT DNA hybrid capture NGS and/or RNA anchored multiplex PCR) and/or IHC+ (clone: D5F3) patients with cancers of any histology were identified as ALK+. ALK IHC was scored as negative (0), equivocal (e: 1+, 2+) or positive (3). Concordance of ALK detection (number of NGS+ and IHC+/total number of patients with NGS and IHC) was calculated. For patients with metastatic disease treated with any ALK TKI in the first-line (1L) setting, progression-free survival (PFS) was reported.

Results

347 ALK+ solid tumor patients were identified. As expected, the majority (96%, n=336) had lung cancer, however, 11 patients with 11 unique non-lung cancer histologies were found (3 gastrointestinal, 2 gynecologic, 1 breast, 1 thyroid, 1 primary brain tumor, 1 DLBCL, 1 PEComa, and 1 CUP). 57% had EML4-ALK fusions; 36 non-EML4 ALK rearrangements were identified, including four novel fusions (PEKHA7-ALK, ZFPM2-ALK, TRIM24-ALK, ALK-MYO3B). ALK was evaluated by IHC alone in 83 patients (23.9%). The concordance rate between NGS and IHC was 85%. Among discordant cases, 11% (n=28) were IHC+/NGS-, 24% (n=63) were IHCe/NGS-, 3% (n=8) were IHCe/NGS+, and 0.4% (n=1) was IHC-/NGS+. The most frequent ALK TKIs were alectinib (n= 87, 58%) and crizotinib (n= 56, 38%). PFS on 1L ALK TKIs for patients with IHC+/NGS+ (n=134), IHC-/NGS+(n=1), IHC+/NGS- (n=8), IHCe/NGS+ (n=4), IHCe/NGS- (n=1) was 26 months, 26 months, 39 months, 41 months, 9 months respectively.

Conclusions

In a population including multiple tumor types, NGS and IHC were highly concordant in ALK fusion detection. ALK TKI benefit may be observed in cases with discordant testing, in which only one assay detects a putative ALK fusion.

Legal entity responsible for the study

The authors.

Funding

NIH Cancer Center grant: P30CA008748.

Disclosure

M.G. Kris: Financial Interests, Personal, Research Grant: Boehringer Ingelheim, National Lung Cancer Partnership, Pfizer, PUMA, Stand up to Cancer; Financial Interests, Personal, Advisory Role: Ariad, AstraZeneca, Bind Bioscience, Boehringer Ingelheim, Chug Pharma, Clovis, Covidien, Daiichi Sankyo, Esanex, Genentech; Financial Interests, Personal, Invited Speaker: Boehringer Ingelheim, Novartis, Millenium, Pfizer, Roche. A. Drilon: Financial Interests, Personal, Advisory Board: Ignyta/Genentech/Roche, Loxo/Bayer/Lilly, Takeda/Ariad/Millennium, TP Therapeutics, AstraZeneca, Pfizer, Blueprint Medicines, Helsinn, BeiGene, BerGenBio, Hengrui Therapeutics, Exelixis, Tyra Biosciences, Verastem Oncology, MORE Health, AbbVie, 14ner/Elevation Oncology, Remedica Ltd, ArcherDX, Monopteros, Novartis, EMD Serono, Melendi, Liberum, Repare RX, Amgen, Janssen, EcoR1, Monte Rosa; Financial Interests, Personal, Other, CME: Medscape, Onclive, PeerVoice, Physicians Education Resources, Targeted Oncology, Research to Practice, PeerView Institute, Paradigm Medical Communications, WebMD, MJH Life Sciences, Med Learning, Imedex, Answers in CME, Medscape, Clinical Care Options, AiCME; Financial Interests, Personal, Other, CME, Consulting: Axis; Financial Interests, Personal, Other, Consulting: Nuvalent, Merus, EPG Health, mBrace, Harborside Nexus, Ology, TouchIME, Entos, Treeline Bio, Prelude, Applied Pharmaceutical Science, Inc; Financial Interests, Personal, Invited Speaker: Chugai Pharmaceutical, Remedica Ltd, RV More; Financial Interests, Personal, Stocks/Shares: Treeline Biosciences; Financial Interests, Personal, Royalties: Wolters Kluwer; Financial Interests, Personal, Other, stocks: mBrace; Financial Interests, Institutional, Funding, Research funding: Pfizer, Exelixis, GlaxoSmithKline, Teva, Taiho, PharmaMar; Financial Interests, Personal, Funding, Research: Foundation Medicine; Non-Financial Interests, Personal, Member: ASCO, AACR, IASLC; Other, Personal, Other, Food/Beverage: Merck, PUMA, Merus; Other, Personal, Other, Other: Boehringer Ingelheim. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

75P - Target identification by TIME phenotypes

Presentation Number
75P
Lecture Time
17:35 - 17:35
Speakers
  • Robert Seitz (Irvine, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Robert Seitz (Irvine, United States of America)
  • Brian Ring (Irvine, United States of America)
  • Catie Cronister (Irvine, United States of America)

Abstract

Background

We have previously shown that a large gene set can classify the tumor immune microenvironment (TIME) of patients with epithelial tumors into one of three phenotypes: immune inflamed (immune hot), immuno-suppressive (immune cold), or immune inert (immune cold). Given that the tumor is constantly evolving, identifying key genes where changes in mutation or cell signaling is associated with a change in phenotypes can lead to identification of potential new targets. Here we show three bioinformatic methodologies for this purpose.

Methods

The first approach to identifies genes hypermethylated in one phenotype and hypomethylated in another as methylation patterns can be surrogates for mutations that drive a transition across the TIME. Second, MUTECT2 annotation was used to detect high impact mutations which were associated with the phenotypes. Third, the IntAct database was used to identify potential cell signaling pairings where one gene was associated with patients classified as hot and a second gene classified patients as cold. All three of these approaches yielded genes with known targeted therapies in active clinical trials.

Results

The methylation approach identified VEGFR2 as a gene where mutations could likely affect TIME phenotypes. VEGFR2 is routinely used in clinical trials and has an approved drug on the market (ramucirumab), and mutations in VEGFR2 are seen to affect patient prognosis and drug sensitivity. With the MUTECT2 approach, mutations in STK11 were identified. Several clinical trials are examining the role that mutations in STK11 can play in altering the treatment paradigm between immunotherapy and KRAS targeted therapy. Finally, the IntAct approach identified the interaction between CRCX4 and CXCL12, known to be involved in TIME evolution and with several clinical trials being conducted designed to disrupt this interaction.

Conclusions

Utilizing a curated gene list to classify patients into TIME phenotypes and then using bioinformatic methods to find genes which may be driving transition across immune of phenotypes resulted in identifying known and well validated targets from current clinical trials. The results argue for a more careful examination of other genes that resulted from this methodology as potential targets for immunotherapy.

Legal entity responsible for the study

The authors.

Funding

Oncocyte.

Disclosure

R. Seitz: Financial Interests, Personal, Advisory Role: Oncocyte, Inc. B. Ring, C. Cronister: Financial Interests, Personal, Full or part-time Employment: Oncocyte, Inc.

Collapse
Cocktail & Poster Display session Abstract session

76P - Prediction of response to palliative chemotherapy by circulating tumor DNA (ctDNA) kinetics in metastatic pancreatic cancer

Presentation Number
76P
Lecture Time
17:35 - 17:35
Speakers
  • Patrick Kirchweger (Linz, Austria)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Patrick Kirchweger (Linz, Austria)
  • Alexander Kupferthaler (Linz, Austria)
  • Jonathan Burghofer (Linz, Austria)
  • Gerald Webersinke (Linz, Austria)
  • Emina Jukic (Innsbruck, Austria)
  • Simon Schwendinger (Innsbruck, Austria)
  • Helwig Wundsam (Linz, Austria)
  • Matthias Biebl (Linz, Austria)
  • Andreas L. Petzer (Linz, Austria)
  • Holger Rumpold (Linz, Austria)

Abstract

Background

Circulating tumor DNA (ctDNA) represents a promising prognostic biomarker for predicting relapse and overall survival in patients with metastatic pancreatic cancer (PC). To test the clinical applicable prognostic value of ctDNA dynamics during treatment, we aimed to detect response to treatment ahead of radiological restaging.

Methods

ctDNA detection using liquid biopsy (ddPCR utilizing KRAS G12/13 (and, if negative, Q61) commercial test kits) was prospectively performed on 70 patients with stage IV PC (i) prior to initiation of systemic chemotherapy and (ii) serially every two weeks until restaging.

Results

Detection rate at baseline was 64.3% (45/70). Reduction of ctDNA levels below 57.9% of its baseline value at week 2 after treatment initiation was significantly predictive for response to treatment (AUC=0.918, sensitivity 91.67%, specificity 100%) and was associated with prolonged overall survival (OS) (5.7 vs. 11.4 months, p=0.006) and progression free survival (PFS) (2.5 vs. 7.7 months, p<0.000) regardless of treatment line. Pretherapeutic ctDNA detection was independently associated with worse OS in patients receiving first line regimen (7 vs. 11.3 months, p=0.046) and regardless of treatment line (11.4 vs. 15.9 months, p=0.045) and associated with worse PFS (3.4 vs. 10.8 months, p=0.018).

Conclusions

The dynamic change of ctDNA during systemic treatment allows the prediction of treatment response and is associated with OS and PFS. Progressive disease was correctly predicted in 100% of patients with preemptive detectable ctDNA after 2 weeks (ctDNA) compared to 12 weeks with current gold standard (CT), enabling change of treatment >80% earlier hereafter.

Legal entity responsible for the study

H. Rumpold.

Funding

Vinzenzgruppe Austria and Krebshilfe Oberösterreich.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

77P - Impact of germline HLA genotypes on clinical outcomes in patients (pts) with solid tumors treated with immunotherapy

Presentation Number
77P
Lecture Time
17:35 - 17:35
Speakers
  • Katerin I. Rojas Laimito (Barcelona, Spain)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Katerin I. Rojas Laimito (Barcelona, Spain)
  • Maria Vieito Villar (Barcelona, Spain)
  • Daniel López (Barcelona, Spain)
  • Roger Berché (Barcelona, Spain)
  • Omar Saavedra Santa Gadea (Barcelona, Spain)
  • Guzman Alonso Casal (Barcelona, Spain)
  • Vladimir Galvao de Aguiar (Barcelona, Spain)
  • Maria Julia Lostes Bardaji (Barcelona, Spain)
  • Irene Braña (Barcelona, Spain)
  • Josep Tabernero (Barcelona, Spain)
  • Elena Garralda Cabanas (Barcelona, Spain)

Abstract

Background

Germline genetic factors can potentially be good biomarkers since they influence immune traits in many diseases. Human leukocyte antigens (HLAs) are expressed in a variety of cells, including cancer cells and immune cells, whereas HLA molecules play critical roles in triggering cytotoxic T lymphocytes (CTL)-mediated tumor cell killing, T cell priming, and clonal expansion. Recent studies have shown that germline HLA gene zygosity, supertype, evolutional divergence, and individual HLA genotypes are associated with the prognosis ofcheckpoint blockade immunotherapy. The purpose of our work was to evaluate biomarker inclusion and response to immunotherapy in pts who underwent germline HLA analysis in Vall d´Hebron Institute of Oncology (VHIO)-Drug Development Unit.

Methods

Pts with solid tumors included in early clinical trials who require germline HLA testing in VHIO's Early Drug Development Unit were included in the analysis from December 2021 to November 2022.

Results

A total of 177 eligible pts. Median age was 59y, main tumor types were colorectal cancer(18%), ovarian cancer(15.8%), breast cancer(12.42%) and pancreatic cancer(12.42%). Most pts(42.93%) were included based on a positive biomarker (21.46% HLA-A01:01, 42.93% HLA-A02:01 and 12.42% HLA-A03:01). Pts treated with anti-PDL1 18 pts(10.17%), anti-PD1 were 14pts(7.90%), anti-PD1 + anti-PDL1 1pts (0.56%), anti-PD1 + other study treatment 19pts(10.73%), anti-PDL1 + other study treatment 4pts(2.26%), other immunotherapy treatment 7pts(3.95%). Association between HLA A02:01 and response to immunotherapy was found significant (p-value 0.0156). The response obtained by CT scan of the pts included in the analysis was progression disease (16pts, 9.03%), stable disease (31pts, 17.51%), partial response (15pts, 8.47%).

Conclusions

High expression of germline HLA-A02:01 genotype is associated with prognosis and response in patients(pts) with solid tumors treated with immunotherapy. HLA-A02:01 genotype potentially be good biomarkers since they influence immune traits in many diseases.

Editorial acknowledgement

Cellex Foundation Institutional grant: research facilities and equipment La Caixa Foundation Institutional grant: LCF/PR/CEO7/50610001

Legal entity responsible for the study

The authors.

Funding

Cellex Foundation Institutional grant: research facilities and equipment La Caixa Foundation Institutional grant: LCF/PR/CEO7/50610001.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

78P - High resolution in situ analysis reveals phenotypic and functional diversification within single pancreatic tumour ducts

Presentation Number
78P
Lecture Time
17:35 - 17:35
Speakers
  • Ellis Michiels (Diepenbeek, Belgium)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Kim Frederix (Diepenbeek, Belgium)
  • Ellis Michiels (Diepenbeek, Belgium)

Abstract

Background

Transcriptomic studies have identified two major subtypes of pancreatic ductal adenocarcinoma (PDAC), i.e. a ‘classical’ and a ‘basal-like’ subtype. These subtypes have differential expression of GATA6, with prognostic and potentially predictive value, and the basal-like subtype has reportedly increased gene dosage of mutant KRAS. Here, we project these findings to the tissue level by high resolution spatial analysis in human PDAC and derived organoids to gain additional biological insights.

Methods

We use formalin-fixed paraffin embedded (FFPE) human PDAC cell lines, tumour organoids and surgical samples that were subjected to hot spot mutation analysis and transcriptomic subtyping by DNA and RNA sequencing. The Basescope assay was optimized for specific in situ detection of KRASpoint mutations in above FFPE samples. BaseScope and RNAScope were combined with multiplex immunostainings and whole tissue sections were analyzed with HALO (Indica™) software.

Results

Apart from inter- and intra-patient heterogeneity, we demonstrate phenotypic diversity of tumour cells within single ducts, revealing spatial phenotypes with varying mRNA expression levels of GATA6 and KRASG12D. Novel gene signature-informed mRNA marker panels underscore the identification of co-existing classical and basal-like zones, as well as co-expressor cells, within single tumour duct. These zones were related to functional (proliferation and epithelial to mesenchymal transition) diversity. PDAC organoids recapitulate the single duct phenotypic diversification that can be shifted experimentally by co-culturing with CAFs.

Conclusions

We successfully establish in situ profiling of transcriptomic subtypes and single point mutations and refine our insights into pancreatic tumour cell plasticity. We reveal extensive intra-tumour diversity that will put extra challenges to novel therapeutic approaches.

Legal entity responsible for the study

VUB and InnoSer Belgie.

