All times are listed in CET (Central European Time)

Displaying One Session

Foyer mezzanine Thu, 07.12.2023
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Location
Foyer mezzanine
Poster Display (ID 34) Poster Display

1P - Integrated Data Analysis within IMMUcan Identifies Prognostic Features of Early NSCLC

Presentation Number
1P
Lecture Time
12:00 - 12:00
Speakers
  • Daniel Schulz (Zurich, Switzerland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The advent of personalized medicine and ever-increasing numbers of available cancer drugs necessitate better prognostic and predictive biomarkers. In turn, this requires a deeper profiling and understanding of tumors and the tumor microenvironment (TME). To address this challenge the Integrated iMMUnoprofiling of large adaptive CANcer patient cohorts (IMMUcan) consortium collects samples and longitudinal clinical data from up to 3000 patients. All samples undergo bulk RNA-seq and whole exome sequencing (WES) for molecular profiling, whole slide multiplexed immunofluorescence (mIF) for broad and imaging mass cytometry (IMC) for detailed cellular profiling.

Methods

We analyzed a retrospective sub-cohort of non-small cell lung cancer (NSCLC) consisting of stage I-IIIA resected tumor samples from 192 patients collected between 2012 and 2018 under the EORTC-SPECTA protocol (NCT 02214134). Median follow up was 2.8 years and 42 patients died of the disease. Bulk RNA-seq and WES data were processed using standard tools. An image analysis workflow for mIF and IMC data paired with a tiling approached from ecology and spatial data analysis were used for feature extraction and subsequent multivariate cox proportional hazard modeling.

Results

The TME did not differ across clinical stage, smoking status or driver mutations. The TME of squamous cell carcinoma (LUSC) subtypes was enriched for immune suppressed cell types compared to adenocarcinoma (LUAD) subtypes in mIF and IMC data. Most samples (78%) contained B cell aggregates or tertiary lymphoid structures but their amount, size and location were not prognostic for overall survival (OS). In this cohort, not the density of CD8+ T cells but the variability of CD8+ T cell density across tissue sections, next to factors such as CD8+ T cell exclusion and broad T cell activation/exhaustion were strong prognostic factors for OS. Finally, we highlight results obtained from multi-modal data integration for the identification of the strongest prognostic features.

Conclusions

Our analyses reveal the potential of multi-modal data analysis and integration, and form the basis for analyses of all molecular and cellular profiling data from IMMUcan patients for biomarker discovery.

Clinical trial identification

NCT 02214134.

Legal entity responsible for the study

EORTC.

Funding

IMI2 JU grant agreement 821558, supported by EU’s Horizon 2020 and EFPIA.

Disclosure

M. Morfouace: Other, Personal, Member, employee: Merck Healthcare. T. Trefzer: Other, Personal, Member, employee: Bayer AG. H.S. Hong: Other, Personal, Member, employee: Merck Healthcare. T. Cufer: Financial Interests, Personal, Invited Speaker: MSD, Pfizer, Roche; Financial Interests, Personal, Advisory Board: Boehringer Ingelheim, Takeda; Financial Interests, Institutional, Local PI: MSD. A.C. Dingemans: Financial Interests, Institutional, Advisory Board: Roche, Sanofi, Amgen, Bayer, Boehringer Ingelheim; Financial Interests, Institutional, Invited Speaker: Takeda, Lilly, Jansen, Pfizer, AstraZeneca; Financial Interests, Institutional, Research Grant: Amgen; Financial Interests, Institutional, Local PI: Lilly, Amgen, Daiichi, JNJ, Mirati; Financial Interests, Institutional, Coordinating PI: Roche; Financial Interests, Institutional, Steering Committee Member: Roche; Non-Financial Interests, Personal, Other, Chair EORTC lung cancer group: EORTC; Non-Financial Interests, Personal, Member: IASCL, ASCO, AACR. B. Besse: Financial Interests, Institutional, Funding: 4D Pharma, AbbVie, Amgen, Aptitude Health, AstraZeneca, BeiGene, Blueprint Medicines, Boehringer Ingelheim, Celgene, Cergentis, Cristal Therapeutics, Daiichi Sankyo, Eli Lilly, GSK, Pfizer, Janssen, Onxeo, OSE Immunotherapeutics, Roche-Genentech, Sanofi, Takeda, Tolero Pharmaceuticals; Financial Interests, Institutional, Research Grant: Genzyme Corporation, Chugai pharmaceutical, Eisai, Inivata, Ipsen, Turning Point Therapeutics. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

3P - Exploratory efficacy analysis by smoking status in PD-L1 high patients in the phase III, non-small cell lung cancer (NSCLC) IMpower110 study

Presentation Number
3P
Lecture Time
12:00 - 12:00
Speakers
  • Luis Paz-Ares (Madrid, Spain)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Metastatic NSCLC is a leading cause of cancer death globally. Although implementation of immunotherapies has improved overall survival (OS), certain subgroups such as never smokers appear to derive less benefit from immunotherapy monotherapy. This analysis of the IMpower110 trial explores the efficacy of atezolizumab compared to chemotherapy in the PD-L1 high (TC≥50%/IC≥10%) patients by smoking status, as well as the achievement of long-term benefit (LTB) in the ever-smoking population.

Methods

We analyzed baseline characteristics and efficacy outcomes (OS and progression-free survival (PFS)) in the aforementioned population and clinically relevant subgroups. Kaplan-Meier curves and forest plots are presented. We characterized the LTB population, defined as patients who lived ≥2 years since randomization. We performed Chi-square tests to assess factors associated with LTB.

Results

In the PD-L1 high population the OS HR was 1.80 (0.71-4.52) for never smokers and 0.71 (0.49-1.04) for ever-smokers. The following results refer to the ever-smoking, PD-L1 high population, which consists of 181 patients (88.3% of all patients with high PD-L1 expression by SP142): The median follow-up was 30.6 months (mo). A median OS of 23.13 mo in the atezolizumab arm was observed compared to 11.04 mo with chemotherapy (HR= 0.71; 95% CI: 0.49-1.04; p=0.079). Likewise, median PFS favored atezolizumab with 8.34 vs 4.88 mo (HR= 0.56; 95% CI: 0.40-0.78; p<0.001). LTB was achieved by 46% (n=45) of patients who received atezolizumab and 33% (n=27) of patients who received chemotherapy. In the atezolizumab arm, clinical factors such as ECOG, liver metastases and race were significantly associated with LTB.

Conclusions

This exploratory analysis of the IMpower110 study reiterates the difference in the efficacy of monotherapy immunotherapy compared to chemotherapy based on smoking status. Atezolizumab is associated with better efficacy outcomes and provided LTB in a higher number of patients compared to chemotherapy in the ever-smoking PD-L1 high population.

Legal entity responsible for the study

Roche Pharma.

Funding

Roche Pharma.

Disclosure

L. Paz-Ares: Financial Interests, Personal, Advisory Board, Speaker fees: Roche, MSD, BMS, AZ, Lilly, PharmaMar, BeiGene, Daichii, Medscape, PER; Financial Interests, Personal, Advisory Board: Merck Serono, Pfizer, Bayer, Amgen, Janssen, GSK, Novartis, Takeda, Sanofi, Mirati; Financial Interests, Personal, Other, Board member: Genomica, Altum sequencing; Financial Interests, Personal, Member of Board of Directors, Board member: Stab Therapeutics; Financial Interests, Institutional, Coordinating PI: BMS, Janssen-Cilag international NV, Novartis, Roche, Sanofi, Tesaro, Alkermes, Lilly, Takeda, Pfizer, PharmaMar, Daiichi Sankyo, AstraZeneca, Merck Sharp & Dohme corp; Financial Interests, Personal, Coordinating PI: Amgen; Financial Interests, Personal, Other, Foundation Board Member: AECC; Financial Interests, Personal, Other, Member: AACR, ASCO, ESMO; Financial Interests, Personal, Other, President: ASEICA (Spanish Association of Cancer Research); Financial Interests, Personal, Other, Foundation president: ONCOSUR; Financial Interests, Personal, Other, member: Small Lung Cancer Group. M.R. García-Campelo: Financial Interests, Personal, Speaker, Consultant, Advisor, travel/accomodation/expenses: Roche, Merck, BMS, AstraZeneca, Lilly, Pfizer; Financial Interests, Personal, Speaker, Consultant, Advisor: PharmaMar, Bayer, AbbVie, GSK, Boehringer. M.D. Isla Casado: Financial Interests, Personal, Speaker, Consultant, Advisor, travel/accomodation/expenses: Roche, Merck, BMS, AstraZeneca, Lilly, Pfizer, Amgen, Boehringer; Financial Interests, Personal, Research Grant: BMS, Roche, AstraZeneca, Lilly; Financial Interests, Personal, Speaker, Consultant, Advisor: Bayer; Financial Interests, Personal, Advisory Role: AbbVie, Takeda. E. Carcereny: Financial Interests, Personal, Advisory Role, travel/accomodation/expenses: Roche, Bristol Myers Squibb, MSD, Takeda; Financial Interests, Personal, Advisory Role: AstraZeneca, Boehringer, Novartis; Financial Interests, Personal, Other, travel/accomodation/expenses: Pfizer. D. Rodriguez Abreu: Financial Interests, Personal, Speaker, Consultant, Advisor, travel/accomodation/expenses: Roche, Merck, BMS, AstraZeneca; Financial Interests, Personal, Advisory Role: Pfizer; Financial Interests, Personal, Speaker, Consultant, Advisor: Boehringer, Takeda. J.J. Garcia Gonzalez: Financial Interests, Personal, Speaker, Consultant, Advisor, travel/accomodation/expenses: Roche; Financial Interests, Personal, Advisory Role, travel/accomodation/expenses: Merck, BMS, Boehringer; Financial Interests, Personal, Advisory Role: AstraZeneca, Lilly, Pfizer. D. Perez Parente: N. Gonzalez Mancha, E. Vilas,P. Ruiz Gracia, A. Greco: Financial Interests, Institutional, Member, employee: Roche Pharma Spain. D.R. Spigel: Consulting or Advisory Role: Genentech/Roche (Inst), Novartis (Inst), Bristol Myers Squibb (Inst), AstraZeneca (Inst), GSK (Inst), Amgen (Inst), Ipsen (Inst), Jazz Pharmaceuticals (Inst), Sanofi/Aventis (Inst), Novocure (Inst), Regeneron (Inst), Lilly (Inst), AbbVie (Inst), BeiGene (Inst), Lyell (Inst), Monte Rosa Therapeutics (Inst). Research Funding: Genentech/Roche (Inst), Novartis (Inst), Celgene (Inst), Bristol Myers Squibb (Inst), Lilly (Inst), AstraZeneca (Inst), University of Texas Southwestern Medical Center-Simmons Cancer Center (Inst), Merck (Inst), G1 Therapeutics (Inst), Neon Therapeutics (Inst), Takeda (Inst), Nektar (Inst), Celldex (Inst), Clovis Oncology (Inst), Daiichi Sankyo (Inst), EMD Serono (Inst), Astellas Pharma (Inst), GRAIL (Inst), Transgene (Inst), Aeglea Biotherapeutics (Inst), Ipsen (Inst), BIND Therapeutics (Inst), Eisai (Inst), ImClone Systems (Inst), Immunogen (Inst), Janssen Oncology (Inst), MedImmune (Inst), Molecular Partners (Inst), Agios (Inst), GSK (Inst), Tesaro (Inst), Cyteir (Inst), Apollomics (Inst), Novocure (Inst), Elevation Oncology (Inst), Calithera Biosciences (Inst), Arcus Biosciences (Inst), Arrys Therapeutics (Inst), Bayer (Inst), BeiGene (Inst), BioNTech (Inst), Blueprint Medicines (Inst), Boehringer Ingelheim (Inst), Hutchison MediPharma (Inst), Incyte (Inst), Kronos Bio (Inst), Loxo Oncology (Inst), Macrogenics (Inst), Oncologie (Inst), Pfizer (Inst), PTC Therapeutics (Inst), PureTech (Inst), Razor Genomics (Inst), Repare Therapeutics (Inst), Rgenix (Inst), Tizona Therapeutics Inc (Inst), Verastem (Inst), AbbVie (Inst), Amgen (Inst), AnHeart Therapeutics (Inst), Ascendis Pharma (Inst), Asher Biotherapeutics (Inst), Ellipses Pharma (Inst), Endeavor (Inst), Erasca (Inst), Evelo Biosciences (Inst), Faeth Therapeutics (Inst), Foundation Bio (Inst), FujiFilm Pharmaceuticals (Inst), Gilead Sciences (Inst), Janux Therapeutics (Inst), Jazz Pharmaceuticals (Inst), Lyell Immunopharma (Inst), Millennium Pharmaceuticals (Inst), Moderna (Inst), Monte Rosa Therapeutics (Inst), Peloton Therapeutics (Inst), SeaGen (Inst), Shenzhen Chipscreen Biosciences (Inst), Strata Oncology (Inst), Stemline Therapeutics (Inst), Synthekine (Inst), Taiho (Inst), Tango Therapeutics (Inst), Tarveda (Inst), and Zai Laboratory (Inst). F. de Marinis: Consulting or Advisory Role: AstraZeneca, MSD Oncology, Bristol Myers Squibb, Roche/Genentech, Pfizer, Novartis, Takeda. J. Jassem: Consulting or Advisory Role: AstraZeneca, Amgen, Boehringer Ingelheim.