Funding

InnoSer Belgie.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

General interest

Lecture Time
17:35 - 17:35
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Cocktail & Poster Display session Abstract session

81P - Implementing genomic profiling as standard-of-care for glioblastoma patients

Presentation Number
81P
Lecture Time
17:35 - 17:35
Speakers
  • Vincent N. Fougner (Copenhagen, Denmark)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Vincent N. Fougner (Copenhagen, Denmark)
  • Anne Dorte Schou Noeroexe (Copenhagen, Denmark)
  • Benedikte Hasselbalch (Copenhagen, Denmark)
  • Thomas Urup (Copenhagen, Denmark)
  • Christina Yde (Copenhagen, Denmark)
  • Laila Belcaid (Copenhagen, Denmark)
  • Martin Hoejgaard (Copenhagen, Denmark)
  • Iben Spanggaard (Copenhagen, Denmark)
  • Kristoffer S. Rohrberg (Copenhagen, Denmark)
  • Hans Skovgaard Poulsen (Copenhagen, Denmark)
  • Ulrik N. Lassen (Copenhagen, Denmark)

Abstract

Background

The introduction of molecular profiling and targeted therapies has only provided few new options for glioblastoma patients (pts). With a median survival of 16 months and no second-line standard therapy, new effective treatments are needed. Here, we report the results from Dept. of Oncology, Rigshospitalet, Denmark, where we investigated the impact of comprehensive molecular profiling in pts with glioblastoma, astrocytoma grade IV and diffuse midline glioma.

Methods

Eligible pts were ≥18 years of age, with newly diagnosed glioblastoma, astrocytoma grade IV or diffuse midline glioma between January 2020 and December 2021. Fresh tumor tissue was used for whole exome sequencing (WES) or whole genome sequencing (WGS) including germline analysis and somatic chromosomal aberrations analysis. Pts enrolled in 2020 had DNA analysed in the WES pipeline, and pts enrolled in 2021 in the WGS pipeline. Each genomic profile was presented at a weekly national molecular tumor board meeting for multidisciplinary evaluation, treatment recommendations and matching with clinical trials. Actionable targets have been classified according to the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT).

Results

133 (92%) of the total 145 pts enrolled had tissue available for WES/WGS. As of now, for the 117 sequenced pts that have progressed after standard therapy, 8 patients (7%) have been treated with molecularly matched experimental therapies. 6 pts were treated in phase 1-2 clinical trials, 1 pt in compassionate use and 1 pt in a Named Patient Program. The actionable targets treated were TMB-high>10mut/MB (n=4), FGFR-mutations/-fusions (n=3) and a NTRK-fusion (n=1).

Conclusions

For glioblastoma, astrocytoma grade IV and diffuse midline glioma, genomic profiling revealed actionable targets and identified new therapeutic options. A full overview of actionable targets and clinical implications will be presented.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

M. Hoejgaard: Financial Interests, Personal, Other, Advisory Role for various diagnostic companies/investors: LingoMedical; Financial Interests, Personal, Stocks/Shares: Bavarian Nordic, Pacific Biosciences, Illumina Inc., Agilent; Financial Interests, Institutional, Funding: Roche; Non-Financial Interests, Personal, Principal Investigator: Repare Therapeutics, Amgen, Incyte Cooperation, Kinnate Biopharma; Other, Personal, Other, Board Member, Tumor Agnostic Board, Public service: Danish Medicines Council. I. Spanggaard: Financial Interests, Institutional, Invited Speaker: Roche, Puma Biotechnology, MSD, Genentech, Incyte, AstraZeneca, Orion, Pfizer; Non-Financial Interests, Personal, Principal Investigator: Roche, Puma Biotechnology, MSD, Genentech, Incyte, AstraZeneca, Orion, Pfizer. K.S. Rohrberg: Financial Interests, Personal, Invited Speaker: Bayer, Amgen, MSD; Financial Interests, Institutional, Invited Speaker, Compensation for conduction of clinical trial: Lilly, Roche/Genentech, Bristol-Myers Squibb, Symphogen, Pfizer, Novartis, Alligator Bioscience, Genmab, Bioinvent, Monta Bioscience; Financial Interests, Institutional, Other, Compensation for conduction of clinical trial: Bayer, Incyte, Puma Biotechnology, Orion Clinical. U.N. Lassen: Financial Interests, Personal, Advisory Board: Bayer, Novartis; Financial Interests, Institutional, Research Grant: Roche, BMS, Pfizer, GSK. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

82P - Cyclic fasting-mimicking diet as a strategy to improve the efficacy of standard antitumor therapies in cancer patients

Presentation Number
82P
Lecture Time
17:35 - 17:35
Speakers
  • Filippo Guglielmo Maria De Braud (Milan, Italy)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Filippo Guglielmo Maria De Braud (Milan, Italy)
  • Giovanni Fucà (Milan, Italy)
  • Francesca Ligorio (Milan, Italy)
  • Veronica Huber (Milan, Italy)
  • Cristina Ferraris (Milan, Italy)
  • Gabriele Martelli (Milan, Italy)
  • Secondo Folli (Milan, Italy)
  • Giulia V. Bianchi (Milan, Italy)
  • Giuseppe Capri (Milan, Italy)
  • Leonardo Provenzano (Milan, Italy)
  • Antonia Martinetti (Milan, Italy)
  • Angela Ficchì (Milan, Italy)
  • Gianfranco Scaperrotta (Milan, Italy)
  • Catherine Depretto (Milan, Italy)
  • Davide Bedognetti (Didra, Qatar)
  • Antonino Belfiore (Milan, Italy)
  • Andrea Vingiani (Milan, Italy)
  • Giancarlo Pruneri (Milan, Italy)
  • Licia Rivoltini (Milan, Italy)
  • Claudio Vernieri (Milan, Italy)

Abstract

Background

Preclinical studies showed that nutrient starvation, in the form of cyclic fasting or fasting-mimicking diet (FMD), sensitizes cancer cells to the antitumor effect of cytotoxic agents and boosts antitumor immunity. These effects are in part mediated by the reduction of blood glucose, insulin and IGF-1 concentration, and by the inhibition of the IGF1-IGF1R axis in tumor cells.

Methods

We conducted a prospective phase Ib clinical trial (NCT03340935) to assess the safety, feasibility and biological effects of a cyclic, 5-day, calorie-restricted, low-carbohydrate, low-protein FMD regimen in a heterogeneous population of cancer patients (pts), and a window-of-opportunity trial (DigesT trial, NCT03454282), in pts with early-stage breast cancer (BC) or melanoma to investigate the immunological effects of a single FMD cycle before surgery.

Results

In 101 pts enrolled in the NCT03340935 trial, and in 22 BC pts included in an interim analysis of the DigesT trial, cyclic FMD was safe, and feasible, and patient compliance was excellent. In addition, the FMD reduced plasma glucose, insulin and IGF-1 concentration and lowered expression/activation of IGF1R in tumor cells. These changes were paralleld by desirable immunologic modifications, including a reduction of circulating immunosuppressive myeloid cells and an increase in activated/cytotoxic T cells and memory T cells at both peripheral blood and tumor levels. Five complete and long-lasting tumor responses were observed in pts with extensive-stage small cell lung cancer (ES-SCLC), advanced pancreatic cancer, metastatic triple-negative BC (TNBC) and metastatic colorectal cancer.

Conclusions

Cyclic FMD may increase the efficacy of standard anticancer therapies. Based on these results we are going to present our next 2 prospective studies: 1) a monocentric, single-arm, phase 2 trial “FASTIMMUNE” to investigate the antitumor efficacy of maintenance atezolizumab plus cyclic FMD in pts with ES-SCLC after 4 cycles of induction chemo-immunotherapy, and 2) the multicentric, open-label, randomized, phase 2 trial “BREAKFAST-2” to investigate if cyclic FMD improves the antitumor activity of neoadjuvant chemoimmunotherapy in patients with stage II/III TNBC.

Clinical trial identification

NCT03340935; NCT03454282.

Legal entity responsible for the study

F.G.M. De Braud.

Funding

Associazione Italiana per la Ricerca sul Cancro (AIRC); European Union Framework Program Horizon 2020.

Disclosure

F.G.M. De Braud: Financial Interests, Personal, Invited Speaker: BMS, Healthcare Research & Pharmacoepidemiology, Merck Group, MSD, Pfizer, Servier, Sanofi, Roche, Amgen, Incyte, Dephaforum, Seagen, Novartis, F.Hoffmann-LaRoche Ltd, BMS, Ignyta Operating INC, Merck Sharp & Dohme Spa, Kymab, Pfizer, Tesaro, MSD, MedImmune LCC, Exelixis Inc, Loxo Oncology Incorporated, Daiichi Sankio Dev. Limited, Basilea Pharmaceutica International AG, Janssen-Cilag International NV, Merck KGAA; Financial Interests, Personal, Other, Consultant Advisory Board: Roche, EMD Serono, NMS Nerviano Medical Science, Sanofi, MSD, Novartis, Incyte, BMS, Menarini, AstraZeneca, Pierre Fabre. G. Pruneri: Financial Interests, Personal, Invited Speaker: Roche, Lilly, Exact Sciences, Novartis; Financial Interests, Personal, Advisory Board: Exact Sciences, ADS Biotec; Financial Interests, Institutional, Research Grant: Roche. L. Rivoltini: Financial Interests, Personal, Invited Speaker, Teaching in educational events: BMS; Non-Financial Interests, Personal, Advisory Role: DKTK. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

83P - Evaluation of response-predictive biomarkers for lisavanbulin: A phase II study in patients with recurrent glioblastoma

Presentation Number
83P
Lecture Time
17:35 - 17:35
Speakers
  • Juanita S. Lopez (Sutton, United Kingdom)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Juanita S. Lopez (Sutton, United Kingdom)
  • Simon Häfliger (Bern, Switzerland)
  • Ruth Plummer (Newcastle upon Tyne, United Kingdom)
  • Paul M. Clement (Leuven, Belgium)
  • Heinz Läubli (Basel, Switzerland)
  • Patrick Roth (Zurich, Switzerland)
  • Thomas Evans (Glasgow, United Kingdom)
  • Lucy Brazil (London, United Kingdom)
  • Ghazaleh Tabatabai (Tübingen, Germany)
  • Antje Wick (Heidelberg, Germany)
  • Yau Wing (Sutton, United Kingdom)
  • Benjamin Wunderlich (Kassel, Germany)
  • Kirk D. Beebe (Research Triangle Park, United States of America)
  • Joel Eisner (Research Triangle Park, United States of America)
  • Marc Engelhardt (Basel, Switzerland)
  • Thomas Kaindl (Basel, Switzerland)
  • Heidi A. Lane (Basel, Switzerland)
  • Peter Hau (Regensburg, Germany)
  • Thomas Hundsberger (St. Gallen, Switzerland)
  • Joachim Steinbach (Frankfurt am Main, Germany)

Abstract

Background

Lisavanbulin (BAL101553, prodrug of BAL27862) destabilizes microtubules, promoting tumor cell death by modulating the spindle assembly checkpoint. BAL27862 is a lipophilic small molecule shown in rodents to penetrate the brain, with antitumor activity in orthotopic glioblastoma (GBM) models. In the phase I part of this study, 2 of 20 patients with recurrent GBM or high-grade glioma showed long-lasting objective responses and strong end-binding protein 1 (EB1) expression in GBM tissue by IHC. EB1, a protein located on the plus-ends of microtubules, is involved in microtubule function. GBM mouse models suggested EB1 is a predictive marker of response to lisavanbulin.

Methods

The objective of the phase 2 study was to investigate prospectively the response-predictive value of EB1, and to identify RNA-based response signatures. A Simon’s Two-Stage design was used with an objective response rate (ORR) ≥ 2/9 required in Stage 1 to enable a final ORR ≥ 6/19. A prescreening program identified patients with EB1-positive archival GBM tissue. Patients with recurrent and measurable disease per RANO receiving treatment for ≥ 6 weeks were evaluable. All patients received 25 mg oral lisavanbulin once daily. RNA-seq was performed on archival GBM tissues.

Results

13 sites in 4 countries participated in this study. Samples from 64 of 629 patients (10.2%) were EB1-positive, and 18 patients received lisavanbulin. Of the 9 patients with measurable disease evaluable for response in Stage 1, there was 1 PR (RANO −58%) and a second patient with a 44% reduction of the target lesion (SD). These patients, and two others with non-measurable disease are ongoing for >10 months. Despite sustained activity in these patients, formal stage transition criteria were not met within the predefined evaluation period, and the study was closed. While IHC testing for EB1 did not show sufficient enrichment for response, RNA-seq analyses identified a 5-gene response signature.

Conclusions

This phase 2a study supports previous study results that lisavanbulin is associated with durable responses and clinical benefit in a subset of patients with GBM. RNA-seq analyses of GBM samples suggest further evaluation of the lisavanbulin predictive response signature.

Clinical trial identification

NCT02490800.

Legal entity responsible for the study

Basilea Pharmaceutica International Ltd, Allschwil.

Funding

Basilea Pharmaceutica International Ltd, Allschwil.

Disclosure

J.S. Lopez: Financial Interests, Personal, Advisory Board: Roche Genentech, Basilea, Ellipses Pharma, Cureteq, Pierre Faber; Financial Interests, Institutional, Research Grant: Roche Genentech, Basilea, Astex. S. Häfliger: Financial Interests, Institutional, Advisory Board, 21.01.2021: Novartis; Financial Interests, Institutional, Invited Speaker: Roche; Financial Interests, Institutional, Advisory Board: Takeda; Financial Interests, Institutional, Expert Testimony: Roche. R. Plummer: Financial Interests, Personal, Advisory Board: Pierre Fabre, Bayer, Novartis, BMS, Cybrexa, Ellipses, CV6 Therapeutics, Astex Therapetics, Sanofi Aventis, Immunocore, Genmab, Medivir, Onexo; Financial Interests, Institutional, Royalties relating to rucaparib licencing: Clovis Oncology; Financial Interests, Personal, Other, Honorarium as Member of IDMC: SOTIO, Alligator Biosciences; Financial Interests, Personal, Other, Honoraria as Member of IDMC: GSK. P.M. Clement: Financial Interests, Institutional, Invited Speaker: Bristol-Myers Squibb, Astra Zeneca, Orbus; Financial Interests, Institutional, Advisory Board: MSD, AbbVie, Bayer, Rakuten, Merck, Vifor, Leo Pharma, Daiichi Sankyo; Financial Interests, Institutional, Research Grant: Astra Zeneca; Financial Interests, Institutional, Invited Speaker: Rakuten; Financial Interests, Personal, Invited Speaker: Basilea; Non-Financial Interests, Personal, Advisory Role: KCE; Non-Financial Interests, Personal, Advisory Role on reimbursement of pharmaceutical specialties in Belgium (Substitute Member): CTG; Non-Financial Interests, Personal, Member: ASCO, EHNS, BANO, SNO, EANO, EORTC, EURACAN, AACR, BSMO; Non-Financial Interests, Personal, Leadership Role: VWHHT; Other, Personal, Other, Advisory Role ad hoc with payment to my institution: EMA. H. Läubli: Financial Interests, Institutional, Expert Testimony: Bristol-Myers Squibb; Financial Interests, Personal, Advisory Board: Palleon Pharmaceuticals, GlycoEra; Financial Interests, Institutional, Invited Speaker: Novartis. P. Roth: Financial Interests, Personal, Expert Testimony: Roche, Merck; Financial Interests, Personal, Advisory Board: BMS, Virometix, QED, Debiopharm; Financial Interests, Personal, Invited Speaker: Novocure; Financial Interests, Institutional, Funding: Novocure; Financial Interests, Institutional, Invited Speaker: MSD. T. Evans: Financial Interests, Institutional, Advisory Board: AstraZeneca, Bayer, Bicycle Therapeutics, Bristol-Myers Squibb, Clovis, Eisai, Medivir, MSD, Nucana, Roche/Genentech. B. Wunderlich: Financial Interests, Personal, Full or part-time Employment: Discovery Life Sciences Biomarker Services GmbH. K.D. Beebe: Financial Interests, Personal, Officer: GeneCentric Therapeutics; Financial Interests, Personal, Stocks/Shares: GeneCentric Therapeutics. J. Eisner: Financial Interests, Personal, Full or part-time Employment, Clinical And Translational Development: GeneCentric Therapeutics. M. Engelhardt, H.A. Lane: Financial Interests, Personal, Full or part-time Employment: Basilea Pharmaceutica International Ltd; Financial Interests, Personal, Stocks/Shares: Basilea Pharmaceutica International Ltd. P. Hau: Financial Interests, Personal, Advisory Board: Bristol-Myers Squibb, GlaxoSmithKline, Merck, Sharp & Dome, Novocure; Financial Interests, Personal, Invited Speaker: Lilly, Medac. T. Hundsberger: Financial Interests, Personal, Advisory Board: Amicus, Sanofi Genzyme, Bayer AG, Novocure. J. Steinbach: Financial Interests, Personal, Advisory Board: Roche, Boehringer Ingelheim, Seagen, Novocure. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