Collapse
Poster Display (ID 34) Poster Display

4P - Immune exoproteome, soluble proteome and immune-related gene expression profiles of anti-PD-1 therapy in stage IIIB/IV Non-Small Cell Lung Cancer: relevance of immunosuppressive factors

Presentation Number
4P
Lecture Time
12:00 - 12:00
Speakers
  • Paulo R. Santos (Coimbra, Portugal)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

This study intended to evaluate the immunological status of patients with stage IIIB/IV non-small cell lung cancer (NSCLC) comparing samples at diagnostics (DX), after chemotherapy (CT) and undergoing immune checkpoint blockade (ICB) immunotherapy.

Methods

Comprehensive immunophenotyping (>200 parameters defining T, B, NK, NKT-like cells, monocytes, dendritic cells, and myeloid-derived suppressor cells) was performed using peripheral blood from 44 stage IIIB/IV NSCLC patients. Immune related gene expression quantification for 103 targets, including 9 reference genes, was performed by RT-qPCR. Additionally, 103 immune-related plasmatic factors (cytokines, chemokines, growth factors and soluble immune checkpoints) were quantified using xMAP (Luminex®) technology.

Results

Most important finding consists in myeloid-derived suppressor cells (MDSCs) significant increase in all NSCLC patients compared to CTRL, particularly monocytic Myeloid-Derived Supressor Cells (M-MDSC). Similarly, type 2 myeloid dendritic cells (mDC2) demonstrated the same results for ICB patients. Particular T cells subsets (CD8, activated CD8, memory CD45RA+ and Tregs), transitional B cells, CD56bright NK cells and NKT-like cells were found significantly increased in ICB patients. Gene expression analysis revealed associations with genes involved in the T cell response, MHC class I expression, metabolism, adhesion molecules and vascular endothelial growth factors. Concerning the soluble factors associated with M-MDSC, a significant increase in VISTA/B7-H5 was observed in NSCLC patients under immunotherapy. A significant increase in IL-10 was observed, thus promoting the differentiation of Treg cells by cDC2. Using unsupervised machine-learning clustering analysis algorithms we were able to identify a global signature associated with checkpoint inhibitor blockade therapy and response to treatment (RECIST1.1).

Conclusions

In conclusion, this study suggests the importance of an extensive evaluation of cellular and soluble immune factors, which may prove useful in the selection and monitoring of patients undergoing immunotherapy in lung cancer.

Legal entity responsible for the study

The authors.

Funding

FCT - Portuguese Foundation for Science and Technology.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

5P - Blood immune-inflammatory dynamic unveils distinctive irAE features in ICI treated NSCLC

Presentation Number
5P
Lecture Time
12:00 - 12:00
Speakers
  • Giulia Mazzaschi (Parma, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

We tested whether the longitudinal monitoring of peripheral blood (PB) immune-inflammatory benchmarks might intercept immune-related adverse events (irAEs) onset and their impact on clinical outcome in ICI-treated NSCLC.

Methods

On PB collected at baseline (T0) and first disease assessment (T1) from 105 advanced NSCLC undergoing ICIs, comprehensive immune cell phenotyping (flow-cytometry), LDH, derived Neutrophil-to-Lymphocyte ratio (dNLR), LIPI and serum multiplex cytokine array were prospectively evaluated, including their delta (Δ) variation [(T1-T0)/T0*100]. irAEs type and grading (G) were defined according to CTCAE (Common Terminology Criteria in Adverse Events) v5.0. Correlations of irAEs with clinicopathological/PB parameters and survival outcome were statistically analyzed.

Results

irAEs occurred in 51 (48.6%) patients with 14.3% G3-4 incidence and involving the skin (36%), diarrhea/colitis (24%), liver (12%), thyroid (4%) and multiple sites (33.3%); median time-to-onset: 48-278 days; and leading to temporary or permanent ICI discontinuation in 11.4% of cases. irAEs were more frequent in females (58.3%) and absent in performance status (PS) 2 patients. Baseline PB parameters didn’t substantially differ according to irAE occurrence, except for higher IFNγ (P=0.014) and lower IL1β and IL6 (P=0.02) levels in irAEs cases. At T1, lower NLR, dNLR and LIPI characterized irAE patients, together with increased CD8+ cytotoxic (Perforin+, P=0.09) and proliferating (Ki67+, P=0.05) activity, higher IFNγ and TNFα and lower TGFβ1 levels compared to no-irAEs cases. Intriguingly, PB immune cells dynamic more closely reflected irAE severity as G3-4 cases displayed a sharp rise (Δpos) in cytotoxic NKs and CD8+GnzB+ lymphocytes (P=0.04) and a drop (Δneg) in CD4+CD25+FOX3high Tregs (P=0.07). Importantly, irAEs patients exhibited greater disease control rate (84% vs 65.1%, P=0.05) and significantly longer PFS (Median PFS=10.4 vs 5.4 mos, P=0.002, HR=0.47) and OS (Median OS=NR vs 11.3 mos, P<0.001, HR=0.34) vs no-irAEs NSCLC.

Conclusions

irAEs involve a distinctive cellular and humoral immune dynamic and might positively condition ICI efficacy.

Legal entity responsible for the study

University Hospital of Parma.

Funding

Associazione Italiana per la Ricerca sul Cancro-AIRC.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

6P - CD161+CD127+CD8+ T cells as a critical predictor of the efficacy of anti-PD-1 immunotherapy in diabetic patients with non-small cell lung cancer

Presentation Number
6P
Lecture Time
12:00 - 12:00
Speakers
  • Jingjing Qu (Hangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

PD-1-based immunotherapy is used for first- or second-line therapeutic regimens against NSCLC. Anti-PD-1 immunotherapy resistance occurs in diabetic patients with NSCLC. However, the characteristics of immune cell infiltration in such patients remain unexplored. Thus, we investigated the possible link between diabetes and immune cell infiltration in NSCLC.

Methods

We included patients (n = 437) with NSCLC treated with anti-PD-1 immunotherapy from the Zhejiang University School of Medicine. We analyzed the objective response rate, progression-free survival (PFS), overall survival (OS), T-cell infiltration, and peripheral blood immunological characteristics in diabetic and nondiabetic NSCLC patients. Differences in tumor microenvironment profiles were measured using CyTOF to elucidate the reasons for poor PFS and OS in diabetic NSCLC patients.

Results

Nondiabetic NSCLC patients had longer PFS than diabetic NSCLC patients (11.0 vs 7.0 months). The OS was 24.0 and 17.0 months in nondiabetic and diabetic NSCLC patients, respectively (P = 0.0065). Diabetic NSCLC patients had significantly lower CD8+T cell infiltration than nondiabetic NSCLC patients (P = 0.0227). Similarly, the anti-tumor effect of PD-1 blockade was decreased in diabetic mice with lung cancer. Additionally, compared with nondiabetic NSCLC patients before anti-PD1 treatment, CyTOF analysis showed that diabetic NSCLC patients had significantly more CD161+CD127+CD8+ T cells (P = 0.0071), and this trend continued after anti-PD1 treatment (P = 0.0393). Flow cytometry showed that diabetic NSCLC patients had a significantly higher CD161+CD127+CD8+T/CD8+ T cells ratio than nondiabetic NSCLC patients. Kaplan–Meier survival analysis showed that high levels of CD161+CD127+CD8+ T cells were positively related to the PFS in diabetic NSCLC patients.

Conclusions

We confirmed that diabetes is a risk factor for patients with NSCLC who undergo anti-PD-1 immunotherapy. CD161+CD127+CD8+ T cells in diabetic NSCLC patients serve as a key indicator of poor prognosis. Our findings provide a reference for understanding the characteristics of the tumor microenvironment in diabetic NSCLC patients.

Editorial acknowledgement

Prof. Nong Yang from the Lung Cancer and Gastroenterology Department, Hunan Cancer Hospital, and the Affiliated Tumor Hospital of Xiangya Medical School of Central South University for their technical support. Editage for English language editing.

Legal entity responsible for the study

Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China.

Funding

This work was supported by grants from the National Natural Science Foundation of China (No. 81802278), Natural Science Foundation of Zhejiang Province (No.LY23H160020), Medicine Health Technology Plan of Zhejiang Province, China (No. 2022KY150), and General Project of Education Department of Zhejiang Province (Y202043420).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

7P - A T-cell-derived circulating DNA as a biomarker for response to anti-PD(L)1 immunotherapy in advanced stage non-small cell lung cancer

Presentation Number
7P
Lecture Time
12:00 - 12:00
Speakers
  • Nuthchaya Mejun (Bangkok, Thailand)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

PD-L1 expression is a current predictive biomarker of anti-PD-(L)1 immunotherapy in advanced non-small cell lung cancer (NSCLC) but has many limitations as a stand-alone biomarker. Tumor infiltrating T-lymphocyte might be the source of T-cell-derived circulating DNA. We explored T-cirDNA as a potential biomarker of anti-PD (L)1 inhibitor and its correlation tumor-infiltrating lymphocytes (TILs).

Methods

The prospective cohort of 78 advanced NSCLC who received anti-PD-(L)1 immunotherapy as monotherapy or combination fashion. Pre-treatment plasma T-cirDNA target rearranged TCR-β CDR3 region were quantified using multiplex real-time PCR assay. FFPE Tissue from 40 patients were stained using CD8 (C8-144B-Dako) and FOXP3 (236A/E7- Abcam). We defined patients into undetectable, low (<1% ratio) and high (>1% ratio) T-cirDNA/cirDNA. Baseline clinicopathological characteristic and prognostic factors were integrated by cox-regression analysis. The correlation of T-cirDNA/cirDNA and CD8/FOXP3 TILs were explored.

Results

77% of participants had detectable T-cirDNA/cirDNA with a median of 0.02 [range 52.3] ngml-1. At median follow-up of 15.5 months, multivariate analysis revealed undetectable T-cirDNA/cirDNA and available subsequent treatment shown overall survival (OS) with the HR of 0.203 [95% CI 0.053-0.781, p-value 0.02] and 0.188 [95% CI 0.051-0.689 p-value 0.01] respectively. High level of T-cirDNA/cirDNA also correlated with better treatment outcome with the HR of 0.211 [95%Cl 0.055-0.815, p-value 0.02]. High level of T-cirDNA/cirDNA correlated with presence of intra-tumoral CD8 TIL 58.3% higher than low and undetectable T-cirDNA/cirDNA, 28.6% and 33.3% (p-value 0.17 and 0.19) respectively.

Conclusions

T-cell-derived circulating DNA shown paradoxical prognostic effect to anti-PD(L)1 treatment. The correlation with intra-tumoral CD8 TIL was demonstrated.

Legal entity responsible for the study

The authors.

Funding

National Research Council of Thailand (Grant number 347/2564).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

9P - Primary NSCLC patient-derived microtumors (PMTs) for clinical-relvant prediction of immunotherapy efficacy

Presentation Number
9P
Lecture Time
12:00 - 12:00
Speakers
  • Fabienne I. Nocera (Innsbruck, Austria)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Although the establishment of checkpoint inhibitors (CPIs) has shown great success in the treatment of cancer patients, a significant number of patients do not respond to it. This demonstrates the highly individualized nature of patient response to therapy, highlighting the need of a more tailored therapeutic approach. To this end, we have developed a functional 3D in vitro model, generated directly from primary tumor to evaluate personalized therapy options in a clinically relevant manner to improve outcome prediction.

Methods

Freshly resected NSCLC tumor material from 20 patients with different TPS Scores was mechanically and enzymatically digested to obtain a single cell suspension. PMTs were then generated by seeding the suspension into ultra-low attachment plates, preserving the original cell composition, which was characterized prior to drug administration after six days of tissue maturation. In addition to treating PMTs with CPIs, combinations with chemotherapies were applied. The dynamic drug response was monitored over 14 days, using bright field imaging. Further analysis of the drug response was performed by cytokine release as well as bulk RNA sequencing.

Results

To test immune modulators in vitro, it is necessary to preserve the immune compartment of the original tumor. FACS data confirmed, that during the maturation process of the NSLCL microtumors and beyond, the original cell compartments, including the various immune cell populations were largely preserved. Overall, the clinical routine categorized objective response rates reflected the clinical outcome distribution. In addition, immunotherapy treatment resulted in characteristic cytokine release and gene-pathway up/down regulation as a proof of concept to build a HUMAN LUNG CANCER RESPONSOME database linking a wide range of patient-specific functional outcomes with in-depth changes in individual pathways.

Conclusions

The present PMT model in combination with a clinically relevant drug response analysis is one of the first to show the typical immunotherapy-dependent alteration of TME as well as immunotherapy-induced cancer cell death in a scalable long-term in vitro assay.

Legal entity responsible for the study

A. Amann.

Funding

PreComb Therapeutics AG.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

10P - Lung Immune Prognostic Index (LIPI) Predicts Response to Immune Checkpoint Inhibitors in NSCLC Patients

Presentation Number
10P
Lecture Time
12:00 - 12:00
Speakers
  • Carlos D. Izquierdo Tolosa (Ciudad de Mexico, Mexico)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

11P - Decreased monocyte-to-lymphocyte ratio was associated with satisfied outcomes of first-line PD-1 inhibitors plus chemotherapy in stage IIIB-IV non-small cell lung cancer

Presentation Number
11P
Lecture Time
12:00 - 12:00
Speakers
  • Liang Zheng (Shanghai, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immunotherapy plus chemotherapy are widely used in patients with advanced non-small cell lung cancer (NSCLC) without oncogenic driver alterations. The monocyte-to-lymphocyte ratio (MLR) might predict thetreatment outcomes of ICI therapy in advanced NSCLC patients but has not yet been investigated. In addition, the cutoff of MLR is controversial. Therefore, the present study aimed to explore the associations between changes in MLR at the initial stage of treatment and clinical outcomes in advanced NSCLC patients receiving first-line PD-1 inhibitor plus chemotherapy.

Methods

The present study included 139 stage IIIB-IV NSCLC patients treated with first-line PD-1 inhibitor plus chemotherapy. The blood results were assessed 10 days before initiation of combination therapy (baseline) and before the third cycle of combined therapy (time point 2). Compared to altered MLR, neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio (PLR) in baseline and in time point 2, patients were divided into decreased MLR/NLR/PLR and increased MLR/NLR/PLR groups. The ORR, PFS, and the association with the changes in blood indicators were analyzed.

Results

Patients with decreased MLR had a significantly higher ORR in the univariate (P<0.001) and multivariate (P<0.001) analyses. Decreased MLR was significantly associated with prolonged PFS in the univariate (P=0.007) and multivariate (P=0.016) analyses. Decreased NLR exhibited high ORR (P=0.001) and prolonged PFS in univariate analysis (P=0.033). Decreased PLR was associated with high ORR in univariate (P<0.001) and multivariate (P=0.017) analyses. The subgroup analyses showed that decreased MLR was significantly associated with satisfactory outcomes in patients with all PD-L1 expressions.

Conclusions

Decreased MLR was associated with high ORR and long PFS and might have a potential predictive value in NSCLC treated with first-line PD-1 inhibitor plus chemotherapy. In addition, changes in MLR might have predictive value in all PD-L1-expressing populations. Decreased NLR and PLR also showed improved survival, suggesting that changes in NLR and PLR may be complementary to predicting prognosis.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

12P - Spatially preserved multi-region transcriptomic subtyping and biomarkers associated with long-term benefit with chemoimmunotherapy in extensive-stage small cell lung cancer (ES-SCLC)

Presentation Number
12P
Lecture Time
12:00 - 12:00
Speakers
  • Melina Peressini Álvarez (Madrid, Spain)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Transcriptomic subtyping holds promise for personalized therapy in SCLC, but intratumoral transcriptomic heterogeneity and its clinical significance remain poorly defined. In this study, we aimed to assess transcription factor-defined subtypes within multiple regions of intact tissues and identify gene sets associated with long-term chemoimmunotherapy benefit.

Methods

We assessed baseline FFPE tumors from 26 ES-SCLC patients enrolled in the CANTABRICO clinical trial (EudraCT: 2020-002328-35). We used GeoMxTM DSP to perform transcriptomic analysis from multiple intratumoral regions of interest (ROIs). We used an assay with +1800 genes (GeoMxTM CTA) plus custom-designed and subsequently validated mRNA probes targeting subtype-defining transcription factors.

Results

We profiled 121 ROIs from 26 tumors. Cluster analysis based on ASCL1, NEUROD1, POU2F3 and YAP1 showed that all samples clustered within 3 groups: SCLC-A (58 ROIs, 47.93 %), SCLC-N (37 ROIs, 30.58 %) and SCLC-Y/I (26 ROIs, 21.49 %). SCLC-P subtype was not identified in this cohort. Gene set enrichment analysis (GSEA) using linear mixed models revealed that SCLC-A subtype was enriched in cell cycle and DNA damage repair genes and showed repression of multiple gene sets associated with anti-tumor immune response. SCLC-Y/I subtype showed the opposite pattern. Six patients (23%) had tumors with co-existence of more than one subtype, not evident through morphological inspection. Predominant subtype or the presence of subtype heterogeneity were not associated with outcomes. Tumors from patients with sustained benefit (time to progression >= 12 months) had mRNA profiles characterized by upregulation of interferon gamma (IFNg)-related genes and down-regulation of gen sets involved in glycolysis and aberrant metabolism.

Conclusions

This study reveals substantial intratumoral subtype heterogeneity in SCLC. In this limited cohort, we did not observe outcome significance for transcriptional subtypes. Our findings related to long-term treatment benefit require validation in independent cohorts.

Clinical trial identification

Data from patients enrolled in the CANTABRICO study (phase IIIb clinical trial, EudraCT 2020-002328-35).

Legal entity responsible for the study

Fundación OncoSur.

Funding

AstraZeneca.

Disclosure

C. Martí Cubells: Financial Interests, Personal, Invited Speaker: MSD, BMS, AstraZeneca; Financial Interests, Personal, Other, Reimbursement of conference attendance: MSD, BMS, Merck. B. Massuti Sureda: Financial Interests, Personal, Advisory Board: AstraZeneca, BMS, Boehringer Ingelheim; Financial Interests, Personal, Invited Speaker: MSD, Roche; Financial Interests, Institutional, Funding, Research on Tumor Board: BMS; Non-Financial Interests, Personal, Principal Investigator: AstraZeneca, MSD; Non-Financial Interests, Personal, Advisory Board: ASCO; Non-Financial Interests, Personal, Member: IASLC, SEOM; Non-Financial Interests, Personal, Leadership Role: SLCG. M. Majem: Financial Interests, Personal, Advisory Board: Amgen, Roche, AstraZeneca, Takeda, Janssen, Cassen Recordati, BMS; Financial Interests, Personal, Invited Speaker: Amgen, Roche, AstraZeneca, Pfizer, Takeda, Helsinn; Financial Interests, Institutional, Funding: BMS, AstraZeneca, Roche. M.R. García-Campelo: Financial Interests, Personal, Advisory Board: MSD, BMS, Roche, Boehringer Ingelheim, Medscape Pfizer, Novartis, AstraZeneca, Lilly, Takeda, Janssen, Sanofi; Financial Interests, Personal, Invited Speaker: MSD, BMS, Roche, Boehringer Ingelheim, Medscape Pfizer, Novartis, AstraZeneca, Lilly, Takeda, Janssen, Sanofi; Financial Interests, Personal, Principal Investigator: MSD, BMS, Roche, Boehringer Ingelheim, Medscape Pfizer, Novartis, AstraZeneca, Lilly, Takeda, Janssen, Sanofi. P. Diz Tain: Financial Interests, Personal, Invited Speaker: BMS, AstraZeneca, Boehringer Ingelheim, Roche, MSD, Takeda, Pfizer, Amgen; Financial Interests, Personal, Advisory Role: BMS, AstraZeneca, Boehringer Ingelheim, Roche, MSD; Financial Interests, Personal, Training: BMS, AstraZeneca, Boehringer Ingelheim, Roche, MSD, Takeda, Pfizer; Financial Interests, Institutional, Principal Investigator: AstraZeneca, Roche, Mirati; Financial Interests, Personal, Advisory Board: Takeda. M.D. Isla Casado: Financial Interests, Personal, Speaker, Consultant, Advisor, Consultation and speaker honoraria: Amgen, AstraZeneca, Bayer, BMS, Boehringer Ingelheim, F. Hoffmann-La Roche, Janssen, MSD, Pfizer, Takeda; Financial Interests, Personal, Speaker, Consultant, Advisor, Consultation honoraria: Lilly, Merck, Sanofi; Financial Interests, Personal, Speaker, Consultant, Advisor, Speaker honoraria: Novartis; Financial Interests, Institutional, Other, Clinical trial: Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, BMS, Daiichi Sankyo, F. Hoffmann-La Roche, GSK, Janssen, Lilly, Merck, Mirati Therapeutics, MSD, Novartis, Pfizer, Sanofi; Financial Interests, Institutional, Research Grant: AstraZeneca, BMS, F. Hoffmann-La Roche, GSK; Other, Personal, Member of Board of Directors, Executive board member: Commission for the Approval of New Drugs, Regional Health Care Department (Spain). A. Moreno Paul: Financial Interests, Personal, Invited Speaker: Roche, MSD, Bristol; Financial Interests, Personal, Other, Reimbursement of conference attendance: Roche, MSD. E. Conde Gallego: Financial Interests, Personal, Research Funding: Roche, ThermoFisher; Financial Interests, Personal, Other, Honoraria: Roche, Lilly, Pfizer, AstraZeneca, Janssen. L. Paz-Ares: Financial Interests, Personal, Advisory Board, Speaker fees: Roche, MSD, BMS, AZ, Lilly, PharmaMar, BeiGene, Daichii, Medscape, PER; Financial Interests, Personal, Advisory Board: Merck Serono, Pfizer, Bayer, Amgen, Janssen, GSK, Novartis, Takeda, Sanofi, Mirati; Financial Interests, Personal, Other, Board member: Genomica, Altum sequencing; Financial Interests, Personal, Member of Board of Directors, Board member: Stab Therapeutics; Financial Interests, Institutional, Coordinating PI: Daiichi Sankyo, AstraZeneca, Merck Sharp & Dohme corp, BMS, Janssen-Cilag international NV, Novartis, Roche, Sanofi, Tesaro, Alkermes, Lilly, Takeda, Pfizer, PharmaMar; Financial Interests, Personal, Coordinating PI: Amgen; Financial Interests, Personal, Other, Member: AACR, ASCO, ESMO; Financial Interests, Personal, Other, Foundation Board Member: AECC; Financial Interests, Personal, Other, President: ASEICA (Spanish Association of Cancer Research); Financial Interests, Personal, Other, Foundation president: ONCOSUR; Financial Interests, Personal, Other, member: Small Lung Cancer Group. J. Zugazagoitia: Financial Interests, Personal, Speaker, Consultant, Advisor, Consultant: Sanofi, AstraZeneca, BMS, Roche, Pfizer, Novartis, Guardant Health; Financial Interests, Personal, Invited Speaker: Sanofi, Takeda, BMS, Pfizer, Roche, AstraZeneca, NanoString, Guardant Health; Financial Interests, Personal, Other, Travel honoraria: Sanofi, Takeda, BMS, Pfizer, Roche, AstraZeneca, NanoString; Financial Interests, Institutional, Research Funding: BMS, AstraZeneca, Roche. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

13P - Efficacy and biomarker exploration of Anlotinib plus Penpulimab for extensive stage Small cell lung cancer who failed from previous Platinum-Based Chemotherapy: Result from A phase II perspective trials

Presentation Number
13P
Lecture Time
12:00 - 12:00
Speakers
  • Yongchang Zhang (Changsha, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The phase II ALTER1202 trial demonstrated that patients who received anlotinib after failing at least two systemic chemotherapy regimens exhibited improved progression-free survival (PFS) and overall survival (OS) compared to the placebo group. In an effort to further enhance outcomes for small-cell lung cancer patients who have exhausted first-line platinum-based chemotherapy options, researchers are investigating the combination of anlotinib and penpulimab, a novel PD-1 inhibitor.

Methods

This open-label, single-arm, multi-center, phase II exploratory study enrolled patients with small-cell lung cancer who had previously undergone platinum-based chemotherapy. Patients received a combination therapy consisting of penpulimab (200mg every three weeks) and anlotinib (10mg daily), following a 2-week on and 1-week off schedule, until disease progression, unacceptable toxicity, or study discontinuation. The primary objective was to assess antitumor activity using the RECIST v1.1 criteria, focusing on the objective response rate (ORR). Secondary objectives included evaluating antitumor activity through the disease control rate (DCR), duration of response (DOR), PFS, OS, and assessing the safety and tolerability of this combination therapy.

Results

As of September 17, 2023, 38 patients were enrolled in the study and had received treatment. Of these 38 patients, 38 were evaluable using RECIST criteria, revealing an ORR of 26.30% (10 out of 38) and a DCR of 68.42% (26 out of 38). The median PFS was 4.36 months, with 6-month and 12-month PFS rates of 21.05% and 10.53%, respectively. The median DOR was 8.03 months, and the median OS was 15.71 months. Among the patients, 26 out of 38 (68.42%) experienced treatment-related adverse events, with 10 patients (26.32%) reporting grade 3 or higher adverse events. The most common adverse events related to anlotinib were hepatic injury, hematoxicity, and hypertension.

Conclusions

The combination of penpulimab and anlotinib showed promising clinical benefits and a favorable safety profile in small-cell lung cancer patients after platinum-based chemotherapy.

Clinical trial identification

NCT05001971.

Legal entity responsible for the study

Hunan Provincial Cancer Hospital.