84P - Racial disparities in phase I lung cancer clinical trials

Presentation Number
84P
Lecture Time
17:35 - 17:35
Speakers
  • Rohit Singh (Burlington, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Rohit Singh (Burlington, United States of America)
  • Akshee Batra (Burlington, United States of America)
  • David Pottinger (Birmingham, United States of America)
  • Numair Rizwan (Prague, Czech Republic)
  • Kayla Goliwas (Birmingham, United States of America)
  • Soumya Niranjan (Birmingham, United States of America)
  • Devika G. Das (Birmingham, United States of America)
  • Bassel El-Rayes (Birmingham, United States of America)
  • Aakash Desai (Jacksonville, United States of America)

Abstract

Background

Lung cancer is the leading cause of cancer-related deaths worldwide and disproportionately affects racial and ethnic minorities. We aimed to assess the enrollment of minorities in phase I clinical trials for lung cancer.

Methods

We searched the clinicaltrials.gov database for completed phase I clinical trials conducted in adult patients from 2010 to 2022. To ensure the relevance and generalizability of our findings, we included trials conducted in North America, Europe, and Australia, while excluding those restricted to Southeast Asia or focused on radiation or screening interventions. We manually abstracted data on the racial distribution of enrollees, tumor histology, therapeutics, and year of trial reporting. To quantify the enrollment of each racial group, we calculated the enrollment fraction for each subgroup as the number of trial enrollees within that subgroup divided by the estimated number of incident cases in that subgroup during a given period. We used the Z test for proportions to compare enrollment fractions between racial groups, with Whites as the comparator group.

Results

We identified 193 phase I clinical trials, and 132 met our inclusion criteria for analysis. Racial data was available in 89 studies (67.5%) that included 11,359 participants: 9439 White (83%), 508 African American (4.5%), 898 Asian (8%), and 513 other participants (4.5%). The frequency of race reporting increased over time, with racial data included in 37% of studies in 2010-2014, 51% in 2015-2019, and 93% in 2020-2022. Comparison of enrollment fractions between 2015-2019 showed that African Americans were significantly underrepresented compared to Whites (z=5.2, p<0.00001). Table: 84P

White African American Asians Other
Proportion of Trial Enrollment, % 87.8 4.4 5.5 2.3
Enrollment fraction, % 0.34 0.14 0.62 0.65

Conclusions

Our study highlights the continued underrepresentation of minorities in phase I clinical trials for lung cancer. To ensure equity in trial participation and the generalizability of developmental therapeutic trials in lung cancer, it is necessary to implement multilevel, culturally, and linguistically tailored strategies.

Editorial acknowledgement

No assistance

Legal entity responsible for the study

R. Singh.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

85P - Stereotactic radiotherapy improves disease control in oligoprogressive patients included in early clinical trials

Presentation Number
85P
Lecture Time
17:35 - 17:35
Speakers
  • Antoine Mavrikios (Villejuif, Cedex, France)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Antoine Mavrikios (Villejuif, Cedex, France)
  • Capucine Baldini (Villejuif, Cedex, France)
  • Yohann Loriot (Villejuif, Cedex, France)
  • Santiago Ponce Aix (Madrid, Spain)
  • Stephane Champiat (Villejuif, Cedex, France)
  • François-Xavier Danlos (Villejuif, France)
  • Clément Quevrin (Villejuif, Cedex, France)
  • Anas Gazzah (Villejuif, Cedex, France)
  • Rastislav Bahleda (Villejuif, France)
  • Eric Deutsch (Villejuif, France)
  • Antonin Levy (Villejuif, France)

Abstract

Background

Patients included in phase I trials are often heavily pre-treated and display strong treatment expectations. In some cases, oligoprogressive disease may occur but with pursued clinical benefit. In such scenarios, local ablative stereotactic radiotherapy (SRT) could allow disease control with prolonged use of current systemic treatment.

Methods

We retrospectively analysed data from patients included in early clinical trials who received SRT for oligo-acquired resistance (≤ 3 lesions of disease progression; OAR) between 01/2014 and 12/2021. OS, PFS1 (trial entry to OAR), PFS2 (SRT to subsequent relapse), time to next treatment (TTNT) were assessed. Subsequent patterns of relapse were distinguished as OAR2 or systemic AR (SAR).

Results

39 patients with 48 oligoprogressive lesions were included. Most frequent primary tumor histologies were NSCLC (33%), melanoma and urothelial carcinoma (13% each). Median age was 59 years, median baseline RMH score was 1 and 93% patients had an ECOG-PS<1. Early clinical trials mostly included ICI (64%) and molecular targeted therapies (MTT) (46%). SRT was mainly delivered to brain (38%) and lymph nodes (26%) at a median dose of 30 Gy. Median follow-up was 19 months. Median OS, PFS1, PFS2, and TTNT were respectively 16, 11, 7 and 9 months. PFS2 included 44% OAR2 and 56% SAR. No SRT-related grade 3-5 toxicity was observed. Increased OS was associated with primary tumor local control, higher baseline lymphocytes count, lower baseline RMH score, lower post-SRT tumor burden and absence of SAR. Increased TTNT was associated with primary tumor local control, absence of baseline polymetastatic disease, lower baseline RMH score, OAR2, OAR2-local treatment, and both lower baseline/post-SRT tumor burden. OAR2 was more often observed in MTT trials and SAR was associated with absence of primary tumor local control, short PFS1 and higher baseline tumor burden.

Conclusions

In pre-treated patients included in phase I trials, OAR managed with SRT led to durable benefit and prolonged continuation of investigational treatments. Predictive factors could be used for patient selection by distinguishing subsequent OAR2 from SAR.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

86P - Validation of the Gustave Roussy Immune (GRIm) score in patients treated with bispecific CD3 T cell engagers in phase I clinical trials

Presentation Number
86P
Lecture Time
17:35 - 17:35
Speakers
  • Noé Herbel (Villejuif, Cedex, France)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Noé Herbel (Villejuif, Cedex, France)
  • Vincent Goldschmidt (Villejuif, Cedex, France)
  • Jean-Marie Michot (Villejuif, France)
  • Ariane Laparra (Villejuif, Cedex, France)
  • Arthur Géraud (Villejuif, France)
  • Kaissa Ouali (Villejuif, France)
  • François-Xavier Danlos (Villejuif, France)
  • Patricia Martin Romano (Villejuif, Cedex, France)
  • Perrine Vuagnat (Villejuif, Cedex, France)
  • Alice Bernard-Tessier (Villejuif, Cedex, France)
  • Anas Gazzah (Villejuif, Cedex, France)
  • Rastislav Bahleda (Villejuif, France)
  • Antoine Hollebecque (Villejuif, Cedex, France)
  • Aurélien Marabelle (Villejuif, France)
  • Sophie Postel-Vinay (Villejuif, France)
  • Christophe Massard (Villejuif, France)
  • Vincent Ribrag (Villejuif, Cedex, France)
  • Santiago Ponce Aix (Madrid, Spain)
  • Stephane Champiat (Villejuif, Cedex, France)
  • Capucine Baldini (Villejuif, Cedex, France)

Abstract

Background

The Gustave Roussy Immune (GRIm)-Score considers neutrophil-to-lymphocyte ratio (NLR), serum albumin concentration and lactate dehydrogenase (LDH) and its prognostic value has been investigated in patients treated with immune check-point blockers (ICBs). According to their GRIm-score, patients can be classified as low risk (GRIm-score 0 or 1) or as high risk (GRIM-score 2 or 3). We aimed to determine whether it could be used as a prognostic tool for patients treated in phase I clinical trials with bispecific CD3 T cell engagers.

Methods

A retrospective analysis on overall survival (OS) and progression free survival (PFS) was performed on all patients treated with bispecific CD3 T cell engagers in phase I clinical trials at DITEP between July 2018 and July 2022, alone or in combination with ICB. Estimations of OS and PFS were made using the Kaplan Meier method and comparisons between low and high-risk patients were done using log-rank test. The power of the GRIm-score was analyzed using Harrell’s C-index to determine its predictive ability.

Results

A total of 67 patients with metastatic solid cancer or hematological malignancy was included. Median age was 62 years old (range 42-78), 47 patients were men (70%) and 20 were women (30%). The most common tumor types were prostate cancer (33%), lymphoma (25%) and small cell lung carcinoma (24%). The median number of previous lines of treatment was 4 (range : 1-9). Most of the patients presented a low GRIm-score (80.5%) at baseline : 0 in 46% of the patients and 1 in 34,5% . A high GRIm-score was found in 19.5% of the patients: 2 in 18% and 3 in 1.5%. A low GRIm-score was associated with better OS and PFS (HR: 0.40; 95% CI: 0.19-0.84, p = 0.01 and HR: 0.36; 95% CI: 0.18-0.7, p = 0.002 respectively) in patients treated with bispecific CD3 T cell engagers. The median OS was 17.3 months for low-risk patients and 4.5 months for high risk. The median PFS was 2.8 months for low risk GRIm and 1.2 month for high risk. The Harrel’s C index for OS was 0.60 and 0.58 for PFS.

Conclusions

The GRIm-score was designed for patients treated with ICB. It can be used to select patients for bispecific CD3 T cell engagers phase I clinical trials. It has prognostic impact on PFS and OS.

Legal entity responsible for the study

Gustave Roussy.

Funding

Has not received any funding.