Funding

Chia Tai Tianqing Pharmaceutical Group Co., Ltd.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

14P - Integrated modelling of T cell repertoires to identify clonotype signatures of ICI response

Presentation Number
14P
Lecture Time
12:00 - 12:00
Speakers
  • Juan Luis Melero (Barcelona, Spain)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immune checkpoint inhibitors (ICI) have shown efficacy in activating immune responses to tumours and in improving survival among cancer patients. However, highly heterogeneous patient responses highlight the need for predictive pipelines that leverage the immune cell dynamics associated with ICI as a biomarker.

Methods

The OS-T platform (Omniscope Inc.) was applied to deeply profile single T cell clonotypes from peripheral blood of colorectal cancer (CRC) and non-small cell lung cancer (NSCLC) patients before and after receiving ICI (e.g., anti-PD1 therapy). For an integrated immune repertoire analysis, we developed a novel computational framework for tracking and modelling TCR clonotype dynamics, combining three complementary approaches: (i) statistical noise model detecting differentially expanded clonotypes after treatment, (ii) sequence similarity and density-based clustering of sequences with inferred similar specificity, (iii) recombination model that computes generation probabilities of immune receptor sequences [Murugan, A., at al., 2012, 109(40), 16161-16166].

Results

We identified T cell clonotype clusters with treatment-related immune receptor sequences specific to or conserved across patients and cohorts. Such clonotype sequences allowed us to quantify treatment efficacy and to track patient response over time. Conserved clonotypes across patients enabled the identification of sequence signatures associated with patient response to ICI, potentially applicable for systematic efficacy tracking or patient stratification.

Conclusions

Our results suggest the combination of deep single-cell T-cell receptor profiling and receptor sequence modelling to efficiently identify candidate clonotypes for ICI response monitoring in CRC, NSCLC, and beyond. Moreover, combining such orthogonal approaches optimises the nomination of cancer-related T cell receptor sequences for targeted cell therapies and similar immune receptor-based strategies.

Legal entity responsible for the study

Omniscope Inc.

Funding

Omniscope Inc.

Disclosure

J. L. Melero, A. Mendizabal-Sasieta, U. Perron, D. Pravdyvets, N. Borcherding, B. Colom-Sanmartí, M. Grzelak, M. Soto, E. Planas Rigol: Financial Interests, Personal, Full or part-time Employment: Omniscope. J.C. Nieto: Financial Interests, Personal, Advisor of Omniscope. H. Heyn: Financial Interests, Personal, Co-founder of Omniscope, SAB member of Nanostring and MiRXES and consultant to Moderna and Singularity.

Collapse
Poster Display (ID 34) Poster Display

16P - Exosomal PD-L1 and lactate predict clinical outcomes of PD-1 blockade combined with chemotherapy in advanced-stage gastric and gastroesophageal junction adenocarcinoma

Presentation Number
16P
Lecture Time
12:00 - 12:00
Speakers
  • Yongshun Chen (Wuhan, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

First-line therapy with PD-1 blockade and chemotherapy is recommended for human epidermal growth factor receptor 2 (HER2)–negative patients with advanced gastric cancer (GC) or gastroesophageal junction cancer (GEJC), but the clinical outcomes of this approach is very variable. Predictive biomarkers are thus urgently required, here we investigated the predictive ability of exosomal PD-L1 and lactate levels in advanced GC or GEJC patients.

Methods

This prospective study (RENMIN-221) enrolled 68 advanced GC or GEJC patients receiving first-line PD-1 blockade and standard chemotherapy from January 2022 to March 2023. Exosomes were isolated by ultracentrifugation method from plasma prior to therapy. PD-L1 and lactate levels of the recovered exosomes were determined by enzyme-linked immunosorbent assay (ELISA), and peripheral blood was obtained before each cycle of treatment. The composition of peripheral CD3+ T cells, CD4+T cells, CD8+T cells, regulatory T (CD4+CD25+CD127low, Treg) cells, and the expression of their PD-1 were assessed by flow cytometry.

Results

Compared to the responders (CR+PR), exosomal PD-L1 (P=0.014) and lactate (P=0.012) was significantly higher than that of the non-responders (SD+PD) before the treatment. The receiver operating characteristic (ROC) curve showed that the combination of exosomal PD-L1 and lactate best distinguished responders from non-responders (area under the curve [AUC]: 0.787 vs. 0.748 vs. 0.706) among all the parameters tested. Combining predictor lower than -0.274 was associated with a better response to the therapy by ORR (82.1% vs. 30.0%, P<0.001), and mPFS (13.8 vs 5.5 months, P<0.001). Exosomal PD-L1 levels were signifcantly correlated with the frequency of CD8+T cells before treatment (P=0.025). And exosomal lactate was associated with less CD8+T cells and more Treg cells after treatment, while exosomal PD-L1was associated with more Treg cells and more PD-1+ Treg cells after treatment.

Conclusions

Exosomal PD-L1 and lactate levels could predict clinical outcomes, which provide accurate and convenient biomarkers in advanced GC or GEJC patients receiving first-line PD-1 blockade combined with 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
Poster Display (ID 34) Poster Display

17P - Spatial Characteristics Associated with the Chemo and Immuno-treatment Response of Gastric Cancer Revealed by Multi-omics Analysis

Presentation Number
17P
Lecture Time
12:00 - 12:00
Speakers
  • Gang Che (Hangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Gastric cancer is a multifaceted condition exhibiting varied responses to treatment, emphasizing the need for tailored therapeutic strategies. This study endeavors to elucidate the cellular interactions and molecular mechanisms that underlie the response to Sintilimab plus SOX (fluorouracil plus oxaliplatin) therapy among gastric cancer patients.

Methods

Single-cell sequencing and multiplex immunohistochemistry (mIHC) were employed to elucidate the spatial characteristics linked to the response of gastric cancer to chemo- and immuno- treatment. By integration of mIHC, feature extraction, and machine learning algorithms, we elucidated the intricate interactions between cellular populations and developed a Support Vector Machine (SVM) model for the prediction of treatment response.

Results

We initially discovered a significant correlation between apical membrane cells and resistance to fluorouracil and oxaliplatin, both crucial components of the treatment regimen. This prompted us to delve into the involvement of apical membrane cells in treatment response. Through a thorough examination of cell interactions, we noted substantial connections between apical membrane cells and resident macrophages. Further analysis of ligand-receptor interactions unveiled specific molecular associations, with TGFB1-HSPB1 and LTF-S100A14 interactions standing out, indicating potential signaling pathways implicated in treatment response. To forecast treatment response, we developed an SVM model integrating six markers (DUOX2, HSPB1, S100A14, C1QA, TGFB1, and LTF), which demonstrated outstanding predictive capacity, achieving high area under the curve (AUC) values of 0.93 in the exploration cohort and 0.84 in the validation cohort.

Conclusions

Our research underscored the importance of integrating multi-omics data alongside spatial information in predictive model, holding the promise of steering personalized therapeutic decisions and enhancing treatment efficacy.

Legal entity responsible for the study

The First Affiliated Hospital, School of Medicine, Zhejiang University.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

18P - Association of DNA methylation profiles with pathologic complete response in early triple negative breast cancer patients receiving neoadjuvant chemoimmunotherapy

Presentation Number
18P
Lecture Time
12:00 - 12:00
Speakers
  • Angelika M. Starzer (Vienna, Austria)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immunotherapy (IO) has become an important component of systemic treatment in early-stage triple-negative breast cancer (eTNBC). Considering toxicity and financial burden, identifying reliable biomarkers of response is urgently warranted. Therefore, we investigated blood cell DNA methylation profiles as biomarker for achieving a pathologic complete response (pCR) at surgery after neoadjuvant chemoimmunotherapy.

Methods

eTNBC patients receiving systemic chemoimmunotherapy before surgery at the Medical University of Vienna were included in this analysis. DNA from peripheral blood mononuclear cells (PBMCs) was isolated from whole blood samples collected at start of neoadjuvant therapy and methylation profiling was performed with Infinium Methylation EPIC microarrays. Routine histologic work-up from surgery determined a pCR or non-pCR according to pathologic standard procedures.

Results

Seven eTNBC patients (all women), median age of 63 years (range 40-83 years), were available for this analysis. All patients received neoadjuvant chemoimmunotherapy with pembrolizumab plus weekly paclitaxel/carboplatin followed by pembrolizumab plus epirubicin/cyclophosphamide. Three out of 7 (42.9%) patients achieved a pCR, while 4/7 (57.1%) had residual disease at surgery. Notably, when focusing on their methylation patterns, patients with pCR and non-pCR could be clearly segregated into two distinct groups. This distinction was based on the methylation profiles of the top 500 differentially methylated CpG sites.

Conclusions

Our results revealed differentially methylated genes between patients w/o pCR that might play a role in response to chemoimmunotherapy. Blood cell methylation profiles might serve as predictive biomarker for pCR in eTNBC and larger patient cohorts are needed to validate our findings.

Legal entity responsible for the study

The authors.

Funding

Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna.

Disclosure

A.M. Starzer: Financial Interests, Personal, Invited Speaker, Lecture honoraria: AstraZeneca; Financial Interests, Institutional, Research Grant, Industry partner of institutional Christian Doppler Laboratory: Roche; Non-Financial Interests, Personal, Member, National Oncology Society: OeGHO; Non-Financial Interests, Personal, Member, Oncology society of USA: ASCO; Other, Personal, Other, Travel support for conference participation: MSD, Lilly. M. Preusser: Financial Interests, Personal, Advisory Board: Bayer, Bristol Myers Squibb, Novartis, Gerson Lehrman, CMC Contrast, GSK, Mundipharma, Roche, BMJ Journals, MedMedia, AstraZeneca, AbbVie, Lilly, Medahead, Daiichi Sankyo, Sanofi, Merck Sharp & Dohme, Tocagen, Adastra, Gan & Lee Pharmaceuticals; Financial Interests, Institutional, Research Grant, Clinical Trials and research: Boehringer Ingelheim, Bristol Meyers Squibb, Roche, Daiichi Sankyo, Merck Sharp & Dohme, Novocure, GSK, AbbVie; Financial Interests, Institutional, Coordinating PI: PharmaMar; Non-Financial Interests, Personal, Leadership Role, Brain Tumor Group Chair (current): EORTC; Non-Financial Interests, Personal, Leadership Role, EANO Past-President (current): EANO; Non-Financial Interests, Personal, Other, Member Multi-Site Guideline Advisory Group: ASCO. R. Bartsch: Financial Interests, Personal, Invited Speaker: AstraZeneca, Seagen, Roche, Novartis, Eli-Lilly, Pierre Fabre, Daiichi, Gilead, MSD, Pfizer, Eisai, Gruenenthal; Financial Interests, Personal, Advisory Board: Daiichi, AstraZeneca, Roche, Novartis, Eli Lilly, Pierre Fabre, MSD, Gilead, Seagen, Eisai, Gruenenthal; Financial Interests, Institutional, Funding, Investigator Initiated Trial: Daiichi; Financial Interests, Institutional, Coordinating PI, Drug support for investigator initiated trial: MSD. A.S. Berghoff: Financial Interests, Personal, Research Funding: Daiichi Sankyo, Roche; Financial Interests, Personal, Speaker, Consultant, Advisor: Roche, Bristol Meyers Squibb, Merck, Daiichi Sankyo, AstraZeneca, CeCaVa; Financial Interests, Personal, Other, travel support: Roche; Financial Interests, Personal, Other, Travel support: Amgen, AbbVie. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

19P - The prognostic value of soluble CD73 in advanced triple-negative breast cancer: an exploratory analysis of the SYNERGY trial

Presentation Number
19P
Lecture Time
12:00 - 12:00
Speakers
  • Denis Zoë (Brussels, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Overexpression of CD73 in breast cancer is associated with immunosuppressive, pro-angiogenic tumor microenvironment and with poor patient clinical outcomes. Anti-CD73 antibodies are being investigated in combination with immune checkpoint inhibitors in clinical trials. Here, we investigated the prognostic value of the soluble form of CD73 (sCD73) in patients with advanced triple negative breast cancer (TNBC) treated with first-line durvalumab and chemotherapy, with or without anti-CD73 antibody oleclumab in the randomized, phase II SYNERGY trial (NCT03616886).

Methods

sCD73 levels were measured by ELISA in baseline plasma of 124 patients included in SYNERGY and in 24 healthy donors (HD). We explored association between sCD73 levels and baseline clinical characteristics, treatment response, and survival. Tumor-infiltrating lymphocytes (TILs) were assessed on H&E staining; PD-L1 was analyzed using immunohistochemistry.

Results

TNBC patients had significantly higher baseline sCD73 levels than HD (Mann-Whitney, p<0.001). High baseline levels of sCD73 were associated with liver metastases, high neutrophil-to-lymphocyte ratio (>5), and elevated CA 15-3 at baseline. sCD73 levels showed no association with clinical benefit at 24 weeks or with progression-free survival (PFS). Median overall survival (OS) was significantly shorter in patients with high baseline sCD73 levels compared to those with low sCD73 (18.8 vs. 26.4 months; log-rank, p=0.03). Higher TILs levels correlated with better PFS (log-rank, p=0.04) and OS (log-rank, p=0.03). High PD-L1 expression correlated with better PFS (log-rank, p=0.004). No association was observed between sCD73 levels and TILs or PD-L1 expression. Multivariate analysis (including treatment arm, TILs, PD-L1, sCD73 and disease presentation) confirmed the independent prognostic value of sCD73 (OS; HR=0.5; p=0.02).

Conclusions

In our analysis high baseline sCD73 levels were associated with negative prognostic clinical factors at baseline and with worse OS. The role of sCD73 as independent prognostic factor requires external validation.

Clinical trial identification

NCT03616886; August 6, 2018.

Legal entity responsible for the study

Institut Jules Bordet.

Funding

Institutional grant from AstraZeneca for the clinical trial. Translational analyses were funded by the Association Jules Bordet and the Fondation contre le Cancer (Belgium).

Disclosure

E. Agostinetto: Financial Interests, Personal, Speaker, Consultant, Advisor, Consultancy fee/honoraria: Eli Lilly , Sandoz, AstraZeneca; Financial Interests, Institutional, Research Grant: Gilead; Financial Interests, Personal, Other, Support to attend medical conferences (travel/accommodation/expenses): Novartis, Eli Lilly, Genetic, Istituto Gentili, Daiichi Sankyo. J. Stagg: Financial Interests, Personal, Advisory Board: Surface Oncology, Tarus Therapeutics, Domain Therapeutics, Iteos Therapeutics; Financial Interests, Personal, Stocks/Shares: Surface Oncology; Financial Interests, Institutional, Research Grant: Surface Oncology, Domain Therapeutics. C. Sotiriou: Financial Interests, Institutional, Advisory Board: Astellas, Vertex, Seattle Genetics, Amgen, INC, Merck & Co; Financial Interests, Personal, Advisory Board: Cepheid, Puma; Financial Interests, Personal, Invited Speaker: Eisai, Prime oncology, Teva; Financial Interests, Institutional, Other, Travel: Roche; Financial Interests, Institutional, Other, Internal speaker: Genentech; Financial Interests, Personal, Other, Regional speaker: Pfizer; Financial Interests, Institutional, Invited Speaker: Exact Sciences. L. Buisseret: Financial Interests, Institutional, Advisory Board: Domain Therapeutics; Financial Interests, Institutional, Other, Steering Committee: iTEOS Therapeutics; Financial Interests, Personal, Other, writing of clinical cases: Mirrors of Medicine; Financial Interests, Institutional, Other, Travel grant: Gilead; Financial Interests, Institutional, Research Grant, Research Grant for an investigator initiated trial: AstraZeneca; Non-Financial Interests, Personal, Principal Investigator: iTeos Therapeutics; Non-Financial Interests, Personal, Member: EORTC, BSMO. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

20P - Deciphering tumor microenvironment heterogeneity between primary tumor and metastases in high grade serous ovarian cancer (HGSC) to predict response to immunotherapy (IT): analyses from the NeoPembrOv/GINECO phase II trial

Presentation Number
20P
Lecture Time
12:00 - 12:00
Speakers
  • Laetitia Collet (Brussels, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Biomarkers of response to IT are lacking in HGSC and PD-L1 expression failed to predict benefit in the randomized NeoPembrOv trial assessing peri-operative chemotherapy with or without pembrolizumab. We hypothesize that heterogeneity of samples site may impact the biomarker evaluation.

Methods

Biopsy of intra-abdominal metastases (M) or primary tubo-ovaries (TO) were obtained before any treatment. PD-L1 expression (Ventana SP263) (N = 85), multiplex immunofluorescence (mIF) (N = 64) and RNAseq data (N = 57) were assessed. Differentially expressed genes (DEG) and gene set enrichment analysis (GSEA) were obtained with DESeq2. Survival was analyzed with Cox models.

Results

No major difference of immune infiltration was seen between TO and M samples (mIF data). TO were enriched in tumor cells and TPS ≥ 1 samples (Table). In M, CPS ≥1 was predictive of better PFS in the pembrolizumab arm as compared to chemotherapy alone arm (HR interaction = 0.26 CI 95% 0.07-0.93 P = 0.034). No significant benefit was seen with TPS and IC, nor in TO. Intra-tumoral CD8 T cell (iCD8) density was higher in the IC, TPS and CPS ≥ 1 subgroups and was correlated with PD-L1 expression in M but not in TO (Table). Higher iCD8 T cells density (P = 0.03) and an enrichment of immune hallmarks (Interferon alpha and gamma, allograft rejection, inflammatory response, P < 0.05) was seen in CPS ≥ 1 M vs CPS ≥ 1 TO. On the other hand, CPS ≥ 1 TO samples were enriched in proliferative pathways (MYC and E2F target, G2M checkpoint, oxidative phosphorylation, P < 0.05) compared to CPS ≥ 1 M.

Tubo-ovary n=29 Metastases, n=56 P-value
PD-L1 expression
Combined positive score (CPS) ≥ 1 79% 60% 0.08
Tumor positive score (TPS) ≥ 1 66% 34% 0.01
Immune cell score (IC) ≥ 1 45% 53% 0.48
Correlation between PD-L1 and iCD8 T cells (spearman ρ)
CPS ρ = 0.27 P = 0.2 ρ = 0.53 P < 0.001
TPS ρ = 0.34 P = 0.1 ρ = 0.50 P < 0.001
IC ρ = 0.02 P = 0.9 ρ = 0.49 P < 0.001

Conclusions

CPS ≥ 1 in M, but not in primitive tumor, is associated with pembrolizumab benefit, consistent with the increase of iCD8 T cells in PD-L1 positive M. This indicates that the site of tumor sampling influence prediction of potential efficacy of PDL1 inhibitors in HGSC.

Clinical trial identification

NCT03275506.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

M. Leheurteur: Financial Interests, Personal, Advisory Board: MSD. F. Selle: Financial Interests, Personal, Invited Speaker: AstraZeneca, MSD, GSK/Tesaro, Eisai; Financial Interests, Personal, Advisory Board: AstraZeneca, MSD, GSK/Tesaro. C. Sotiriou: Financial Interests, Institutional, Advisory Board: Astellas, Vertex, Seattle Genetics, Amgen, INC, Merck & Co; Financial Interests, Personal, Advisory Board: Cepheid, Puma; Financial Interests, Personal, Invited Speaker: Eisai, Prime oncology, Teva; Financial Interests, Institutional, Other, Travel: Roche; Financial Interests, Institutional, Other, Internal speaker: Genentech; Financial Interests, Personal, Other, Regional speaker: Pfizer; Financial Interests, Institutional, Invited Speaker: Exact Sciences. I.L. Ray-Coquard: Financial Interests, Personal, Advisory Board: Roche, GSK, AstraZeneca, Mersana, Deciphera, Amgen, Oxnea, Merck Sereno, Agenus, Novartis, Macrogenics, Clovis, EQRX, Adaptimmune, Eisai, SUTRO, BMS, Adaptimmune, Daiichi Sankyo; Financial Interests, Institutional, Other, COLIBRI translational research: BMS; Financial Interests, Institutional, Advisory Board, translational research NEOPREMBROV trial: MSD; Non-Financial Interests, Personal, Principal Investigator: PAOLA1; Non-Financial Interests, Personal, Other, President: GINECO. O. Le Saux: Financial Interests, Personal, Advisory Board: Novartis, MSD, GSK; Financial Interests, Personal, Invited Speaker: Lilly, AstraZeneca, Clovis; Financial Interests, Institutional, Trial Chair: Novartis, Hospira-Pfizer foundation, Astellas. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

21P - Mass cytometry reveals a population of exhausted CD8+ T cells associated with durvalumab/tremelimumab/vinorelbine efficacy in advanced cervical cancer (iMOVIE).

Presentation Number
21P
Lecture Time
12:00 - 12:00
Speakers
  • Alexandre Bertucci (Marseille, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immune checkpoint inhibitors (ICI) were recently developed in advanced cervical cancer (CC). However, the efficacy of ICI monotherapy is limited and predictive biomarkers of ICI efficacy are scarce. To improve ICI efficacy in advanced CC, a promising strategy is to combined anti PD(L)-1, anti CTLA-4 and chemotherapy. Our objective is to discover immune circulating biomarkers in patients with metastatic CC in ≥2nd line, treated with anti PD-L1 and anti CTLA-4 in combination with metronomic oral vinorelbine (MOV).

Methods

Three immune panels of up to 40 markers were developed to explore the immune landscape (T cells, NK cells and myeloid cells) of advanced cervical cancer in liquid biopsy. We used high-dimensional mass cytometry (CyTOF) in baseline blood samples from patients with advanced CC treated with durvalumab/tremelimumab and MOV. CyTOF datas were analyzed by machine-learning algorithms for dimensionality reduction, automated clustering and candidate prediction. Immune candidates were confirmed by manual gating. Maxstat and log-rank test were used to determine optimal cut-off and compare groups, respectively.

Results

From the cervix cohort of the phase 1/2 MOVIE multicentric prospective clinical trial (NCT03518606, sponsor UNICANCER France), 29 patients were analyzed. Median age was 56 years old. Compared to healthy donors, CC patients presented a decrease of CD4+CD127+TCF1+ T cells and an increase in CD8+TIGIT+ T cells. In CC patients treated in MOVIE trial (durvalumab, tremelimumab and MOV), clustering analyses, machine-learning analyses and manual gating confirmation identified a population of exhausted and senescent CD8+ T cells (CD8+CD45RA+CCR7-TIGIT+CD57+) associated with treatment efficacy. An optimal cut-off at 0.95% of CD45+ cells was determined. Patients with a high percentage of CD8+CD45RA+CCR7-TIGIT+CD57+ T cells had an improved clinical benefit rate (p=0.005), an improved PFS (HR=0.35, CI95, 0.13-0.95, p=0.013) and an improved OS (HR=0.23, CI95, 0.08-0.69, p<0.001).

Conclusions

This study identified a population of exhausted and senescent CD8+ T cells associated with response and survival with dual ICI and MOV in advanced cervical cancer.

Legal entity responsible for the study

Unicancer France.

Funding

GIRCI PACA.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

22P - Predictive value of Tertiary Lymphoid Structure in patients with mismatch repair deficient advanced/ recurrent endometrial cancer treated with Dostarlimab.

Presentation Number
22P
Lecture Time
12:00 - 12:00
Speakers
  • Maria Kfoury (Marseille, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

In the phase 1 GARNET study, Dostarlimab (Dmab) a PD-1 inhibitor, demonstrated clinically meaningful and durable antitumor activity in patients (pts) with advanced/ metastatic mismatch repair deficient endometrial cancer (Adv MMRd EC) after chemotherapy (chemo). Recently, the RUBY trial showed a significant improvement in progression-free survival (PFS) with Dmab plus chemo vs chemo alone, changing the standard of care in the first-line setting of Adv MMRd EC. To date, there are no validated predictive biomarkers of response to Dmab. There is growing evidence that tertiary lymphoid structure (TLS) could be predictive of response and prognostic in early EC. In our pilot study, we aim to determine the predictive value of TLS in Adv MMRd EC.

Methods

Pts with Adv MMRd EC treated with Dmab as a second-line after chemo in GARNET and the French early access program at Institut Paoli-Calmettes were included. Tissue samples from hysterectomy, endometrial or metastasis biopsies were collected. Microscopic examination was performed using paraffin-embedded tissue blocks on HES-stained sections. The presence of TLS was defined by histomorphological and phenotypic (immunohistochemistry) criterias: a dense nodule of more than 200 closely grouped lymphoid cells, with an occasional germinal center; a mixture of CD20+ B cells forming a follicle and surrounded by CD3+ T cells. Mature TLS were defined by the presence of follicular dendritic cells CD23+, CD21+, L1CAM+.

Results

14 pts were included from 12/2017-10/2021. One patient was excluded (tissue sample not evaluable). 11/13 (84%) pts had endometrioid EC. TLS were identified in 6/13 (46%) pts (TLS+) on HES sections. Immunohistochemistry staining confirmed the diagnosis of TLS, but did not detect further TLS. Median PFS : 9 months in pts with TLS vs 3.5 months in pts with no TLS.

Conclusions

Presence of TLS on HES sections seemed associated with better PFS in Adv MMRd EC pts treated with second-line Dmab. HES seemed sufficient to identify TLS, and could in routine practice, help better select MMRd pts that could benefit from Dmab alone. Larger cohorts are required to confirm our observations and explore predictive factors of reponse to Dmab in first-line.

Legal entity responsible for the study

Institut Paoli-Calmettes.

Funding

Has not received any funding.

Disclosure

M. Kfoury: Financial Interests, Personal, Invited Speaker: AZ, GSK. F. Rousseau: Financial Interests, Personal, Financially compensated role: Eisai; Financial Interests, Personal, Other: Viatris. R. Sabatier: Financial Interests, Personal, Advisory Board: GSK; Financial Interests, Personal, Invited Speaker: Eisai, Clovis Oncology; Financial Interests, Institutional, Research Grant: AstraZeneca; Non-Financial Interests, Personal, Other, Travel fees: MSD, Novartis; Non-Financial Interests, Personal, Other, Congress fees: GSK. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

23P - Circulating immune cells and activity of immune checkpoint inhibitors in metastatic renal cell carcinoma

Presentation Number
23P
Lecture Time
12:00 - 12:00
Speakers
  • Ronan Flippot (Villejuif, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immune checkpoint inhibitors (IO) combinations are standard in metastatic renal cell carcinoma (mRCC) but validated biomarkers are lacking to guide strategies towards either dual IO or IO plus tyrosine kinase inhibitors (TKI). Circulating immune cells (CIC) may inform antitumor immune response but have been mostly studied with IO monotherapy. We aimed at exploring the association between CIC and outcomes in mRCC patients treated with IO including combinations.