Disclosure

V. Goldschmidt: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, AstraZeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies, BeiGene, BicycleTx, Blueprint Medicines, Boehringer Ingelheim, Boston Pharmaceuticals, Bristol Myers Squibb, Celgene Corporation, Chugai Pharmaceutical, Clovis Oncology, Cullinan-Apollo, Curevac, Daiichi Sankyo, Debiopharm, Eisai, Eli Lilly, Exelixis, Faron Pharmaceuticals, Forma Tharapeutics, Gamamabs, Genentech, Glaxosmithkline, H3 Biomedicine, Hoffmann La Roche, Imcheck Therapeutics, Innate Pharma, Institut De Recherche Pierre Fabre, Iris Servier, Iteos Belgium, Janssen Cilag, Janssen Research Foundation, Kura Oncology, Kyowa Kirin Pharm, Loxo Oncology, Lytix Biopharma, MedImmune, Menarini Ricerche, Merck Sharp & Dohme Chibret, Merrimack Pharmaceuticals, Merus, Millennium Pharmaceuticals, Molecular Partners, Nanobiotix, Nektar Therapeutics, Novartis Pharma, Octimet Oncology, Oncoethix, Oncopeptides, Orion Pharma, Ose Pharma, Pfizer, PharmaMar, Pierre Fabre, Medicament, Roche, Sanofi Aventis, Seattle Genetics, Sotio, Syros Pharmaceuticals, Taiho Pharma, Tesaro, Turning Point Therapeutics, Xencor; Financial Interests, Institutional, Proprietary Information: Lilly France; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. J. Michot: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, Astra Zeneca, Aveo, Basilea Pharmaceutica Internationa, Bayer Healthcar, Bbb Technologies, BeiGene, BicycleTx, Blueprint Medicines, Boehringer Ingelheim, Boston Pharmaceuticals, Bristol Myers Squibb, Ca, Celgene Corporation, Chugai Pharmaceutical, Clovis Oncology, Cullinan-Apollo, Curevac, Daiichi Sankyo, Debiopharm, Eisai, Eli Lilly, Exelixis, Faron Pharmaceuticals, Forma Tharapeutics, Gamamabs, Genentech, Glaxosmithkline, H3 Biomedicine, Hoffmann La Roche, Imcheck Therapeutics, Innate Pharma, Institut De Recherche Pierre Fabre, Iris Servier, Iteos Belgium, Janssen Cilag, Janssen Research Foundation, Kura Oncology, Kyowa Kirin Pharm. Dev, Lilly France, Loxo Oncology, Lytix Biopharma As, MedImmune, Menarini Ricerche, Merck Sharp & Dohme Chibret, Merrimack Pharmaceuticals, Merus, Millennium Pharmaceuticals, Molecular Partners, Nanobiotix, Nektar Therapeutics, Novartis Pharma, Octimet Oncology Nv, Oncoethix, Oncopeptides, Orion Pharma, Ose Pharma, Pfizer, PharmaMar, Pierre Fabre, Medicament, Roche, Sanofi Aventis, Seattle Genetics, Sotio, Syros Pharmaceuticals, Taiho Pharma, Tesaro, Turning Point Therapeutics, Xencor; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. K. Ouali: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, AstraZeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies Bv, BeiGene, BicycleTx; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, R. F. Danlos: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, AstraZeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies Bv, BeiGene, BicycleTx; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. P. Martin Romano: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, Astra Zeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies Bv, BeiGene, BicycleTx; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. P. Vuagnat: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, AstraZeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies Bv, BeiGene, BicycleTx; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. A. Bernard-Tessier: Financial Interests, Personal, Invited Speaker: Astellas; Financial Interests, Personal, Other, Travel fees: Orion, Bayer; Financial Interests, Institutional, Advisory Board: Novartis; Non-Financial Interests, Personal, Principal Investigator: Amgen. A. Gazzah: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, AstraZeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies Bv, BeiGene, BicycleTx; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. R. Bahleda: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, AstraZeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies Bv, BeiGene, BicycleTx; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. A. Hollebecque: Financial Interests, Personal, Invited Speaker: Servier, Incyte, Eisai; Financial Interests, Personal, Advisory Board: Basilea, Tahio, Relay Theraeutics, QED Therapeutics, Debiopharm; Financial Interests, Institutional, Funding: Incyte; Financial Interests, Institutional, Research Grant: AstraZeneca; Non-Financial Interests, Personal, Principal Investigator, M19-345: AbbVie; Non-Financial Interests, Personal, Principal Investigator, CO42216: Roche; Non-Financial Interests, Personal, Principal Investigator, MCLA-158: Merus; Non-Financial Interests, Personal, Principal Investigator, SGNB6A: Seatle Genetics; Non-Financial Interests, Personal, Principal Investigator, TAS-120-202: Tahio; Non-Financial Interests, Personal, Principal Investigator, Krystal-10: Mirati; Non-Financial Interests, Personal, Principal Investigator, ADP-0033: Adaptimmune; Non-Financial Interests, Personal, Principal Investigator, ACT16902: Sanofi; Non-Financial Interests, Personal, Principal Investigator, C4201002: Pfizer; Non-Financial Interests, Personal, Principal Investigator, RLY-4008: Relay Therapeutics; Non-Financial Interests, Personal, Principal Investigator, CC-90011: Celgene/BMS; Non-Financial Interests, Personal, Principal Investigator, Loxo-IDH: Loxo/Lilly; Non-Financial Interests, Personal, Principal Investigator: AstraZeneca. A. Marabelle: Financial Interests, Personal, Advisory Board: Gritstone, Innate Pharma, Neogene, Deka Biosciences, Hotspot Therapeutics, Johnson & Johnson, Depth Charge, Bioline Rx, Clover Biopharmaceuticals, Grey Wolf Therapeutics, Lytix Biopharma, RedX Pharma, HiFiBiO Therapeutics, ImCheck Therapeutics, Shattuck Labs, Marengo Therapeutics, PegaOne; Financial Interests, Personal, Other, Associate Editor: European Journal of Cancer; Financial Interests, Personal, Stocks/Shares: HiFiBiO Therapeutics, Deka Biosciences, HotSpot Therapeutics, Shattuck Labs; Financial Interests, Institutional, Research Grant: BMS, Astra Zeneca, Sanofi; Financial Interests, Institutional, Invited Speaker: IMCheck, SOTIO, Roche/Genentech, MSD, BMS, OSE Immunotherapeutics, Eisai, Pierre Fabre, Adlai Nortye, Molecular Partners; Financial Interests, Personal, Invited Speaker: Roche; Financial Interests, Institutional, Funding: Transgene; Non-Financial Interests, Personal, Other, Associate Editor: IOTECH Journal; Non-Financial Interests, Institutional, Product Samples: BMS, IDERA, MSD, Transgene; Non-Financial Interests, Personal, Leadership Role: Société Française d'Immunothérapie des Cancers; Non-Financial Interests, Personal, Member: Society for Immunotherapy of Cancer, American Association for Cancer Research, American Society for Clinical Oncology; Other, Personal, Other, Consultant: Third Rock Ventures, Guidepoint; Other, Personal, Other, Advisor: Medicxi. S. Postel-Vinay: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, AstraZeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies Bv, BeiGene, BicycleTx, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. C. Massard: Other, Personal, Other, Christophe Massard: Consultant/Advisory fees from Amgen, Astellas, AstraZeneca, Bayer, BeiGene, BMS, Celgene, Debiopharm, Genentech, Ipsen, Janssen, Lilly, MedImmune, MSD, Novartis, Pfizer, Roche, Sanofi, Orion Principal/sub-Investigator of Clinical Trials for AbbVie, Aduro, Agios, Amgen, Argen-x, Astex, AstraZeneca, Aveo Pharmaceuticals, Bayer, BeiGene, Blueprint, BMS, Boeringer Ingelheim, Celgene, Chugai, Clovis, Daiichi Sankyo, Debiopharm, Eisai, Eos, Exelixis, Forma, Gamamabs, Genentech, Gortec, GSK, H3 Biomedecine, Incyte, Innate Pharma, Janssen, Kura Oncology, Kyowa, Lilly, Loxo, Lysarc, Lytix Biopharma, MedImmune, Menarini, Merus, MSD, Nanobiotix, Nektar Therapeutics, Novartis, Octimet, Oncoethix, Oncopeptides, Orion, Pfizer, PharmaMar, Pierre Fabre, Roche, Sanofi, Servier, Sierra Oncology, Taiho, Takeda, Tesaro, Xencor Company. V. Ribrag: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, AstraZeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies Bv, BeiGene, BicycleTx; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. S. Ponce Aix: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, AstraZeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies Bv, BeiGene, BicycleTx; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. S. Champiat: Financial Interests, Personal, Invited Speaker: Amgen, Astellas, AstraZeneca, Bristol Myers Squibb, Eisai, Genmab, Janssen, Merck, Merck Serono, Novartis and Roche; Financial Interests, Personal, Other, Principal Investigator of Clinical Trials: AbbVie, Amgen, Boehringer Ingelheim, Cytovation, Eisai, Imcheck Therapeutics, Molecular Partners, Merck, Ose Pharma, Pierre Fabre, Sanofi Aventis, Sotio, Transgene; Financial Interests, Personal, Advisory Board: Alderaan Biotechnology, Amgen, AstraZeneca, Avacta, Celanese, Domain Therapeutics, Ellipses Pharma, Genmab, Immunicom, Nanobiotix, Nextcure, Oncovita, Pierre Fabre, Seagen, Tatum Bioscience, Tollys SAS, UltraHuman8; Financial Interests, Personal, Other, Travel and congress: Amgen, AstraZeneca, Bristol Myers Squibb, Merck, Ose Pharma, Roche, Sotio; Financial Interests, Institutional, Other, As part of the Drug Development Department (DITEP) =Principal/sub-Investigator of Clinical Trials: Adaptimmune, Adlai Nortye USA Inc, Aduro Biotech, Agios Pharmaceuticals, Amgen, Adaptimmune, AdlaiAstex Pharmaceuticals, AstraZeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies Bv, BeiGene, BicycleTx, Blueprint Medicines, Boehringer Ingelheim, Boston Pharmaceuticals, Bristol Myers Squibb, Ca, Casi Pharmaceuticals, Celgene Corporation, Cellcentric, Chugai Pharmaceutical, Cullinan-Apollo, Curevac, Daiichi Sankyo, Debiopharm, Eisai, Eli Lilly, Exelixis, Faron Pharmaceuticals Ltd, Forma Tharapeutics, Gamamabs, Genentech, Glaxosmithkline, H3 Biomedicine, Hoffmann La Roche, Imcheck Therapeutics, Incyte Corporation, Imcheck Therapeutics, Incyte Corporation, Innate Pharma, Institut De Recherche Pierre Fabre, Iris Servier, Iteos Belgium, Janssen Cilag, Janssen Research Foundation, Janssen R&D, Kura Oncology, Kyowa Kirin Pharm. Dev, Lilly France, Loxo Oncology, MedImmune, Menarini Ricerche, Merck Sharp & Dohme Chibret, Merrimack Pharmaceuticals, Merus, Molecular Partners, Nanobiotix, Nektar Therapeutics, Novartis Pharma, Octimet Oncology Nv, Oncoethix, Oncopeptides, Orion Pharma, Genomics, Ose Pharma, Pfizer, Pharma Mar, Pierre Fabre Medicament, Relay Therapeutics, Roche, Sanofi Aventis, Seattle Genetics, Sotio, Syros Pharmaceuticals, Taiho Pharma, Tesaro, Transgene, Turning Point Therapeutics, Xencor; Financial Interests, Institutional, Research Grant, As part of the Drug Development Department (DITEP) =Research Grants: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Product Samples, As part of the Drug Development Department (DITEP) =Non-financial support (drug supplied): AstraZeneca, BMS, Boehringer Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. C. Baldini: Financial Interests, Institutional, Principal Investigator: AbbVie, Adaptimmune, Adlai Nortye USA, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, AstraZeneca, Aveo, Basilea Pharmaceutica International, Bayer Healthcare, Bbb Technologies Bv, BeiGene, BicycleTx, Blueprint Medicines, Boehringer Ingelheim, Boston Pharmaceuticals, Bristol Myers Squibb, Ca, Celgene Corporation, Chugai Pharmaceutical Co, Clovis Oncology, Cullinan-Apollo, Curevac, Daiichi Sankyo, Debiopharm, Eisai, Eisai Limited, Eli Lilly, Exelixis, Faron Pharmaceuticals, Forma Tharapeutics, Gamamabs, Genentech, Glaxosmithkline, H3 Biomedicine, Hoffmann La Roche, Imcheck Therapeutics, Innate Pharma, Institut De Recherche Pierre Fabre, Iris Servier, Iteos Belgium, Janssen Cilag, Janssen Research Foundation, Kura Oncology, Kyowa Kirin Pharm. Dev, Lilly France, Loxo Oncology, Lytix Biopharma As, MedImmune, Menarini Ricerche, Merck Sharp & Dohme Chibret, Merrimack Pharmaceuticals, Merus, Millennium Pharmaceuticals, Molecular Partners, Nanobiotix, Nektar Therapeutics, Novartis Pharma, Octimet Oncology Nv, Oncoethix, Oncopeptides, Orion Pharma, Ose Pharma, Pfizer, Pharma Mar, Pierre Fabre, Medicament, Roche, Sanofi Aventis, Seattle Genetics, Sotio, Syros Pharmaceuticals, Taiho Pharma, Tesaro, Turning Point Therapeutics, Xencor; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Other, Drug supplied: AstraZeneca, Bayer, BMS, Boringher Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

87P - Clinical features and genetic profile of MDM2-altered solid tumors

Presentation Number
87P
Lecture Time
17:35 - 17:35
Speakers
  • Julia Tejerina-Peces (Madrid, Spain)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Julia Tejerina-Peces (Madrid, Spain)
  • Jorge Bartolomé Arcilla (Madrid, Spain)
  • Miguel De La Hoya (Madrid, Spain)
  • Pilar Garre (Madrid, Spain)
  • Mateo Paz Cabezas (Madrid, Spain)
  • Alfonso López de Sá (Madrid, Spain)
  • Santiago Cabezas-Camarero (Madrid, Spain)
  • Beatriz García Paredes (Madrid, Spain)
  • Natalia Vidal Cassinello (Madrid, Spain)
  • Carlos Aguado (Madrid, Spain)
  • Gloria Marquina Ospina (Madrid, Spain)
  • Gonzalo Fernandez-Hinojal (Madrid, Spain)
  • Alberto Orta (Madrid, Spain)
  • Luis Ortega (Madrid, Spain)
  • Alberto Ocana Fernandez (Madrid, Spain)
  • Pedro Pérez Segura (Madrid, Spain)
  • Aranzazu Manzano Fernández (Madrid, Spain)

Abstract

Background

MDM2 is a negative regulator of p53 that directly inhibits the transcriptional activation of p53 targeting its ubiquitination and degradation by the proteasome. In addition, MDM2 has a p53-independent effect by modulating other molecular pathways involved in cell growth, proliferation and death. Information about the accompanying genetic co-alterations in MDM2-altered tumors, including its relationship with the immune system and other oncogenic pathways, is scarce. New agents targeting MDM2 are currently under development. A better knowledge of the clinical and molecular features of MDM2-altered tumors is needed to establish the treatment for these tumors.

Methods

We performed an observational retrospective study at Hospital Clínico San Carlos (Madrid, Spain) in 816 advanced solid tumor patients undergoing Next Generation Sequencing (NGS) on tumor samples (n=807) or liquid biopsies (n=9) between July 2019 and December 2022. Clinical data, tumor mutational burden (TMB) and accompanying genetic alterations of the MDM2-altered tumors were recorded (n= 33, 4%).

Results

Of the 33 MDM2-altered tumors, we found 26 (79%) amplifications (2 of them with co-existing MDM2 rearrangements) and 7 (21%) non-previously described frameshift mutations. The most frequent tumors were liposarcoma (n=8, 24%), lung cancer (n=7; 21%) and head and neck cancer (n=4; 12%). Most common accompanying genetic alterations were related to cell cycle (TP53 and/or CDK4, 88%), DNA repair pathway (BRCA2, ATRX, ATM, RAD54L and/or POLE, 52%) and chromatin modulation pathway (BRD4, MLL2 and/or SMARCA4, 30%). There were 7 TMB-high (cut-off > 10 mpm) tumors (3.78-174) in 2 melanoma, 2 lung cancer, 1 breast cancer, 1 parotid cancer and 1 cancer of unknown primary. 5 of them related to mutations in MDM2. There was only one microsatellite instability (MSI) in a patient with TMB-high breast tumor.

Conclusions

In our cohort, solid tumors with MDM2 alterations had accompanying genetic alterations affecting chromatin modulation and DNA repair pathway. MDM2 mutated solid tumors presented a trend to high TMB. These findings could help to develop new combination strategies for MDM2-altered tumors with immunotherapy or chromatin or DNA repair targeted agents.

Legal entity responsible for the study

J. Tejerina-Peces, Hospital Clínico San Carlos.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

88P - Evaluation of trending drug targets and technologies in current drug development

Presentation Number
88P
Lecture Time
17:35 - 17:35
Speakers
  • Matteo Repetto (Milan, Italy)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Matteo Repetto (Milan, Italy)
  • Edoardo Crimini (Milan, Italy)
  • Carmen Belli (Milan, Italy)
  • Luca Boscolo Bielo (Milan, Italy)
  • Liliana Ascione (Milan, Italy)
  • Alexander Drilon (New York, United States of America)
  • Giuseppe Curigliano (Milan, Italy)

Abstract

Background

Novel investigational agents (IAs) aim to exploit new mechanisms and cancer vulnerabilities, expanding our arsenal of anticancer drugs. Recently, highly selective biomarker-directed IAs have allowed advancements in the precision treatment of oncogene-driven cancers by decreasing toxicity and increasing activity. In phase I drug development, a global problem of unknown magnitude is the multiplicity of similar drugs being investigated against the same target, colloquially known as the ‘me too’ phenomenon.

Methods

The Clinicaltrials.gov online database was queried for phase I clinical trials active during a two-year timeframe from December 2020 to December 2022. Natural language processing was employed to extract IAs from experimental interventions. Autoritative sources (Cancer.gov, Pubmed, et similia) were queried to classify IAs based on target and drug class/structure. Whenever data was unavailable from these websites, lower tier sources were employed. Overall target frequency and co-occurrence analyses were subsequently evaluated.

Results

By surveying the current landscape of phase I clinical trials for a two-year timeframe (December 2020 to December 2022), we identified 4480 distinct IAs. Excluding agents without direct mechanistic targets, non-human genome encoded targets (such as viral proteins, metabolites, carbohydrates et similia) or undisclosed data, 1016 gene targets and 3495 investigational agents were identified. While only a minority of gene targets had 4 or more IAs directed against them (26.7%) the majority of IA, 83.3% of single-target and 92.5% of multi-targeted, were directed against this minority of gene targets. The most frequent IA classes were Inhibitors(1105), Cell products(933), Monospecific-monoclonal antibodies(488) and Vaccines(252). The most frequent gene targets by number of IA were: CD19(316), EGFR(131), CD3(126), ERBB2(125), BCMA(108) and PD-1(106), CTLA4-PD1 and PDL1-CTLA4 were the most frequent cooccurring target pairs.

Conclusions

Only a minority (9.3%) of phase I IAs were explored against a target without a competitive agent; Unfortunately, the majority of IAs (86%) shared targets with at least 3 other agents. We argue that these duplicative efforts could be redirected toward unmet needs instead.

Editorial acknowledgement

This abstract did not receive any editorial assistance.

Legal entity responsible for the study

M. Repetto.

Funding

Has not received any funding.