Methods

Phenotyping of peripheral blood mononuclear cells was performed by flow cytometry before systemic treatment in PREMIS (NCT03984318) and NIVOREN trials (NCT03013335). CIC included T cells (total CD8+, senescent CD8+, total CD4+, senescent CD4+) and B cells (naive, naive transitional, switched and unswitched memory, double negative, plasmablasts) subpopulations. We assessed associations between CIC and objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) across treatment subgroups. An exploratory analysis of CIC and tissue immune infiltrate was conducted.

Results

Among 112 patients, median age was 61 y.o; 75% had clear cell histology, 64% intermediate/poor risk disease. Regimens included IO-TKI in 28%, dual IO in 15%, IO monotherapy in 54%. A lower proportion of CD8+ T cells was associated with improved ORR in the overall population (p=0.008), and improved ORR (p=0.002) and PFS (HR 0.6, p=0.030) in patients treated with IO monotherapy or dual IO. No association between T cells and outcomes in patients treated with IO-TKI was observed. Senescent CD8+ T cells were highly associated with CD8+ T cell levels (r=0.51, p<0.001), and inversely associated with the presence of tertiary lymphoid structures or lymphoid aggregates within the tumor. Among B cells, plasmablasts were inversely associated with ORR (p=0.019) in the overall population with no impact on survival. Across treatment regimens, B cell subtypes were only predictive in IO monotherapy, with switched memory B cells associated with improved ORR (p=0.014) and PFS (HR 0.54, p=0.020).

Conclusions

Circulating CD8+ T cells and CD8+ senescence may inform resistance to dual IO but not IO-TKI. The role of B cells warrants further investigation. Longitudinal analyses are ongoing.

Clinical trial identification

NCT03013335, NCT03984318.

Legal entity responsible for the study

The authors.

Funding

French National Cancer Institute, ARC Foundation, Artur Foundation.

Disclosure

R. Flippot: Financial Interests, Institutional, Funding: Bayer; Financial Interests, Personal, Invited Speaker: Astellas, Janssen, MSD; Financial Interests, Personal, Non-financial benefits: BMS; Financial Interests, Personal, Speaker, Consultant, Advisor: Ipsen, Pfizer. B. Escudier: Financial Interests, Personal, Financially compensated role: Ipsen, Pfizer, Oncorena, Aveo, BMS, Ipsen; Financial Interests, Personal and Institutional, Research Funding: BMS. L. Albiges: Financial Interests, Institutional, Other, Consulting: Astellas, BMS, Eisai, Ipsen, Janssen, MSD, Novartis, Pfizer, Roche, Merck; Financial Interests, Personal, Other, Honoraria: Novartis; Non-Financial Interests, Personal, Principal Investigator, Clinical trial steering committee: Pfizer, BMS, AVEO, AstraZeneca, MSD; Non-Financial Interests, Personal, Principal Investigator: Ipsen; Non-Financial Interests, Personal, Other, Clinical trial steering committee: Roche, Exelixis; Non-Financial Interests, Personal, Member: ASCO; Non-Financial Interests, Personal, Other, Medical Steering Committee: Kidney Cancer Association; Non-Financial Interests, Personal, Other, Member of the Renal Cell Carcinoma Guidelines Panel: European Association of Urology (EAU). All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

24P - Chromosome 3p-related gene alterations (GA) as biomarkers for immunocombinations in metastatic renal cell carcinoma (mRCC): a hypothesis-generating analysis

Presentation Number
24P
Lecture Time
12:00 - 12:00
Speakers
  • Matteo Rosellini (Bologna, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Identifying biomarkers for mRCC is an unmet need in actual immunotherapy (IO) era. Available data regarding chromosome 3p-related genes (i.e., VHL, PBRM1, SETD2) as potential predictors for therapy response is conflicting. We describe the correlation of these GA with clinical outcomes in mRCC pts treated with IO/IO or IO/tyrosine kinase inhibitor (TKI).

Methods

We performed a single-center retrospective analysis on mRCC pts treated with first line IO/IO or IO/TKI. A multi-gene panel was used, allowing the amplification of 841 amplicons (54.93kb, human reference sequence hg19/GRCh37) in the coding sequences of the following genes: ATM, BAP1, KDM5C, MET, MTOR, NF2, PBRM1, PIK3CA, PTEN, SETD2, SMARCB1, TP53, TSC1, TSC2, VHL.

Results

30 mRCC pts undergoing IO/IO and IO/TKI were included. Of these, 18 had tumor tissue adequate for molecular analysis: 77.8% male, 22.2% female. Histology was 100% clear cell. IMDC risk was 50% intermediate, 33.4% good, 16.6% poor. First line therapy was 89% IO/TKI, 11% IO/IO. 83.3% pts (n=15) carried GA, among which 61.1% had a known pathogenic mutation (PAT). Most common GA included VHL in 44% (n=8; 7 PAT and 1 variant of unknown significance - VUS), PBRM1 in 44% (n=8; 5 PAT and 3 VUS) and SETD2 in 33% (n=6; 4 PAT and 2 VUS). With the limit of a small sample that did not allow proper statistical analyses, SETD2 mutated pts had lower median progression free (mPFS) and overall survival (mOS) than non-SETD2 mutated pts. Higher mPFS and mOS were shown with VHL or PBRM1 GA, especially in PBRM1+VHL mutated pts (Table). Of note, all PBRM1+VHL mutated pts underwent IO/TKI.

mPFS (months) mOS (months)
Mutated Non-mutated Mutated Non-mutated
SETD2 12 14.5 15 16
VHL 19 11 20 14
PBRM1 19 7.5 20 13
PBRM1+VHL 20 7.5 20.5 13.5

Conclusions

Our data shows a possible negative predictive role of SETD2 GA for IO-based therapy in RCC. Concomitant VHL and PBRM1 GA could act as a predictor for IO/TKI efficacy. Our hypothesis-generating analysis highlights the need of an integrated evaluation of these genes as promising biomarkers in RCC. Further larger studies are required.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

25P - Change in Splenic Volume as a Surrogate Marker for Immunotherapy Response in Patients with Advanced Urothelial and Renal Cell Carcinoma—Evaluation of a Novel Approach of Fully Automated Artificial Intelligence Based Splenic Segmentation

Presentation Number
25P
Lecture Time
12:00 - 12:00
Speakers
  • Gregor Duwe (Mainz, Germany)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

In the treatment of advanced urothelial (aUC) and renal cell carcinoma (aRCC), biomarkers such as PD-1 and PD-L1 are not robust prognostic markers for immunotherapy (IO) response. Previously, a significant association between IO and a change in splenic volume (SV) was described for several tumour entities. To the best of our knowledge, this study presents the first correlation of SV to IO in aUC and aRCC.

Methods

All patients with aUC (05/2017–10/2021) and aRCC (01/2012–05/2022) treated with IO at our academic centre were included. SV was measured at baseline, 3 and 9 months after initiation of IO using an in-house developed convolutional neural network-based spleen segmentation method. Uni- and multivariate Cox regression models for overall survival (OS) and progression-free survival (PFS) were used.

Results

In total, 35 patients with aUC and 30 patients with aRCC were included in the analysis. Lower SV at the three-month follow-up was significantly associated with improved OS in the aRCC group.

Conclusions

We describe a new, innovative artificial intelligence-based approach of a radiological surrogate marker for IO response in aUC and aRCC which presents a promising new predictive imaging marker. The data presented implicate improved OS with lower follow-up SV in patients with aRCC.

Legal entity responsible for the study

University Medical Center, Johannes-Gutenberg University University Medical Center, Johannes-Gutenberg University University Medical Center, Johannes-Gutenberg University University Medical Center of the Johannes Gutenberg-University Mainz.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

26P - Liquid biopsy as promising source of plasma extracellular vesicle biomarkers of response to Cabozantinib (CABO) plus Durvalumab (DURVA) in advanced urothelial carcinoma (UC) or non-UC variant histologies (VH) patients (the Phase 2 ARCADIA trial)

Presentation Number
26P
Lecture Time
12:00 - 12:00
Speakers
  • Veronica Huber (Milan, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

As components of the liquid biopsy, Extracellular Vesicles (EVs) have gained major interest as biomarkers of diagnosis, prognosis and prediction of response/resistance therapies. Here we investigated if plasma EV immune profile, size and concentration with plasma proteomics might discriminate responding from non-responding pts affected by UC or VH undergoing CABO+DURVA.

Methods

We evaluated 40 pts for their plasma EV profile at baseline and at the first reassessment. Baseline samples of 50 pts were evaluated for predictive potential. EVs were profiled using modified MACSplex technology coupled with flow cytometry and nanoparticle tracking analysis. Whole plasma was also searched for further indicators of response/resistance by proteomics (92 analytes, Immune-oncology panel, Olink).

Results

Preliminary analysis of EV markers measured in baseline samples evidenced no major association with response, although VH histology (n=13) showed a significant enrichment of CD1c and CD14 expressing EVs. A significant increase of immune markers were detected on-therapy time point (CD14,CD1c,CD2,CD20,CD8 and CD69; EV markers CD9, CD63 and CD81; platelet markers CD29, CD31 and CD326). This was also reflected by the global distribution of EV markers, which evidenced unexpectedly high levels of EVs exposing CD81 EV marker and CD8 already at baseline, with a further increase after therapy initiation. Finally, the dichotomization by response (responders n=21 vs non-responders n=19) highlighted that a statistically significant increase of immune EVs was detectable almost exclusively in responding.

Conclusions

Our preliminary results suggest that the early dynamics in plasma EVs may inform on the clinical outcome to DURVA plus CABO. The significant increase of EVs expressing immune markers measured at first reassessment in responding may derive from the activation of the immune system induced by therapy. The comprehensive analysis of EV profiles, size and concentration together with plasma proteomics could give rise to predictive/prognostic biomarkers of response, especially in pts with non-UC VHs.

Clinical trial identification

NCT03824691.

Legal entity responsible for the study

Fondazione IRCCS Istituto Nazionale dei Tumori, Milan.

Funding

AIRC IG-25078, NET-2016-02361632 from Italian Health Ministry.

Disclosure

P. Giannatempo: Financial Interests, Personal, Advisory Board: Pfizer. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

27P - Peripheral biomarker analysis in patients with advanced urothelial carcinoma (UC) after platinum chemotherapy treated with Cabozantinib (CABO) plus Durvalumab (DURVA): preliminary analysis from the Phase 2 ARCADIA trial.

Presentation Number
27P
Lecture Time
12:00 - 12:00
Speakers
  • Francesco Sgambelluri (Milan, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Preliminary results of the interim analysis of the ARCADIA trial have shown that combining multitargeted receptor tyrosine kinase inhibitor CABO with the checkpoint inhibitor DURVA has promising activity in patients (pts) affected by UC after chemotherapy. To identify peripheral blood biomarkers potentially associated with clinical response, we carried out a quantitative profiling of innate and adaptive immune subsets from a subset of treated pts.

Methods

From 09/2019 and 08/2023 blood samples from 65 pts (45= pure UC, 20=VHs) were collected at baseline and before the third treatment cycle. Absolute cell counts for 29 innate and adaptive immune subsets were determined by multiparametric flow cytometry.

Results

In pre-therapy samples a significant higher counts for all CD45+ leukocytes were found in non-responders compared to responders (p=0.0053, Mann Whitney test, n= 27 patients). This was, explained by higher counts for CD16+CD15+ neutrophils (p=0.0005), classical CD14+CD16- (p=0.0119) and CD14++CD16+ intermediate (p=0.0186) monocytes, CD56dim CD16+ NK cells (p=0.0365) and Lin-HLA-DR-/LoCD33+CD14+CD15- M-MDSCs (p=0.0281). At baseline, higher neutrophils counts were associated with worse PFS (p=0.0117, log rank test), while higher eosinophils counts were associated with improved PFS (p=0.0158). Compared to responders, non-responders underwent a significant reduction in post-treatment counts for all CD45+ leukocytes (p=0,0024, Wilcoxon matched pair test), due to reduction of neutrophils (p=0.0068), CD15+CD16- eosinophils (p=0.0068), CD3+ T cells (p=0.0425) CD19+ B cells (p=0.0068), classical monocytes (p=0.0034), activated (HLA-DR+) CD56dim CD16- NK cells (p=0.0068), M-MDSCs (p=0.0161), Lin- HLA-DR-/Lo CD33+ CD14- CD15+ PMN-MDSCs (p=0.001), Lin- HLA-DR+ CD33- pDCs (p=0.0269) and Lin- HLA-DR+ CD33+ mDCs (p=0.0005).

Conclusions

These preliminary findings suggest that high baseline counts for granulocytes, monocytes and MDSCs may negatively impact on response in pts treated with CABO+DURVA. Moreover, baseline neutrophils and eosinophils counts show opposite impact on PFS.

Clinical trial identification

NCT03824691.

Legal entity responsible for the study

Fondazione IRCCS Istituto Nazionale Tumori.

Funding

NET-2016-02361632 from Italian Health Ministry to A. Anichini.

Disclosure

P. Giannatempo: Financial Interests, Personal, Advisory Board: Pfizer. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

28P - 3-year follow-up analysis of disease-free survival in CheckMate 274 by PD-L1 expression using tumor cell and combined positive scoring algorithms

Presentation Number
28P
Lecture Time
12:00 - 12:00
Speakers
  • Frank Stenner-Liewen (Basel, Switzerland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

CheckMate 274 met the primary endpoints of improved disease-free survival (DFS) with nivolumab (NIVO) v placebo (PBO) both in the intent-to-treat (ITT) population and in patients (pts) with programmed death-ligand 1 (PD-L1) expression ≥1% assessed by tumor cell score (TC). We report updated analysis of DFS by PD-L1 expression as assessed by both TC and combined positive score (CPS; measuring PD-L1 on both tumor and immune cells) with minimum follow-up in the ITT population of 31.6 months.

Methods

CheckMate 274 (NCT02632409) is a phase 3, randomized, double-blind, trial of adjuvant NIVO v PBO in pts with muscle-invasive urothelial carcinoma who are at high risk of recurrence after radical resection. Pts were randomized 1:1 to NIVO 240mg or PBO every 2 weeks intravenously for ≤1 year. Primary endpoints are DFS in the ITT population and in pts with TC PD-L1≥1%. CPS was determined retrospectively from the previously stained immunohistochemistry slides. This analysis only included pts with PD-L1 quantifiable by both CPS and TC.

Results

630 pts had quantifiable PD-L1 by TC and CPS; 250 (40%) had TC≥1% (NIVO, n=125; PBO, n=125), 380 (60%) had TC<1% (NIVO, n=191; PBO, n=189), 558 (89%) had CPS≥1 (NIVO, n=282; PBO, n=276), and 72 (11%) had CPS<1 (NIVO, n=34; PBO, n=38). In pts with TC<1%, 81% (n=309) had CPS≥1. Table shows the number of pts and DFS outcomes in pts with TC≥1% and CPS≥1. In pts with TC<1% who also had CPS≥1, median DFS (95% CI) was 19.2 (16.1-25.6) months with NIVO vs. 10.4 (8.2-19.4) months with PBO; HR for NIVO v PBO in these pts was 0.79 (95% CI, 0.60-1.05).

Median DFS (95% CI), mo HR (95% CI) DFS probability, % 24-mo DFS probability, % 33-mo
TC ≥ 1% NIVO (n=125) 52.6 (25.8–NE) 0.48 (0.34–0.69) 62 58
PBO (n=125) 8.4 (5.6–17.9) 36 32
CPS ≥ 1 NIVO (n=282) 25.6 (19.3–41.8) 0.64 (0.51–0.80) 51 47
PBO (n=276) 8.5 (7.8–15.2) 38 35

HR, hazard ratio; mo, months; NE, not estimable.

Conclusions

With extended follow-up, this exploratory analysis of PD-L1 expression by CPS showed that most pts with TC<1% had CPS ≥1. In pts with TC<1% and CPS≥1, median DFS with NIVO was nearly double that with PBO. These results support the interpretation that most pts with TC<1% also benefit from adjuvant NIVO.

Clinical trial identification

CheckMate 274 (NCT02632409).

Legal entity responsible for the study

BMS.

Funding

BMS.

Disclosure

F. Stenner-Liewen: Consulting or Advisoy role: BMS/Roche, Ipsen, MSD, Novartis, Pfizer. Research funding: BMS GmbH & Co. KG, Takeda. Travel, accommodations, expenses: BMS GmbH & Co. KG, Roche. M. Galsky: Consulting or Advisoy role: Biomotiv, Janssen, Dendreon, Merck, GSK, Lilly, Astellas Pharma, Genentech, BMS, Novartis, Pfizer, EMD Serono, AstaZeneca, Seagen, Incyte, Aileron Therapeutics, Dracen, Inovio Pharmaceuticals, NuMab, Dragongly Therapeutics, Basilea, UroGen Pharma, Infinity Pharmaceuticals, Gilead Sciences, Silverback Therapeutics, AbbVie; Patents, Royalties, Other Intellectual Property: Methods and compositions for treating cancer and related methods. Mount Sinai School of Medicine July 2012, Application number: 20120322792. Stock and Other Ownership Interests: Rappta Therapeutics. Research funding (my institution): AstraZeneca, Bristol Myers Squibb, Dendreon, Genentech/Roche, Janssen Oncology, Merck, Novartis. D. Bajorin: Consulting or Advisoy role: Merck, Dragonfly Therapeutics, Fidia Farmaceutici S.p.A., Bristol Myers Squibb Foundation; Travel, accommodations, expenses: Merck; Research funding (my institution): Novartis, Merck, BMS, AstraZeneca, Astellas Pharma, Seattle Genetics/Astellas. J. Witjes: Consulting or Advisoy role: Nucleix, BMS, MSD, Ipsen, Sanofi/Aventis, Janssen Oncology, Oncodiag, BeiGene, Ferring, AstraZeneca, Asieris Pharmaceuticals, Photocure; Honoraria: Astellas Pharma, BeiGene, Ferring, AstraZeneca, Janssen, MSD, BMS/Pfizer. J. Gschwend: Consulting or Advisoy role: Janssen-Cilag, Bayer Schering Pharma, BMS; Honoraria: Janssen-Cilag, Bayer Schering Pharma, BMS. Y. Tomita: Consulting or Advisoy role: Eisai, MSD Onco Pharmaceutical, Taiho Pharmaceutical; Honoraria: Astellas Pharma, BMS Japan, Chugai Pharma, Ono Pharmaceutical, Takeda, Merck, Pfizer, MSD; Research funding (my institution): Astellas Pharma, AstraZeneca, Chugai Pharma, Eisai, MSD, Ono Pharmaceutical, Pfizer, Takeda. F. Nasroulah, M. Askelson, J.M. David: Financial Interests, Institutional, Full or part-time Employment: BMS; Stock and Other Ownership Interests: BMS. B. Perez Valderrama: Consulting or Advisoy role: BMS/Medarex, MSD Oncology, Astellas Pharma, Novartis, Bayer, Advanced Accelerator Applications/Novartis; Travel, accommodations, expenses: Merck/Pfizer; Honoraria: BMS/Medarex, Roche, EUSA Pharma, Pfizer, Astellas Pharma, Bayer, Merck/Pfizer, Merck. M-O. Grimm: Consulting or Advisoy role: AstraZeneca, BMS, Ipsen, MSD, Pfizer, EUSA Pharma, Merck Serono, Takeda, Eisai, Bayer Vital, Janssen Cilag, Gilead Sciences, Novartis; Honoraria: AstraZeneca, BMS, MSD, Pfizer, Ipsen, Merck Serono, EUSA Pharma, Janssen Cilag; Travel, accommodations, expenses: BMS, Merck Serono, MSD, Janssen Cilag, Ipsen, AstraZeneca; Research funding (my institution): BMS, Intutive Surgical, Bayer Vital. L. Appleman: Consulting or Advisoy role: AADi; Travel, accommodations, expenses: BMS, Merck Serono, MSD, Janssen Cilag, Ipsen, AstraZeneca; Other relationship: Pfizer; Research funding (my institution): Pfizer, Exelixis, BMS, Astellas Pharma, Acerta Pharma, Novartis, Bayer, Agensys, Merck, Genentech/Roche, Tokai Pharmaceuticals, AVEO, Peloton Therapeutics, Calithera Bioscences, Seattle Genetics, Inovio Pharmaceuticals, Eisai, Lilly, Amgen, Surgace Oncology, BioNTech AG. G. Gravis: Research funding (my institution): BMS; Payment or honoraria for speakers’ bureaus (institutional): Janssen Oncology, Ipsen, BMS, Amgen, Sanofi/Aventis, MSD Oncology, Astellas Pharma; Support for attending meetings and/or travel: Janssen Oncology, BMS, Astellas Pharma, Pfizer, Ipsen, Sanofi, AstraZeneca; Participation on a Data Safety Monitoring Board or Advisory Board (institutional): BMS, Janssen, Ipsen, Sanofi/Aventis, MSD Oncology, Pfizer, Bayer, AstraZeneca. A. Necchi: Consulting or Advisoy role: Merck Sharp & Dohme, Roche, Bayer, AstraZeneca, Clovis Oncology, Janssen, Incyte, Seattle Genetics/Astellas, BMS, Rainier Therapeutics, GSK, Basilea Pharmaceutical, Catalym; Travel, accommodations, expenses: Roche, Merck Sharp & Donhme, AstraZeneca, Janssen, Rainier Therapeutics, Pfizer; Employment, other relationship and stock and other ownership interest: Bayer (An Immediate Family Member); Honoraria: Roche, Merck, AstraZeneca, Janssen, Foundation Medicine, BMS, Astellas Pharma; Research funding: Ipsen, Gilead Sciences Research funding (institution): Merck Sharp & Dohme, AstraZeneca. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

29P - The Impact of Body Mass Index on Survival Endpoints among Patients with Metastatic Urothelial Carcinoma Undergoing Treatment with Immune Checkpoint Inhibitors: A Real-World Multicenter Analysis

Presentation Number
29P
Lecture Time
12:00 - 12:00
Speakers
  • Yu-Li Su (Kaohsiung City, Taiwan)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Studies on the correlation between high body mass index (BMI) and extended survival among patients receiving immune checkpoint inhibitors (ICIs) have been made, although findings have shown variability. Our research explored the phenomenon of the “obesity paradox” in patients with metastatic urothelial carcinoma (mUC) undergoing treatment with ICIs.

Methods

We conducted a retrospective analysis of patients diagnosed with mUC who received a minimum of one cycle of ICI treatment at two medical centers in Taiwan from September 2015 to January 2023. Features of patients’ clinicopathologic factors, including age, sex, primary or metastatic location, treatment line and BMI were examined. The primary outcome were overall survival (OS) and progression-free survival (PFS), which were assessed utilizing the Kaplan–Meier method. We employed the Cox regression model to adjust for multiple covariates.

Results

A total of 215 patients were included, with 128 (59.5%) being male, and the median age was 70 years. In the obese group (BMI ≥ 25), patients demonstrated significantly better median OS compared to the non-obese group (BMI < 25) (21.9 vs. 8.3 months; p = 0.021). However, there was no significant difference in median PFS between the high and low BMI groups (4.7 vs. 2.8 months; p = 0.16). Post-hoc subgroup revealed a survival benefit from ICI treatment in male patients within the BMI ≥ 25 group (HR 0.49, 95% CI 0.30-0.81, p = 0.005).

Conclusions

Based on real-world data from the Asia-Pacific region, there appears to be a correlation between obesity and prolonged OS in patients receiving ICI treatment for mUC.

Legal entity responsible for the study

The authors.

Funding

Chang Gung Memorial Hospital, Kaohsiung, Taiwan.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

30P - CD4+ T cells within the tumor microenvironment are an independent predictor of recurrence, but do not improve the performance of a predictive model in oral squamous cell carcinoma

Presentation Number
30P
Lecture Time
12:00 - 12:00
Speakers
  • Sangeeta K. Bisheshar (Groningen, Netherlands)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The recurrence risk of patients diagnosed with oral squamous cell carcinoma (OSCC) is currently insufficiently predicted by conventional clinicopathological characteristics. This study investigates the added predictive value of the tumor microenvironment immune cell composition (TMICC) in addition to conventional clinicopathologic characteristics.

Methods

Primary tumor samples of 290 OSCC patients were immunohistochemically stained for TMICC, consisting of CD4, CD8, CD20, CD68, CD163, CD57, FoxP3 and Programmed cell Death Ligand 1 (PD-L1). Additionally, clinicopathologic characteristics were obtained from patients’ medical files. Predictive models were trained and validated by conducting Least Absolute Shrinkage and Selection Operator (LASSO) regression analyses with cross-validation. To quantify the added predictive power of TMICC within models, receiver operating characteristic (ROC) analyses were used. Recurrence was defined to include locoregional recurrences and distant metastasis.

Results

Recurrence occurred in 74 (25.5%) of patients. Conventional clinicopathologic characteristics consisting of tumor localization, pathological T-stage, pathological N-stage, extracapsular spread, resection margin, differentiation grade, perineural invasion, lymph angioinvasion and treatment modality, were used to build a LASSO logistic regression-based predictive model. Addition of TMICC to the model resulted in a comparable AUC of respectively 0.79 (± 0.01) and 0.76 (± 0.1) in the training and test sets. The model showed that high numbers of CD4+ T cells and absence of lymph node metastasis, extracapsular spread and perineural invasion protected against recurrence. Positive surgical margins and reception of adjuvant treatment increased the risk for recurrence.