Disclosure

M. Repetto: Financial Interests, Personal, Funding: Sanofi. A. Drilon: Financial Interests, Personal, Advisory Board: Ignyta/Genentech/Roche, Loxo/Bayer/Lilly, Takeda/Ariad/Millennium, TP Therapeutics, AstraZeneca, Pfizer, Blueprint Medicines, Helsinn, BeiGene, BerGenBio, Hengrui Therapeutics, Exelixis, Tyra Biosciences, Verastem Oncology, MORE Health, AbbVie, 14ner/Elevation Oncology, Remedica, ArcherDX, Monopteros, Novartis, EMD Serono, Melendi, Liberum, Repare RX, Amgen, Janssen, EcoR1, Monte Rosa; Financial Interests, Personal, Other, CME: Medscape, Onclive, PeerVoice, Physicians Education Resources, Targeted Oncology, Research to Practice, PeerView Institute, Paradigm Medical Communications, WebMD, MJH Life Sciences, Med Learning, Imedex, Answers in CME, Medscape, Clinical Care Options, AiCME; Financial Interests, Personal, Other, CME, Consulting: Axis; Financial Interests, Personal, Other, Consulting: Nuvalent, Merus, EPG Health, mBrace, Harborside Nexus, Ology, TouchIME, Entos, Treeline Bio, Prelude, Applied Pharmaceutical Science, Inc; Financial Interests, Personal, Invited Speaker: Chugai Pharmaceutical, Remedica Ltd, RV More; Financial Interests, Personal, Stocks/Shares: Treeline Biosciences; Financial Interests, Personal, Royalties: Wolters Kluwer; Financial Interests, Personal, Other, stocks: mBrace; Financial Interests, Institutional, Funding, Research funding: Pfizer, Exelixis, GlaxoSmithKline, Teva, Taiho, PharmaMar; Financial Interests, Personal, Funding, Research: Foundation Medicine; Non-Financial Interests, Personal, Member: ASCO, AACR, IASLC; Other, Personal, Other, Food/Beverage: Merck, PUMA, Merus; Other, Personal, Other, Other: Boehringer Ingelheim. G. Curigliano: Financial Interests, Personal, Invited Speaker: Roche, AstraZeneca, Daiichi Sankyo, Novartis, Pfizer, Pfizer; Financial Interests, Personal, Advisory Board: Ellipsis, Roche, AstraZeneca, Daiichi Sankyo, Lilly, Pfizer, Veracyte, BMS, Merck, Exact Sciences, Celcuity; Financial Interests, Institutional, Research Grant, Investigator Initiated Trial: Merck; Financial Interests, Institutional, Funding, Phase I studies: BMS, Novartis, AstraZeneca, Daiichi Sankyo, Roche, Blueprint Medicine, Kymab, Astellas, Sanofi, Philogen; Financial Interests, Institutional, Invited Speaker, Phase I clinical basket trial: Relay Therapeutics; Non-Financial Interests, Personal, Officer, Italian National Health Council as Advisor for Ministry of Health: Consiglio Superiore di Sanità; Non-Financial Interests, Personal, Advisory Role, Member of the Scientific Council. Patient Advocacy Association: Europa Donna; Non-Financial Interests, Personal, Advisory Role, Cancer Research Foundation: Fondazione Beretta; Non-Financial Interests, Personal, Invited Speaker, No compensation for this role, This a public national company for cancer prevention: Lega Italiana Lotta ai Tumori; Non-Financial Interests, Personal, Officer, Member of the Advisory Council: EUSOMA. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

89P - Impact of metformin on glucocorticoid-induced changes in systemic metabolism in patients with brain metastases from solid malignancies

Presentation Number
89P
Lecture Time
17:35 - 17:35
Speakers
  • Filippo Guglielmo Maria De Braud (Milan, Italy)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Filippo Guglielmo Maria De Braud (Milan, Italy)
  • Riccardo Lobefaro (Milan, Italy)
  • Paola Corsetto (Milan, Italy)
  • Francesca Ligorio (Milan, Italy)
  • Emma Zattarin (Milan, Italy)
  • Michele Del Vecchio (Milan, Italy)
  • Lorenza A. Di Guardo (Milan, Italy)
  • Giuseppe Lo Russo (Milan, Italy)
  • Claudia Proto (Milan, Italy)
  • Sara Cresta (Milan, Italy)
  • Cristina Ferraris (Milan, Italy)
  • Gabriele Martelli (Milan, Italy)
  • Secondo Folli (Milan, Italy)
  • Veronica Huber (Milan, Italy)
  • Leonardo Provenzano (Milan, Italy)
  • Antonia Martinetti (Milan, Italy)
  • Angela Ficchì (Milan, Italy)
  • Licia Rivoltini (Milan, Italy)
  • Giovanni Fucà (Milan, Italy)
  • Claudio Vernieri (Milan, Italy)

Abstract

Background

Glucocorticoids (GC) are commonly used in patients (pts) with brain metastases to palliate symptoms and to prevent radiotherapy-induced side effects. GCs can cause metabolic adverse events (AEs), such as hyperglycemia or diabetes, which could negatively impact on clinical outcomes. Here, we evaluated if the antidiabetic compound metformin (MET) prevents GC-induced hyperglycemia and diabetes in pts with brain metastases from melanoma, breast or lung cancer.

Methods

We conducted a monocentric, prospective, phase II clinical trial (OPTIMAL: NCT02705157) that randomized pts with a de novo diagnosis of brain metastases to receive dexamethasone (DEX) at a minimum daily dosage of 8 mg, or DEX plus oral MET, up to a maximum daily dosage of 2550 mg. The primary study objective was to investigate if MET reduces the incidence of diabetes at 14 days after DEX initiation. Secondary study endpoints were changes in other systemic metabolic parameters, as well as in peripheral blood cell counts. Here we present an interim analysis of the study, which included 33 pts.

Results

Of 33 pts randomized between October 2019 and July 2021, 15 pts received DEX, while 18 pts received DEX plus MET. None of pts enrolled in the study developed treatment-induced diabetes at 14 days; in addition, DEX, with or without MET, did not result in increased fasting plasma glucose concentration. Based on these findings, the OPTIMAL trial was precociously interrupted. Of note, MET use resulted in statistically significant reduction of post-prandial glycemia, as measured as capillary blood glucose levels, and in an attenuation of DEX-induced increased in fasting plasma HDL cholesterol. Plasma triglyceride, total cholesterol and LDL cholesterol levels are not affected by GCs, either alone or in combination with MET. Finally, MET did not reverse DEX-induced increase in blood leukocyte, neutrophil and monocyte counts.

Conclusions

High-dose GCs, +/- MET, do not precociously affect fasting blood glucose concentration in pts with brain metastases from solid malignancies. MET could potentially reverse some GC-induced metabolic changes. However, the impact of these metabolic modifications on systemic immunity and on clinical outcomes remains unclear.

Clinical trial identification

NCT02705157.

Legal entity responsible for the study

F.G.M. De Braud.

Funding

Has not received any funding.

Disclosure

F.G.M. De Braud: Financial Interests, Personal, Invited Speaker: BMS, Healthcare Research & Pharmacoepidemiology, Merck Group, MSD, Pfizer, Servier, Sanofi, Roche, Amgen, Incyte, Dephaforum, Seagen, Novartis, F.Hoffmann-LaRoche Ltd, BMS, Ignyta Operating INC, Merck Sharp & Dohme, Kymab, Pfizer, Tesaro, MSD, MedImmune, Exelixis Inc, LOXO Oncology Incorporated, Daiichi Sankio, Basilea Pharmaceutica International, Janssen-Cilag International, Merck KGAA; Financial Interests, Personal, Other, Consultant Advisory Board: Roche, EMD Serono, NMS Nerviano Medical Science, Sanofi, MSD, Novartis, Incyte, BMS, Menarini, AstraZeneca, Pierre Fabre. G. Lo Russo: Financial Interests, Personal, Advisory Board: MSD, Novartis, AstraZeneca, BMS, Pfizer, Roche, Sanofi; Financial Interests, Personal, Invited Speaker: Italfarmaco, Sanofi; Financial Interests, Institutional, Invited Speaker: BMS, MSD, GSK, Celgene, Novartis, Roche, AstraZeneca, Amgen. L. Rivoltini: Financial Interests, Personal, Invited Speaker, teaching in educational events: BMS; Non-Financial Interests, Personal, Advisory Role: DKTK. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

90P - Effect of age on safety and efficacy of novel cancer drugs investigated in early phase clinical trials

Presentation Number
90P
Lecture Time
17:35 - 17:35
Speakers
  • Jacopo Uliano (Milan, Italy)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Jacopo Uliano (Milan, Italy)
  • Eleonora Nicolo (Milan, Italy)
  • Sara Gandini (Milan, Italy)
  • Federica Giugliano (Milan, Italy)
  • Antonio Marra (New York, United States of America)
  • Stefania Morganti (Milan, Italy)
  • Emanuela Ferraro (Milan, Italy)
  • Paolo Tarantino (Boston, United States of America)
  • Paola Zagami (Milan, Italy)
  • Laura Boldrini (Milan, Italy)
  • Marzia A. Locatelli (Milan, Italy)
  • Angela Esposito (Bristol, United Kingdom)
  • Carmen Belli (Milan, Italy)
  • Ida Minchella (Milan, Italy)
  • Carmen Criscitiello (Milan, Italy)
  • Giuseppe Curigliano (Milan, Italy)

Abstract

Background

Although cancer often develops in advanced age, elderly patients (pts) are still underrepresented in clinical trials, especially phase I trials. Our study aimed to compare safety and efficacy of treatments in early phase clinical trials between younger and elderly pts (aged ≤/>65 years).

Methods

We retrospectively collected clinicopathologic characteristics and treatment outcomes of pts consecutively enrolled in early phase clinical trials at our institution from Jan 2014 to Apr 2021. Toxicity and clinical outcomes were compared between younger and elderly pts.

Results

419 pts were included in the analysis. Median age was 56 years (range 26-84); 107 (26%) pts were older than 65. The most common tumor types were breast (48%), lung cancer (9%), and melanoma (5%). Pts were treated in 64 trials primarily receiving immunotherapy (47%) or targeted agents (46%). Younger pts more frequently received targeted therapies compared to elderly (50% vs 35%; p=0.007). Elderly pts had worse baseline ECOG performance status (PS) (p=0.002), received fewer prior lines (p=0.04), and had fewer metastatic sites (p=0.04). Median follow-up was 20 months (range 1.7-85). Grade ≥3 adverse events were similar in both groups (31% vs 33%; p=0.7), even when treated with combination therapies. Elderly pts had a higher rate of treatment discontinuation due to treatment-related toxicities compared to younger (17% vs 7.9%; p=0.008). The objective response rate (19% vs 25%; p=0.22), disease control rate (48% vs 58%; p=0.07), and median progression-free survival (3.57 vs 5 months; p=0.08) were comparable in both groups. No significant difference in overall survival (OS) was observed by age groups (HR 0.91, 95% CI 0.69-1.19; p=0.47) after multivariate adjustment for ECOG PS, target lesions sum, and Royal Marsden Hospital prognostic score, which conversely were independently associated with OS.

Conclusions

Safety and efficacy of new targeted and immunotherapy drugs in elderly pts enrolled in early phase clinical trials are comparable to younger pts. These data suggest that age should not limit consideration of early phase clinical trials as a treatment option; enrollment of elderly pts should be even more encouraged in the era of precision medicine. Jacopo Uliano and Eleonora Nicolò have equally contributed to this work and share first authorship.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

P. Tarantino: Financial Interests, Personal, Invited Speaker: AstraZeneca; Financial Interests, Personal, Advisory Board: Daiichi Sankyo. C. Criscitiello: Financial Interests, Personal, Invited Speaker: Pfizer, Novartis, Eli Lilly, Roche, Gilead; Financial Interests, Personal, Advisory Board: MSD, Seagen, AstraZeneca, Daiichi Sankyo. G. Curigliano: Financial Interests, Personal, Invited Speaker: Roche, AstraZeneca, Daiichi Sankyo, Novartis, Pfizer, Pfizer; Financial Interests, Personal, Advisory Board: Ellipsis, Roche, AstraZeneca, Daiichi Sankyo, Lilly, Pfizer, Veracyte, BMS, Merck, Exact Sciences, Celcuity; Financial Interests, Institutional, Research Grant, Investigator Initiated Trial: Merck; Financial Interests, Institutional, Funding, Phase I studies: BMS, Novartis, AstraZeneca, Daiichi Sankyo, Roche, Blueprint Medicine, Kymab, Astellas, Sanofi, Philogen; Financial Interests, Institutional, Invited Speaker, Phase I clinical basket trial: Relay Therapeutics; Non-Financial Interests, Personal, Officer, Italian National Health Council as Advisor for Ministry of Health: Consiglio Superiore di Sanità; Non-Financial Interests, Personal, Advisory Role, Member of the Scientific Council, Patient Advocacy Association: Europa Donna; Non-Financial Interests, Personal, Advisory Role, Cancer Research Foundation: Fondazione Beretta; Non-Financial Interests, Personal, Invited Speaker, No compensation for this role, This a public national company for cancer prevention: Lega Italiana Lotta ai Tumori; Non-Financial Interests, Personal, Officer, Member of the Advisory Council: EUSOMA. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

91P - Molecular predictors of response to the therapy with mitotane in adrenocortical cancer

Presentation Number
91P
Lecture Time
17:35 - 17:35
Speakers
  • Erika Porubayeva (Moscow, Russian Federation)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Erika Porubayeva (Moscow, Russian Federation)
  • Liliya Urusova (Moscow, Russian Federation)

Abstract

Background

Adrenocortical cancer (ACC) therapy is characterized by insufficient effectiveness. Currently, mitotane, an adrenolytic drug, is the only drug approved for treatment of ACC and is used in the adjuvant setting and in case of metastatic or advanced disease. However, the administration of mitotane to certain groups of patients remains controversial due to the low response rates, high toxicity and limited data on the benefit of treatment. Expression levels of the large subunit of ribonucleotide reductase M1 (RRM1), cytochrome P450 2W1 (CYP2W1) and sterol-O-acyltransferase-1 (SOAT1) are considered as potential predictors of response to mitotane therapy. The aim of this study was to estimate the immunohistochemical expression of RRM1, CYP2W1 and SOAT1 in ACC as markers of clinical outcomes and response to the therapy with mitotane.

Methods

The study included 62 patients older than 17 years of age with a diagnosis of ACC confirmed histologically and immunohistochemically. Pathomorphological examination of surgical and consultative material from patients treated between 2005 and 2020 in Endocrinology research center and other health care centers of Russia was performed. Antibodies to SF-1, Ki-67, RRM1, CYP2W1, SOAT1 were used diluted in accordance with recommendations of firms-manufacturers for immunohistochemical detection. Kaplan-Meier method was used to estimate disease-free survival (DFS) and its predictors.

Results

Mitotane therapy was initiated in 29 patients in the postoperative period, the control group comprised 33 patients according to results of immunohistochemical examination (level of expression of Ki-67). In the control group of patients with low and moderate RRM1, CYP2W1 and SOAT1 immunoreactivity, a better DFS was observed (p=0.037, p=0.020 and p=0.001, respectively) compared to the study group at this level of marker expression. In case of high expression levels of the markers, no statistically significant differences were found.

Conclusions

In this study we have identified tendences in the correlation of the expression of potential prognostic markers and DFS in our sample of patients. Evaluation of RRM1, SOAT1 and CYP2W1 immunoreactivity opens new possibilities for personalized mitotane therapy in ACC.

Legal entity responsible for the study

Endocrinology Research Center, Moscow, Russia.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

93P - Circulating cell-free DNA fragmentation profiles during systemic therapy of advanced-stage non-small cell lung cancer patients

Presentation Number
93P
Lecture Time
17:35 - 17:35
Speakers
  • Jelena Milovanovic (Belgrade, Serbia)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Jelena Milovanovic (Belgrade, Serbia)
  • Ivana Boljevic (Belgrade, Serbia)
  • Jelena Spasic (Belgrade, Serbia)
  • Marijana Topalovic (Belgrade, Serbia)
  • Ana Krivokuca (Belgrade, Serbia)
  • Milena Cavic (Belgrade, Serbia)
  • Miljana Tanic (Belgrade, Serbia)

Abstract

Background

Circulating cell-free DNA (cfDNA) may be used for monitoring response to systemic treatment in patients with advanced carcinomas. Tumor-derived cfDNA is characterized by a shift of fragment sizes. The aim of the study was to evaluate longitudinally the changes in cfDNA fragmentation profile and correlate it with disease progression in non-small cell lung cancer (NSCLC) patients receiving anticancer systemic therapy.

Methods

We recruited 14 patients aged 36 - 72 years at diagnosis with advanced stage IIIb/IV NSCLC and performance status 0-2 at diagnosis. All patients were initially tested on PD-L1 expression as well as on mutations in the EGFR and the ALK genes. Patients with positive PD-L1 expression (n=3) received immunotherapy pembrolizumab as the first-line therapy, those with mutations in the EGFR gene (n=3) received tyrosine kinase inhibitor - gefitinib, one ALK-positive patient received alectinib, while other patients received standard chemotherapy - Carboplatin/Etoposide (n=3) or Carboplatin/Paclitaxel (n=4). Consecutive plasma samples were collected at diagnosis, during systemic treatment and after termination of the therapy. cfDNA was extracted from 0.5 mL of plasma using magnetic-based MagMax cfDNA extraction kit, and quantified using Qubit HS dsDNA assay and ddPCR. Contamination with genomic DNA was determined using a B-cell-specific ddPCR assay. Fragment size distribution of cfDNA was determined by Agilent Bioanalyzer HS dsDNA assay.

Results

Concentrations of cfDNA measured by Qubit (∼14.3 ng/mL of plasma) showed good correlation to the absolute quantification determined by ddPCR (∼13.22 ng/mL of plasma). In a 38% of samples there was small contamination by peripheral blood cells gDNA. Besides expected fraction of cfDNA (168 bp mode) observed in all samples, we observed in 48% of samples an ultra-short fraction of cfDNA (50bp mode).

Conclusions

Application of magnetic-based cfDNA extraction method allowed us to detect an ultra-short cfDNA fraction, previously unrecognized in lung cancer patients. Studies to correlate it to progression-free survival are ongoing.

Legal entity responsible for the study

Institute for Oncology and Radiology of Serbia.

Funding

The Science Fund of the Republic of Serbia (PROMIS/2020/6060876).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

94P - Targeted therapy in pediatric acute myeloid leukemia: An unmet need

Presentation Number
94P
Lecture Time
17:35 - 17:35
Speakers
  • Banda R. Teja (Hyderabad, India)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Banda R. Teja (Hyderabad, India)

Abstract

Background

Acute Myeloid Leukemia (AML) in children is rare with incidence of seven per million children annually. Recently a number of novel targeted therapies like FLT3 inhibitors, IDH inhibitors and BCL2 antagonists have been approved for adult AML. However conventional chemotherapy and allogenic stem cell transplantation remains the standard of care for pediatric AML. In the present study we analysed molecular genetics and their impact on induction outcome in paediatric AML patients.

Methods

This is a retrospective observational study. The study included newly diagnosed non M3 AML patients with age less than 18 years during 2018 and 2021. Age, gender, initial blood counts, FAB subtype of AML, molecular genetics, karyotype,-post induction bone marrow aspiration, MRD were analysed. All patients received standard 7+3 (Daunomycin+cytarbine) induction regimen.

Results

A total of 45 patients were evaluated. The median age at presentation was 13 years (range, 4-17 years) with male to female ratio of 1.5:1. Of these 45 patients, 11(24%) had t(8:21) abnormality, 6(13%) had NPM1 mutation without FLT3-ITD, 6(13%) patients had FLT3-ITD mutation, 3(6%) patients had inv(16), 3(6%) patients were positive for FLT3-TKD mutation and 16(35%) patients did not have any cytogenetic abnormality. Post induction marrow examination was complete remission, partial remission and persistence of disease in 25(56%), 11(24%) and 9(20%) patients respectively. Five (83%) out of 6 patients with FLT3-ITD and all FLT3 TKD mutation positive patients had persistence of disease (p<0.01). Post induction MRD was available in 14 patients. In all these 14 cases MRD correlated with bone marrow aspiration findings and all patients with CR had MRD negativity after induction. No mortality during induction was observed.

Conclusions

FLT3 mutations was observed in 19%{FLT3 ITD(13%), FLT3 TKD(6%)} of pediatric AML patients and appears to have an adverse impact on induction outcome in paediatric AML. FLT3 ITD/TKD inhibitors during induction have shown improved survivals in adult AML. With increasing incidence of pediatric AML in recent years, there is an unmet need to identify targeted and novel therapies for better outcomes in paediatric AML.

Legal entity responsible for the study

The author.

Funding

Has not received any funding.

Disclosure

The author has declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

95P - Anticancer activity of Inula recemosa root extract in human liver cancer cell line by attenuation of OCT4/Sox2 axis

Presentation Number
95P
Lecture Time
17:35 - 17:35
Speakers
  • Tania Ghosh(Sarkar) (Kolkata, India)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Tania Ghosh(Sarkar) (Kolkata, India)
  • Anwesha Mukherjee (Kolkata, India)
  • Bratoti Nag (Pune, India)
  • Arijit Ghosh (Kolkata, India)
  • Suvra Mondal (Kolkata, India)
  • Rittwika Bhattacharyya (Kolkata, India)
  • Soma Mukhopadhyay (Kolkata, India)

Abstract

Background

Hepatocellular carcinoma (HCC) has become one of the leading causes of death globally in males and is also rising in females at an alarming rate. The growth, oncogenicity, metastasis and therapeutic resistance of hepatocellular carcinoma is maintained by hepatic cancer stem cells (HCSCs). The development of new therapeutic approaches specifically targeting hepatic cancer stem cells using herbal medicine could propose new hope for advanced HCC treatment. To achieve therapeutic success, emphasis should also depend on inhibitors that would not only be more effective on tumors but have minimal normal tissue toxicity. Considering the hepatoprotective role of Inula recemosa suggested before, the aim of our present study was to understand the role of Inula recemosa root (IRE) on hepatocellular carcinoma, and hepatic cancer stemness.

Methods

The cytotoxicity effect of n-hexane extract of Inula recemosa was evaluated by MTT assay on HepG2 liver cancer and WRL68 liver normal cell lines. Colony formation was also performed to measure reproductive integrity after treating with different concentration over a prolonged period of time. Flowcytometry method was used for cell cycle analysis, apoptosis and the production of reactive oxygen species (ROS) by DCFDA. Effect of IRE on cancer stemness transcription factors like SOX 2 and OCT4 transcript level was determined by RT-PCR method. Immunocytochemistry method was used to check the change in cancer stemness marker protein expression after IRE treatment.

Results

IRE showed very low toxicity on WRL68 normal liver cell line but robust antiproliferative effect on HepG2 cell line by induction of apoptosis. The induction of apoptosis was supported by increase in ROS production level in IRE treatment groups when compared with untreated cells. Cell cycle analysis showed arrest in Sub G0 phase. IRE also attenuated transcript level and protein expression of stemness markers.

Conclusions

Our data endorses the potential of IRE in hepatocellular carcinoma on targeting the cancer stem cell transcription factors for the first time. We can conclude that IRE might open up new therapeutic avenues on advanced therapies of hepatocellular carcinoma in near future.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

96P - The Hammersmith score optimises patient selection and predicts for overall survival in early-phase cancer trial participants independent of tumour burden

Presentation Number
96P
Lecture Time
17:35 - 17:35
Speakers
  • James A. Korolewicz (London, United Kingdom)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • James A. Korolewicz (London, United Kingdom)
  • Bernhard Scheiner (Vienna, Austria)
  • Claudia Fulgenzi (London, United Kingdom)
  • Antonio D'Alessio (Rozzano, Italy)
  • Alessio Cortellini (London, United Kingdom)
  • Chynna Pascual (London, United Kingdom)
  • Aman Mehan (London, United Kingdom)
  • Sarah E. Partridge (London, United Kingdom)
  • Oreoluwa Mohammed (London, United Kingdom)
  • Aneta Gupta (London, United Kingdom)
  • Lauren Booker (London, United Kingdom)
  • Susan Cleator (London, United Kingdom)
  • James Rackie (London, United Kingdom)
  • Yasmine Needham (London, United Kingdom)
  • Jonathan Krell (London, United Kingdom)
  • Laura Tookman (London, United Kingdom)
  • Won-Ho Edward Park (London, United Kingdom)
  • Muzamil Asif (London, United Kingdom)
  • Joanne Evans (London, United Kingdom)
  • David J. Pinato (London, United Kingdom)

Abstract

Background

Significant anti-tumour response is increasingly seen in early-phase cancer trials (EPCT), requiring optimal patient selection and accurate prognostication. Hammersmith Score (HS), a simple prognostic index derived on routine biochemical measures (Albumin <35g/L, Lactate Dehydrogenase (LDH) >450 U/L, Sodium <135mmol/L) predicts response and survival in EPCT participants. HS has not been validated in the cancer immunotherapy era.

Methods

We retrospectively analyzed characteristics and outcomes of 212 referrals to our early-phase unit (12/2019-12/2022). Independent predictors for overall survival (OS) were identified from univariable (Kaplan Meier) and multivariable (Cox regression). HS was calculated for 66 patients and compared with the Royal Marsden Score (RMS) to predict OS and 90-day mortality (90DM). Multivariable logistic regression and ROC curves were used to estimate 90DM and c-index was used to compare prognostic models.

Results

Of 212 referrals, 147 patients were screened and 82 patients treated. Median age 62.1 years (52.2-71.9); median OS 7.3 months (95% CI: 6.2-8.4); 45.1% male; 63.4% performance status ≥1, median number of metastatic sites was 2. Experimental therapies included immunotherapy (n=53), targeted therapy (n=6) and chemotherapy (n=23). Prognostic stratification by HS identifies significant difference in median OS: 11.28 months for HS0 (95%CI: 8.7-13.8), 5.29 months for HS1 (95%CI: 2.0-8.6) and 1.2 months for HS2-3 (95% CI: 1.0-1.3, p<0.001). ROC analysis showed HS and RMS perform similarly in predicting 90DM (AUROC 0.59 and 0.56 respectively). HS was confirmed as a multivariable predictor for OS (HR: HS1 vs. 0 2.6 (95%CI: 1.1-6.3), p=0.030; HS 2/3 vs. 0: 8.5 (95%CI: 1.9-37.4), p=0.005, C-index 0.775) with similar multivariable predictive ability as RMS (HR: RMS 2 vs. 0/1 7.8 (95%CI: 2.1-28.6), p=0.002; HS3 vs. 0/1 11.1 (95%CI: 2.9-42.5), p<0.001, C-index 0.741).

Conclusions

HS is a validated prognostic index for patients with advanced cancer treated in the context of modern EPCTs, independent of tumour burden, removing the need for quantification during patient selection. HS is a simple, inexpensive selection tool to optimise referral for EPCT.

Legal entity responsible for the study

D.J. Pinato.

Funding

Has not received any funding.

Disclosure

B. Scheiner: Financial Interests, Personal, Other, Travel support: AbbVie, Ipsen, Gilead. A. D'Alessio: Financial Interests, Personal, Other, Travel support and consultancy fees: Roche. A. Cortellini: Financial Interests, Personal, Advisory Board: AstraZeneca, MSD, OncoC4; Financial Interests, Personal, Invited Speaker: AstraZeneca, MSD, Eisai, BMS. D.J. Pinato: Financial Interests, Personal, Advisory Board: Mina Therapeutics, Eisai, Exact Sciences, MURSLA, H3B, DaVolterra, AstraZeneca, Bayer Healthcare; Financial Interests, Personal, Invited Speaker: BMS, Ipsen, Roche; Financial Interests, Personal, Other, Editor in Chief role: Wiley; Financial Interests, Institutional, Research Grant: BMS, MSD; Non-Financial Interests, Personal, Principal Investigator: Incyte, H3B, Starpharma, Roche, Ribon Therapeutics, Turning Point Therapeutics, Apollomics; Non-Financial Interests, Personal, Other, Charity Trustee: Cancer Treatment and Research Trust. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

97P - Bcl-xL prevents the arginine starvation induced by PEGylated arginine deiminase (ADI-PEG20) from inducing apoptosis

Presentation Number
97P
Lecture Time
17:35 - 17:35
Speakers
  • Brian A. Van Tine (St. Louis, United States of America)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Brian A. Van Tine (St. Louis, United States of America)
  • Prashanta Panda (St. Louis, United States of America)
  • Leonard Rogers (St. Louis, United States of America)
  • Toshinao Oyama (St. Louis, United States of America)

Abstract

Background

Multiple clinical trials are using PEGylated arginine deiminase (ADI-PEG20) to induce arginine starvation in cancers that are argininosuccinate synthetase (ASS1) low. ASS1 low tumors include sarcomas, melanomas, non-small cell and small cell lung cancers, triple-negative breast, pancreatic cancer, and many others. While ADI-PEG20 clearly induces an arginine starvation response, no cell death is initiated at a meaningful level. While many studies have seen low levels of apoptosis (<10%), the primary effect of ADI-PEG20 is cytostatic, as tumors metabolically reprogram themselves by upregulating ASS1, gaining the ability to produce arginine.

Methods

Cell lines were treated with ADI-PEG20 and A-1331852 (Bcl-xL inhibitor) and monitored for cell death using the Incucyte. CRISPR models confirmed that the Bcl-xL effects of A-1331852 were on target. CDK2 activity, Bcl-xL and Mcl-1 were measured in response to ADI-PEG20 using Wes capillary electrophoresis. These cell lines were grafted into mice and treated with ADI-PEG20 and A-1331852.

Results

Arginine starvation inhibits CDK2, resulting in the loss of Mcl-1 expression. Neither Bcl-2 nor Mcl-1 inhibitors initiated cell death when combined with ADI-PEG20. Dual treatment of Bcl-xL inhibitor A-1331852 with ADI-PEG20 led to mitochondrial BAX levels increasing fivefold, corresponding to a drop in cytoplasmic levels that suggests translocation to the mitochondria. A similar but less dramatic pattern of significant increase in mitochondrial levels was observed with BAK. Inhibition of Bcl-xL with ADI-PEG20 induced CASP3/7 and PARP cleavage, resulting in apoptosis in a panel of ASS1 low cell lines. This was confirmed in vivo.

Conclusions

ADI-PEG20 does not induce cell death as a monotherapy, but the arginine starvation instead results in metabolic adaptation. This begins to explain the lack of efficacy seen in early ADI-PEG20 monotherapy trials. Bcl-xL, but not Mcl-1 or Bcl-2, prevents ADI-PEG20 from inducing apoptosis. Bcl-xL inhibition synergies with ADI-PEG20 to induce apoptosis. This study provides the preclinical rationale for combining ADI-PEG20 with next generation Bcl-xL inhibitors such as the Bcl-xL PROTAC in a phase I clinical trial.

Legal entity responsible for the study

The authors.

Funding

Polaris, NIH.

Disclosure

B.A. Van Tine: Financial Interests, Personal, Invited Speaker, Educational Speaker: Targeted Oncology; Financial Interests, Personal, Advisory Board, Also, Travel paid to conference to present abstract: Adaptimmune; Financial Interests, Personal, Invited Speaker, Also attended an Ad Board meeting, Travel was paid to present abstract at conference: GSK; Financial Interests, Personal, Other, Consulting, Also attended and Ad Board meeting, Travel was paid to attend an Ad board meeting: Epizyme; Financial Interests, Personal, Other, Consulting- ADRx working on a cancer project and they are requesting my expertise: ADRx; Financial Interests, Personal, Advisory Board, tenosynovial giant cell tumors (TGCT): Ayala Pharmaceuticals; Financial Interests, Personal, Other, Consulting/Advisor: Cytokinetics; Financial Interests, Personal, Other, Consulting: Bayer; Financial Interests, Personal, Other, 60 minute interview regarding Synovial Sarcoma: Bionest Partners; Financial Interests, Personal, Other, OncLive Virtual Workshop: Intellisphere; Financial Interests, Personal, Advisory Board, Attended Advisory Board Meeting: Apexigen; Financial Interests, Personal, Advisory Board, Attended an Advisory Board Meeting: Daiichi Sankyo, Deciphera Pharmaceuticals; Financial Interests, Personal, Advisory Board, Attended an Advisory Board Meeting: Novartis; Financial Interests, Personal, Advisory Board: Lilly, PTC Therapeutics; Financial Interests, Personal, Invited Speaker, Sigma-2 Receptor Ligands and Therapeutic uses therefor (006766), Modular Platform for Targeted Therapeutic Delivery (006755), Sigma-2 Receptor Ligand Drug Conjugates as Antitumor Compounds, Methods of synthesis and Uses Thereof (014229): Accuronix Therapeutics; Financial Interests, Institutional, Research Grant: Pfizer, Merck, Tracon Pharm, GSK; Non-Financial Interests, Personal, Invited Speaker: Polaris. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

98P - Cationic dendrimers as prospective vehicles of therapeutic nucleic acids into tumor cells: Approaches, advantages and challenges

Presentation Number
98P
Lecture Time
17:35 - 17:35
Speakers
  • Nadezhda Knauer (Novosibirsk, Russian Federation)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Nadezhda Knauer (Novosibirsk, Russian Federation)
  • Ekaterina Pashkina (Novosibirsk, Russian Federation)
  • Olga Boeva (Novosibirsk, Russian Federation)
  • Alina Aktanova (Novosibirsk, Russian Federation)
  • Valeria Arkhipova (Novosibirsk, Russian Federation)
  • Maria Meschaninova (Novosibirsk, Russian Federation)
  • Jean Pierre Majoral (Toulouse, France)
  • Ann-Christin Nickel (Düsseldorf, Germany)
  • Sajjad Muhammad (Düsseldorf, Germany)
  • Daniel Hänggi (Düsseldorf, Germany)
  • Ulf Dietrich Kahlert (Magdeburg, Germany)
  • Vladimir Kozlov (Novosibirsk, Russian Federation)
  • Evgeny Apartsin (Toulouse, France)

Abstract

Background

Challenges of nanomedicine in oncology require therapeutic tools to affect metabolic pathways in the target cells. Therapeutic nucleic acids (NAs) are promising instruments as siRNAs target them selectively, while microRNAs can reach several targets. However, oligonucleotides suffer from rapid nuclease degradation in biological media. Cationic dendrimers – highly symmetric hyperbranched molecules – can be efficient vehicles. In the current work we studied challenges of using polycationic phosphorus and carbosilane dendrimers and their complexes with oligonucleotides (dendriplexes).

Methods

Different tumor models were used: Jurkat (single-cells suspension culture in serum-containing media), BTSC233, JHH520, NCH644, GBM1 (glioblastoma cancer stem cells (GSCs) as neurospheres in serum-free medium), U87 (“standard” glioblastoma culture). Human PBMCs and iPSCs were used as non-tumor controls. We used miR-34, synthetic inhibitor of miR-21 and siRNA against LYN kinase (siLyn) as therapeutics. Such biological effects as internalization, cytotoxicity (incl. comparison to standard chemodrugs), apoptosis induction, expression of surface markers, related to interactions with immune microenvironment (PD-L1, TIM-3, CD47), LYN-expression were investigated.

Results

Dendrimers have their own cytotoxic effects on tumor cells, which are comparable or superior to standard chemodrugs; in some models the toxic effect on tumor cells was higher than on non-tumor cells. Moreover, GSCs showed higher sensitivity to molecules. Cationic dendrimers efficiently delivered NAs into tumor cells. Dendriplexes showed antitumor effects: decrease of cell viability, apoptosis induction. Dendrimers and dendriplexes affected the expression of PD-L1 and TIM-3, which could be a sign of cell stress. siLyn-dendriplexes decreased tumor cell viability but did not change significantly LYN expression. Patterns of changes strongly depended on the cell type and cultivation mode.

Conclusions

We showed promising antitumor activity of cationic dendrimers and their complexes with therapeutic NAs. However, further investigations of the impact of different cell type and model features in view of pre-clinical testing is to be performed.

Legal entity responsible for the study

The authors.

Funding

European Cooperation in Science and Technology CA17140.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

99P - Quantitative indicators of TREC and KREC excision rings in malignant neoplasms

Presentation Number
99P
Lecture Time
17:35 - 17:35
Speakers
  • Alexander V. Sultanbaev (Ufa, Russian Federation)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Alexander V. Sultanbaev (Ufa, Russian Federation)
  • Shamil Musin (Ufa, Russian Federation)
  • Konstantin Menshikov (Ufa, Russian Federation)
  • Nadezda Sultanbaeva (Ufa, Russian Federation)
  • Irina Menshikova (Ufa, Russian Federation)
  • Alfiya Fatikhova (Ufa, Russian Federation)
  • Mikhail Sultanbaev (Ufa, Russian Federation)
  • Vadim Askarov (Ufa, Russian Federation)
  • Dmitry Kudlay (Moscow, Russian Federation)

Abstract

Background

The excisional rings of the T cell receptor rearrangement (TREC) and the κ-deletion element (KREC) are extrachromosomal DNA structures formed during V(D)J-recombination. A decrease in the number of TREC and KREC below age-related values may be a manifestation of immunodeficiency conditions, which can be caused by oncological and hematological diseases.

Methods

To study the change in the amount of TREC and KREC in malignant neoplasms. Methods. For the study, blood was taken from patients with malignant neoplasms in the main group and group of healthy individuals of various ages. At the stage of intermediate evaluation 206 studies were performed out of 700 planned. Of these 26 healthy individuals and 180 patients with malignant neoplasms. Sex distribution: 22.8% (46/206) men, 77.2% (159/206) women, median age 54 years (Q1-Q3: 44-66 years). From MN: breast cancer 35% (63/180), ovarian cancer 21.1% (38/180), melanoma 15.6% (28/180), lymphoma 11.1% (20/180), lung cancer 8.9%(16/180), colorectal cancer 8.3%(15/180). Quantification of TREC and KREC was performed by real-time PCR using the IMMUNO-BIT reagent kit (ABV-test LLC) in accordance with the instructions for the kit. DNA extraction from whole blood was performed using the AmpliPrime RIBO-prep reagent kit (NextBio LLC).

Results

According to our study, in healthy individuals, the TREC level is 75.6/105 PBMC (Q1-Q3: 19.2-135.3), the KREC level is 317.3/105 (Q1-Q3: 118.1-565.9). Whereas in patients with MN, the level of TREC is 4.6/105 PBMC (Q1-Q3: 0.9-17.3), the level of KREC is 111.9/105 (Q1-Q3: 29.3-339.28). When comparing the TREC and KREC indicators in different groups, statistically significant differences were established (p<0.001, p=0.006). The levels of TREC and KREC in the healthy population were significantly higher than among patients with cancer (median TREC were 75.6 and 4.6, median KREC 317.3 and 111.9, respectively).

Conclusions

The data obtained demonstrate significant changes in T- and B-cell lymphopoiesis in patients with cancer. Quantitative determination of TREC and KREC makes it possible to assess the state of the T- and B-cell link of the immune system in patients with malignant neoplasms.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

100P - RS-0139, a novel tumor-targeted delivery of docetaxel, with potent anti-tumor activity in a broad spectrum of tumor cell lines and xenograft models

Presentation Number
100P
Lecture Time
17:35 - 17:35
Speakers
  • Gulsah Nomak (Istanbul, Turkey)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Gulsah Nomak (Istanbul, Turkey)
  • Merve Karacivi (Istanbul, Turkey)
  • Sedef Kutlu Ozkaya (Istanbul, Turkey)
  • Uboldi Valeria (Lugano, Switzerland)
  • Burcu Sumer Bolu (Istanbul, Turkey)
  • Tugce Karatas (Istanbul, Turkey)
  • Carlo V. Catapano (Lugano, Switzerland)
  • Hakan Sedat Orer (Istanbul, Turkey)
  • Amitav Sanyal (Istanbul, Turkey)
  • Rana Sanyal (Istanbul, Turkey)

Abstract

Background

Docetaxel (DTX), an anti-microtubule agent, suffers from limitations due to its toxicity profile, which includes hypersensitivity reactions, neutropenia, neuropathy, fatigue, and nausea. SagittaTM platform technology is discovered to reduce toxicity while maximizing the efficacy of cytotoxic drugs. Sagitta™ Bir, a water-soluble polymeric carrier, allows covalent binding of multiple active substances and targeting moieties to the polymer backbone. RS-0139 is the first drug candidate developed with this platform.

Methods

RS-0139 is a tumor-targeted, DTX-releasing prodrug. The active molecule (DTX) is covalently bound to the polymer backbone via a short peptide releasing only upon entry to the cell via endocytosis. The carrier is also equipped with a targeting peptide which shows high affinity to αvβ3, αvβ5, and αvβ6 integrin receptors highly expressed on the tumor cell. A comprehensive set of in vivo studies are conducted to assess potential anti-tumor activity on A549, NCI-H460, MDA-MB-231, 22Rv1 cell lines.

Results

Physical properties of RS-0139 such as enhanced aqueous solubility provide an advantage over the currently used taxanes. Also, the pharmacokinetic profile is not only improved in terms of half-life and AUC but also is demonstrated to be translatable across species. In preclinical studies, the enzymatically cleavable covalent conjugation of DTX enabled sustained release, reducing adverse effects due to limited circulating free DTX. Hence, RS-0139 showed superior tolerability in healthy mice, rats, and rabbits compared to DTX. Besides, the dosing frequency was reduced due to the 5-fold increase in the half-life. Preclinical data strongly supported the clinical translation of this novel nanomedicine for the treatment of solid tumors. The phase I clinical trial is ongoing on NSCLC patients.

Conclusions

Tumor cell expression of the integrins is correlated with disease progression in various tumor types such as breast, pancreatic, prostate, non-small cell lung cancers and glioblastoma. Preclinical studies demonstrate promising results for NSCL, breast and prostate cancers.

Legal entity responsible for the study

RS Research.

Funding

RS Research.

Disclosure

G. Nomak, R. Sanyal: Financial Interests, Personal, Member of the Board of Directors: RS Research. M. Karacivi, S. Kutlu Ozkaya, B. Sumer Bolu, T. Karatas: Financial Interests, Personal, Full or part-time Employment: RS Research. H.S. Orer: Financial Interests, Personal, Principal Investigator: RS Research. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

101P - Prediction of radiation responses in patients with locally advanced rectal cancer with a patient-derived organoid-based radiosensitivity model

Presentation Number
101P
Lecture Time
17:35 - 17:35
Speakers
  • Samart Phuwapraisirisan (Bangkok, Thailand)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Samart Phuwapraisirisan (Bangkok, Thailand)
  • Praewphan Ingrungruanglert (Bangkok, Thailand)
  • Nipan Israsena (Bangkok, Thailand)
  • Chucheep Sahakitrungruang (Bangkok, Thailand)
  • Songphol Malakorn (Bangkok, Thailand)

Abstract

Background

Identifying locally advanced rectal cancer patients responsive to chemoradiotherapy before surgery remains a challenge. Patient-derived organoid (PDOs) biobanks may potentially predict therapeutic response to chemotherapy and radiation. We conducted a prospective clinical trial with treatment-naive rectal cancer patients and matched patient-derived tumor organoids to determine whether a correlation exists between experimental results obtained after irradiation in patients and organoids.

Methods

Between May 2019 and June 2021, 55 patients who were diagnosed with pathologically confirmed mid-to-lower rectal cancer were prospectively enrolled. High-risk patients with locally advanced rectal cancer (LARC) indicating neoadjuvant chemoradiotherapy (NCRT) according to ESMO guidelines. Pre-NCRT rectal cancer tissue samples were obtained. Tumor organoids were isolated and cultured. The mutational status of tissues and organoids was analyzed. Long course NCRT with total dose of 50.4-55.8Gy in 28 to 31 fractions was performed. Infusional 5-FU or oral capecitabine or XELOX was administered. Radical surgeries performed at 8-12 weeks after completing radiation. Pathologic response after NCRT was evaluated using the tumor regression grade (TRG) system. To validate the response of PDTOs to irradiation in vitro, we performed a radiation dose-dependent (0, 2, 4,6 and 8Gy) survival analysis of 37 PDOs.

Results

PDOs radiation dose-viability curves showed substantial inter-patient heterogeneity in radiosensitivity. PDOs from post-NCRT tissues displayed higher radioresistance than those from treatment-naïve specimens. We observed strong correlation between each group and clinical outcome post NCRT. 6 of 7 patients (85.7%) whose PDOs assigned to PDOs-radioresistant group showed poor response (TRS score = 2) after NRCT, while 4 of 7 patients (57.1%) whose PDOs assigned to PDOs-radiosensitive group displayed good response (TRS = 1) and 2 of 7 patients (28.5 %) showed complete response (TRS = 0).

Conclusions

Our data provide promising evidence for correlations between in vitro organoid responses and clinical responses to NCRT in locally advanced rectal cancer.

Legal entity responsible for the study

Faculty of Medicine, Chulalongkorn University.

Funding

C2F Funding, Chulalongkorn University.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

102P - Co-expression analysis of genes encoding proteasome subunits and XPO1-related proteins in the Cancer Genome Atlas (TCGA) and the Gene Tissue Expression (GTEx) databases as a tool to devise new treatment strategies

Presentation Number
102P
Lecture Time
17:35 - 17:35
Speakers
  • Vito Spataro (Bellinzona, Switzerland)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Vito Spataro (Bellinzona, Switzerland)
  • Antoine Buetti-Dinh (Manno, Switzerland)

Abstract

Background

The 26S proteasome is a multiprotein complex encoded by 32 genes. Proteasome inhibitors (PI) used in the clinic target subunit (SU) β5 encoded by the PSMB5 gene, but several additional SU are functionally relevant and potential drug targets. The nucleocytoplasmic export (NE) protein XPO1 is a novel established target for anticancer treatment and homologues of SU PSMD14 and of XPO1 are involved in AP-1 mediated drug resistance in the fission yeast model . An integrative study on the expression of proteasome genes and XPO1-related genes in tumors can be informative for the development of novel treatment strategies.

Methods

We extracted data on RNA expression of all 32 proteasome genes and all 37 genes encoding XPO1-interacting proteins according to OpenCell database in all tumours (T) of TCGA and in normal tissues (NT) of the GTEx Project. We performed gene co-expression analysis (GCA) for each gene pair and calculated Pearson correlation R coefficients. The matrices of GCA were compared across T and NT and the Euclidean distance was used to cluster the correlation matrices into a dendrogram. A machine learning algorithm was used to identify the genes with highest classification weights.

Results

The R coefficients for co-expression of proteasome genes are very high in NT and significantly lower in the majority of T. Based on GCA, cluster analysis can clearly separate T and NT. Sixteen of the 20 genes contributing most to the separation are proteasome genes. This set includes PSMB5 (the target of approved PI), 8 SU of the proteasome core, 7 SU of the regulatory particle and XPO1 (the target of Selinexor). Several gene pairs encoding proteasome SU and NE proteins have highly correlated expression (R>0.7). PSMD14 and XPO1 are highly correlated (R>0.7) in some T types (breast, prostate, lung) and not in their NT counterparts.

Conclusions

We conclude that: 1) the expression of proteasome genes is severely altered in T and a subset of them is more frequently deregulated 2) there is a high positive correlation between the expression of some proteasome genes and several genes involved in NE 3) the gene pair PSMD14-XPO1 is frequently correlated in T and not in NT.

Legal entity responsible for the study

V. Spataro.

Funding

Has not received any funding.

Disclosure

V. Spataro: Non-Financial Interests, Personal, Advisory Board: Roche Pharma, Takeda Pharma; Non-Financial Interests, Personal, Invited Speaker: Novartis Pharma. All other authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

103P - Microsomal triglyceride transfer protein as a prognostic and therapeutic marker for brain cancer

Presentation Number
103P
Lecture Time
17:35 - 17:35
Speakers
  • Ryuk Jun Kwon (Yangsan, Korea, Republic of)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Ryuk Jun Kwon (Yangsan, Korea, Republic of)
  • Soomin Son (Yangsan, Korea, Republic of)

Abstract

Background

Glioblastoma multiforme (GBM) is the common malignant brain cancer and has poor prognosis despite development of modern medicine. The lipid metabolism is known to be correlated to cancer development and progression. Microsomal Triglyceride Transfer Protein (MTTP) is a molecule to lower lipids and its inhibitor, lomitapide, is involved in the improvement of hyperlipidemia. However, whether the altered expression of MTTP affects the development and prognosis of GBM is not identified.

Methods

Data for patients with brain cancer were prepared from The Cancer Genome Atlas (TCGA) database. Using t test or Mann-Whitney U test, datasets were compared to the expression of MTTP in normal and brain cancer tissues. The log-rank test and multivariable Cox proportional hazard regression were performed to assess if MTTP significantly affects the prognosis of patients with brain malignant tumor.

Results

MTTP expression was significantly higher in brain cancer than that in normal and the expression of MTTP in GBM was significantly higher than that in LGG. Moreover, the expression level of MTTP was significantly correlated with and cancer stage. MTTP expression was significantly increased in the IDH-WT than in the IDH mutant. Overall Survival (OS) of MTTP high and low expression groups was estimated 36.82 ± 6.612 and 75.02 ± 8.096, respectively (p < 0.001). Similar to this result, high MTTP expression had a poorer survival than low MTTP expression in LGG and GBM, respectively. In addition, brain cancer patients with older age and high MTTP expression had a significantly poor survival (median OS: 12.56 ± 0.876 months, p < 0.001).

Conclusions

The expression of MTTP was higher in brain cancer than in normal tissues. In addition, its expression was correlated to the age and stage of brain cancer patients. This results suggest that the function of MTTP may depend on the cancer type. The OS, PFS, and DSS of patients with high MTTP expression showed shorter than those with low MTTP expression. These results suggest that MTTP is a prognostic indicator in brain cancer. Taken together, our study will provide a framework for investigating the molecular mechanism of brain cancer development and progression and the additional use of lomitapide for GBM treatment.

Editorial acknowledgement

The authors would like to acknowledge The Cancer Genome Atlas database and the GEO database.

Legal entity responsible for the study

The authors.

Funding

This study was supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (NRF-2022R1F1A107476911; RJK) and Research Institue for Convergence of biomedical science and technology, Pusan National University Yangsan Hospital (30-2023-005).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

104P - Choline transporter-like protein 1 is a novel molecular target for the treatment of hepatocellular carcinoma

Presentation Number
104P
Lecture Time
17:35 - 17:35
Speakers
  • Masato Inazu (Tokyo, Japan)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Masato Inazu (Tokyo, Japan)
  • Akane Hase (Shinjuku-ku, Japan)
  • Naoko Tajima (Shinjuku-ku, Japan)
  • Tsuyoshi Yamanaka (Shinjuku-ku, Japan)

Abstract

Background

Hepatocellular carcinoma (HCC), the most common type of primary liver cancer, has a high incidence and mortality rate, and it is desirable to develop new therapeutic agents to overcome unmet medical needs. Choline is essential for the synthesis of phospholipids, the main component of cell membranes, and abnormal choline accumulation in cancer cells is strongly correlated with malignant tumor growth. Recently, choline transporter-like protein 1 (CTL1), which is highly expressed in cancer cells, has attracted attention as a novel target molecule in cancer therapy. In this study, we analyzed the functional expression of choline transporters in human HCC cells and investigated the antitumor effects of CTL1 inhibitors.

Methods

The characteristics of [3H]choline uptake were examined using a highly differentiated human HCC cell line, HuH-7.Expression analysis of choline transporters was performed by RT-PCR and Western blotting. Cell viability and caspase-3/7 activity were measured using the CellTiter-Glo Luminescent Cell Viability Assay and Caspase-Glo 3/7 Assay System, respectively.

Results

CTL1 was highly expressed in HuH-7 cells and localized to the cell membrane. [3H]Choline uptake was Na+-independent and pH-dependent, via a single transport system. Annexin V-positive cells were observed when cells were cultured in choline-deficient conditions. CTL1 inhibitors decreased cell viability, increased caspase-3/7 activity, and inhibited migration of HuH-7 cells. CTL1 inhibitors activated the sphingomyelinase/ceramide pathway, and ceramide inhibited cell survival and increased caspase-3/7 activity.

Conclusions

The transport of extracellular choline is mediated by CTL1 in HuH-7 cells, and inhibition of CTL1 function induces apoptosis, suggesting that CTL1 may be a new target molecule for cancer therapy. Furthermore, CTL1 inhibitors induced apoptotic cell death through the ceramide pathway, suggesting that it may be a novel therapeutic agent for liver cancer. It also has an inhibitory effect on the migration of HuH-7 cells and is expected to suppress lymph node metastasis in HCC patients.

Legal entity responsible for the study

Tokyo Medical University.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

106P - Knockout of lncRNA-CCAT1 with the use of CRISPR-Cas9 system and G7 PAMAM dendrimers influences apoptosis and proliferations of NSCLC cells

Presentation Number
106P
Lecture Time
17:35 - 17:35
Speakers
  • Mateusz Iwanski (Rzeszow, Poland)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Mateusz Iwanski (Rzeszow, Poland)
  • Konrad Kwaśniak (Rzeszow, Poland)
  • Joanna Olszewska (Rzeszow, Poland)
  • Anna Głowacka (Rzeszow, Poland)
  • Konrad Wróbel (Rzeszow, Poland)
  • Staniwsław Wołowiec (Rzeszow, Poland)
  • Jacek Tabarkiewicz (Rzeszow, Poland)

Abstract

Background

Colon cancer-associated transcript 1 (CCAT-1) is an oncogenic lncRNA that has been emerged as a vital biomarker for diagnosis, prognosis and therapeutic interventions in multiple malignancies. The previous studies showed that lncRNA-CCAT1 was upregulated in NSCLC cells and its expression was related to tumor growth and reduced survival rate. The aim of our study was to evaluate influence of the knockout of lncRNA-CCAT1 with the use of CRISPR-Cas9 system and G7 PAMAM dendrimers on apoptosis and proliferations of NSCLC cells.

Methods

We used two human lung adenocarcinoma cell lines: A549, H1975 and H1703 squamous cell carcinoma cell line. The knockout of the lncCCAT expression was performed using the CRISPR-Cas9 system and G7 PAMAM dendrimers. We used 4 combinations of gRNAs. The apoptosis of NSCLC after lncRNA-CCAT1 knockout was estimated with the use of flow cytometry and Annexin V staining, evaluation of caspase-3/7 and measurement of mitochondrial membrane potential changes. Expression of Ki67 was measured by flow cytometry to evaluate NSCLC cells proliferation. All mentioned above parameters were evaluated 24, 48 and 72 hours after transfection. Nonparametric ANOVA tests was used for statistical analysis.

Results

We found that transfection with conjugates of G7 PAMAM dendrimers and px459 v2.0, the appropriate gRNAs (for lncCDH5-3:3 knockout), and pcDNA3.1 plasmid are downregulating expression of lncRNA-CCAT1. We confirmed that apoptosis of NSCLC was increased after transfection and cells proliferation was reduced. We also found differences in timing and intensity of biological effects when different combination of gRNAs are used in particular NSCLC cell lines.

Conclusions

The conjugates of G7 PAMAM dendrimers and px459 v2.0, the appropriate gRNAs (for lncCDH5-3:3 knockout), and pcDNA3.1 plasmid can be used for knockout of the expression of lncRNA-CCAT1. On the other hand the gRNAs shall be individually chosen for particular NSCLC cells according to their genetic mutation status.

Legal entity responsible for the study

University of Rzeszow.

Funding

Polish Ministry for Higher Education and Science.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

107P - Censoring imbalance in ACIS trial for prostate cancer

Presentation Number
107P
Lecture Time
17:35 - 17:35
Speakers
  • Noa Zimhony-Nissim (Beer Sheva, Israel)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Noa Zimhony-Nissim (Beer Sheva, Israel)
  • Shai Gilboa (Tel Aviv, Israel)
  • David Bomze (St. Gallen, Switzerland)
  • Tomer Meirson (Petah Tikva, Israel)
  • Gal Markel (Ramat Gan, Israel)

Abstract

Background

In the ACIS trial, apalutamide plus abiraterone-prednisone has demonstrated longer radiographic progression-free survival (rPFS) compared with abiraterone-prednisone in chemotherapy-naïve patients with metastatic castrate-resistant prostate cancer. Despite reporting prolonged rPFS, no improvement in overall survival or quality of life was found and significant difference in rPFS was lost when assessed by blinded independent central review (BICR). Here we examined whether informative censoring might explain the discrepancy between the outcomes.

Methods

Patient-level survival data were extracted and reconstructed using the semi-automated open-source tool WebPlotDigitizer and R statistical software. We utilized the reverse-Kapan-Meier method, in which the status indicator “event” and “censored” are flipped.

Results

We found that the rPFS of ACIS was associated with a significant censoring imbalance (reverse hazard ratio [HR] 1.32 [95% CI 1.08–1.61]; p=0.007), with 14% excess censoring in the intervention group. After performing sensitivity analysis, in which the balance in censoring between groups was partially restored, the difference in rPFS was lost (HR 0.86 [95% CI 0.73–1.00]; p=0·095), consistent with the non-significant results of the BICR assessment. To show this is unique to the trial we analyzed similar studies in mCRPC demonstrating anti-tumor activity and found that they were not associated with censoring imbalance.

Conclusions

When a study lacking survival benefit relies on surrogate endpoint to claim the presence of a clinical benefit, it is essential to exclude informative censoring. The measured effect in ACIS could be a product of the inherent limitations of survival analysis when excessive censoring exists.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Cocktail & Poster Display session Abstract session

108P - Establishing a multi-modal tissue preparation and imaging workflow to study heterogeneity in neuroblastoma tumors

Presentation Number
108P
Lecture Time
17:35 - 17:35
Speakers
  • Viktoria Humhal (Vienna, Austria)
Location
Hall Bordeaux, Paris Marriott Rive Gauche, Paris, France
Date
Mon, 06.03.2023
Time
17:15 - 18:15
Authors
  • Viktoria Humhal (Vienna, Austria)
  • Daria Lazic (Vienna, Austria)
  • Simon Gutwein (Vienna, Austria)
  • Eva Bozsaky (Vienna, Austria)
  • Fikret Rifatbegovic (Vienna, Austria)
  • Marie Bernkopf (Vienna, Austria)
  • Bernd Bodenmiller (Zurich, Switzerland)
  • Sabine Taschner-Mandl (Vienna, Austria)

Abstract

Background

Neuroblastoma (NB) is the most common solid tumor in infants and accounts for 15 % of childhood related cancer deaths. Clinical, genetic and phenotypic heterogeneity of NB hamper patient stratification, challenging therapeutic approaches. Methods to investigate tumor cell phenotypes and cells of the microenvironment in the primary tumors have so far mostly focused on transcriptomics or have been limited to only a few phenotypic markers on the protein and tissue level.

Methods

Multiplexed imaging techniques allow simultaneous or sequential detection of a high number of phenotypic and genetic markers. We aimed to establish a multi-modal tissue preparation and imaging workflow to integrate these multiplexed imaging techniques and thereby increase the confidence in tumor cell identification while studying tumor heterogeneity in NB at a high-dimensional scale. We combined three different imaging methods, i.e., 3-plex immuno-fluorescence staining (IF), 3-plex interphase fluorescence-in-situ-hybridization (iFISH) and 42-plex imaging mass cytometry (IMC).

Results

The multi-modal imaging method was established on NB tumor sections. First, all imaging techniques were optimized separately and subsequently in a combined fashion to generate sample preparation and staining protocols for combined IF and iFISH as well as for combined IF and IMC. Once the workflow was set, we assembled a patient cohort of 34 fresh frozen, high-risk NB tumors for analysis. For each sample we prepared two consecutive 4 μm thick cryosections, which were IF-stained according to the protocol to identify regions of interest for IMC and to align both tumor slices. Upon IF, one section was used for IMC to investigate the presence of different tumor cell phenotypes and immune infiltrates, while the other slice was used for iFISH to detect patient specific genetic aberrations.

Conclusions

The proposed workflow enables the analysis of the same tissue region with different imaging modalities, which would otherwise not be possible due to destructive properties or chemical incompatibilities of standard imaging protocols. This workflow will be of great benefit to reveal tumor heterogeneity and spatial relations of tumor cells with their microenvironment in NB.

Legal entity responsible for the study

S. Taschner-Mandl.

Funding

Austrian Science Fund (FWF).

Disclosure

All authors have declared no conflicts of interest.

Collapse