Conclusions

CD4+ T cells are the strongest predictor within the TMICC, however, addition to conventional clinicopathologic characteristics does not improve the performance of a predictive model for recurrence in OSCC treated with curative intent.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

E. Schuuring: Financial Interests, Institutional, Advisory Board, Unrelated to the publication of this article: AstraZeneca, Bayer, BMS, Roche, Pfizer, Novartis, Amgen, Lilly, BioCartis, Illumina, Astellas Pharma, Agena Biosciences, MSD/Merck, CC Diagnostics, Janssen Cilag (Johnson&Johnson), Diaceutics; Financial Interests, Institutional, Invited Speaker, Unrelated to the publication of this article: Bio-Rad, Abbott, Roche, Biocartis, Illumina, Pfizer, AstraZeneca, Agena Biosciences; Financial Interests, Institutional, Research Grant, Unrelated to the publication of this article: Pfizer, Biocartis, AstraZeneca, Bayer/Invitae, Biocartis, Cancer-ID, BMS, Bio-Rad, Roche, Agena Biosciences, Promega, Qiagen, CC Diagnostics, Boehringer Ingelheim. B. van der Vegt: Financial Interests, Institutional, Speaker, Consultant, Advisor, Unrelated to the publication of this article: Visiopharm; Financial Interests, Institutional, Advisory Board, Unrelated to the publication of this article: Phillips, AstraZeneca, Daiichi Sankyo; Financial Interests, Institutional, Invited Speaker, Unrelated to the publication of this article: Diaceutics, MSD, MSD. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

31P - Characterization of pre-exhausted / exhausted state of CD8+ T cells in HRAS mutant head and neck carcinomas (HNSCCs). Implications for response to immune checkpoint blockade (ICB).

Presentation Number
31P
Lecture Time
12:00 - 12:00
Speakers
  • Ioannis Kotsantis (Haidari, Greece)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

HRAS mutations have been found in 6% of HNSCCs. T cell exhaustion, defined as dysfunctional T cells stimulated by continuous antigen exposure. To guide immunotherapeutic approaches, we sought to assess the immune landscape of HRAS-mutant tumors by investigating the subpopulations of pre-exhausted and exhausted T cells.

Methods

We found 10 cases of HRAS mutant tumors and 39 cases of HRAS wild-type (WT) tumors. We sought to characterize exhausted CD8+ T cell subpopulations by measuring the expression of T-cell Factor-1 (TCF-1) and Programmed Cell Death-1 (PD-1) in the tumor's center (C) and periphery (P). Multiplex immunohistochemistry (IHC) was performed in FFPE tissue sections using three primary antibodies (PD1-CD8-TCF1/7), followed by analysis of a manually trained algorithm in Qupath software.

Results

HRAS mutant tumors present numerically higher numbers of total immune cells both in the C and the P than HRAS-WT tumors, that reached statistical significance only in the P (5123.17/mm2vs. 3527.93/mm2, p=0.002). In addition, the density of CD8+ T Cells was increased in both the C (694.10/mm2 vs. 356.02/mm2) and the P (851.10/mm2 vs. 333.30/mm2) in HRAS mutant tumors. Importantly, the percentage of pre-exhausted CD8 (+) T Cells was elevated in the P of HRAS mutant tumors (384.67/mm2vs. 51.18/mm2, p=0.040), indicating a possible association of response to ICB, since pre-exhausted T cells mediate the proliferative response to immunotherapy. On the contrary, exhausted T cells, defined as PD-1(+)TCF-1 (-) were more abundant in the C of HRAS mutant tumors compared to WT (13.77% vs. 2.67% of total CD8+ cells, p=0.022). Moreover, increased area occupied by CD11c+ dendritic cells and numbers of CD8+ T cells were found in regional lymph nodes from HRAS mutant patients (79.25% vs 38.80%, p=0.036 and 10160.43 vs 4438.28/mm2, p=0.036, respectively), consistent with data showing that maintenance of TCF1 by intratumoral T cells requires continuous migration from draining lymph nodes.

Conclusions

Pre-exhausted PD-1(+)TCF-1(+) T cells are significantly increased at the periphery of HRAS mutant tumors, suggesting a potential sensitivity of these tumors to ICB.

Legal entity responsible for the study

The authors.

Funding

Kura Oncology.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

32P - Tumor-agnostic plasma assay for circulating tumor DNA predicts outcome in recurrent and/or metastatic squamous cell carcinoma of the head and neck treated with a PD-1 inhibitor

Presentation Number
32P
Lecture Time
12:00 - 12:00
Speakers
  • Natasha Honoré (Woluwe-Saint-Lambert, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Only 15-20% of recurrent and/or metastatic squamous cell carcinoma of the head and neck (R/M SCCHN) patients derive long-term benefit from nivolumab or pembrolizumab. We developed a ctDNA tumor-agnostic assay aimed at the early prediction of single agent PD-1 inhibitor efficacy in R/M SCCHN.

Methods

Our tumor-agnostic assay included 37 genes frequently mutated in R/M SCCHN and two HPV16 genes. Primary endpoint was the concordance between ctDNA kinetics (ΔctDNA) and best overall response (BOR) according to Response Evaluation Criteria in Solid Tumors version 1.1 (RECISTv1.1). ΔctDNA was defined as the difference in mean variant allele frequency (VAF) between the on-treatment sample harvested 6-10 weeks (FU1) after PD-1 inhibitor initiation and the pre-treatment plasma sample (ΔctDNA = mean FU1 VAF - mean pre-treatment VAF).

Results

ctDNA was detected in 35/44 (80%) of the pre-treatment plasma samples. The concordance between ΔctDNA and imaging response was observed in 74%. Median PFS was 8.6 months in the favorable ΔctDNA group and 2.5 months in the unfavorable ΔctDNA group (p=0.057). Median OS was 18.1 and 8.2 months in the favorable and unfavorable ΔctDNA groups, respectively (p=0.13). In patients with PD-L1 expressing SCCHN (Combined Positive Score ≥1), OS was significantly better in patients with favorable ΔctDNA compared with patients with unfavorable ΔctDNA: median OS was 8.4 and 41.5 months (p=0.033), respectively.

Conclusions

Tumor-agnostic ctDNA analysis for HPV-negative and HPV-positive R/M SCCHN is feasible. ctDNA kinetics show promising results in predicting the efficacy of PD-1 inhibitors in R/M SCCHN.

Clinical trial identification

NCT02139020.

Legal entity responsible for the study

The authors.

Funding

Foundation Saint-Luc.

Disclosure

C. Van Marcke de Lummen: Financial Interests, Institutional, Advisory Board: Eli Lilly, Novartis, AstraZeneca; Non-Financial Interests, Personal, Member of Board of Directors: BSMO. R. Galot: Other, Personal, Other, Travel expenses: Merck. J. Machiels: Financial Interests, Institutional, Advisory Board: Novartis, MSD, Pfizer, Roche, Debio, AstraZeneca, Innate, Nanobiotix, Bayer, Merck Serono, Boehringer Ingelheim, BMS, Pfizer, Cue Pharma, Incyte, Janssen, Johnson & Johnson, ALX Oncology, F-star, Nektar, F-star, Seagen, Astellas, Genmab; Financial Interests, Institutional, Other, Travel expense: Gilead, MSD, Sanofi; Financial Interests, Institutional, Steering Committee Member: AstraZeneca, MSD; Financial Interests, Institutional, Coordinating PI: MSD, iTeos, eTheRNA; Financial Interests, Institutional, Local PI: Pfizer, Ceylad, MSD, Novartis, KURA, Roche, Lilly, Boehringer, Sanofi-Aventis, Incyte, Bayer, Merck Serono, Janssen, Johnson & Johnson, Amgen, AbbVie, GSK; Non-Financial Interests, Personal, Leadership Role, Chair: EORTC head and neck group. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

33P - Impact of prior treatment on baseline immune biomarkers and nivolumab + relatlimab (NIVO+RELA)-induced changes in the tumor microenvironment (TME) in patients (pts) with melanoma from RELATIVITY-020

Presentation Number
33P
Lecture Time
12:00 - 12:00
Speakers
  • Paolo A. Ascierto (Napoli, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

In RELATIVITY-020, NIVO+RELA was well tolerated and demonstrated durable anti-tumor activity in pts with advanced melanoma refractory to prior immunotherapy (IO-rf), with enriched responses seen in pts whose tumors expressed programmed death ligand 1 (PD-L1) or lymphocyte activation gene-3 (LAG-3). To better understand subgroups of pts that may derive increased benefit from NIVO+RELA, we performed exploratory IO biomarker analyses in RELATIVITY-020.

Methods

LAG-3, PD-L1, and CD8 levels were evaluated by immunohistochemistry in tumor samples from pts at baseline who were IO-naïve (1L) or IO-rf, and within 4 weeks on NIVO+RELA. Immune-related gene expression signatures were evaluated by RNA-seq in pts with modified definitions of primary vs secondary anti–programmed death-1/PD-L1 resistance. Association between biomarkers and response was determined by RECIST v1.1.

Results

Baseline LAG-3, PD-L1, and CD8 levels were higher in IO-rf pts whose last therapy was IO (IO-prior) vs non-IO; objective response rate (ORR) with NIVO+RELA was 12.8 vs 8.3%, respectively. Baseline biomarkers were higher in NIVO+RELA responders vs non-responders (P<0.05) only in IO-prior pts. Lower immune and inflammatory gene expression signatures were observed in pts with primary vs secondary IO resistance. Baseline LAG-3, PD-L1, and CD8 levels were similar for 1L vs IO-rf pts despite a higher response to NIVO+RELA in 1L pts. After NIVO+RELA, a significant increase in LAG-3 in the TME was noted in both IO-rf and 1L pts (log2 fold change: 0.73 and 1.46; both P<0.01), whereas PD-L1 and CD8 increased significantly in 1L pts only.

Conclusions

IO-rf pts whose last line of therapy prior to RELATIVITY-020 was IO vs non-IO had elevated inflammation-associated tumor biomarkers and may be more responsive to NIVO+RELA, although the ORR difference was modest. Variance in immune infiltration gene-expression signatures in primary vs secondary IO-resistant tumors suggest these subgroups have distinct TMEs and IO sensitivities. Modulation of LAG-3 in both IO-rf and 1L pts suggests NIVO+RELA can alter the TME in both settings, although modulation was greater in 1L pts.

Clinical trial identification

NCT01968109.

Editorial acknowledgement

Editorial support was provided by Allyson Koyen Malashevich, PhD, and Agata Shodeke, PhD, of Spark Medica Inc., funded by Bristol Myers Squibb.

Legal entity responsible for the study

Bristol Myers Squibb.

Funding

Bristol Myers Squibb.

Disclosure

P.A. Ascierto: Financial Interests, Personal, Other, Consultant and Advisory Role: BMS, Roche Genentech, MSD, Novartis, Merck Serono, Pierre Fabre, AstraZeneca, Sun Pharma, Sanofi, Idera, Sandoz, Immunocore, 4SC, Nektar, Boehringer Ingelheim, Regeneron; Financial Interests, Personal, Other, Consultant and Advisory Role. Travel support: Pfizer/Array; Financial Interests, Personal, Other, Consultant Role: Italfarmaco; Financial Interests, Personal, Other, Advisory Role: Eisai, Seagen; Financial Interests, Personal, Other, Consultant Role: Daiichi Sankyo, Pfizer, Oncosec, Nouscom, Lunaphore; Financial Interests, Personal, Other, Consultant role and travel support: Bio-AI Health; Financial Interests, Personal, Other, Consultant role: Medicenna; Financial Interests, Personal, Advisory Board, Consultant and Advisory Role: iTeos; Financial Interests, Personal, Advisory Board, Consultant and Advisory role: ValoTx; Financial Interests, Personal, Advisory Board, Consultant and Advisor role. Travel support: Replimmune; Financial Interests, Personal, Advisory Board, Advisor role: Bayer; Financial Interests, Institutional, Funding, Clinical Trial: Pfizer/Array, Roche Genentech, Sanofi; Financial Interests, Personal, Advisory Board: Erasca; Financial Interests, Institutional, Funding, Clinical trial and translational research: BMS; Non-Financial Interests, Personal, Leadership Role, President since 2010: Fondazione Melanoma Onlus Italy; Non-Financial Interests, Personal, Leadership Role, President since 2014: Campania Society of ImmunoTherapy of Cancer (SCITO) Italy; Non-Financial Interests, Personal, Member: SITC, ASCO, EORTC Melanoma Cooperative Group, AIOM, SMR; Non-Financial Interests, Personal, Other, Member of Steering Committee since 2016: Society for Melanoma Research (SMR); Non-Financial Interests, Personal, Member of Board of Directors, November 2017 - December 2021: Society for Immunotherapy of Cancer (SITC). H. Tang, S. Dolfi: Financial Interests, Personal, Full or part-time Employment: Bristol Myers Squibb; Financial Interests, Personal, Stocks/Shares: Bristol Myers Squibb. M.S. Nyakas: Financial Interests, Personal, Speaker, Consultant, Advisor, Lectures and/or expert meetings: Bristol Myers Squibb, Novartis, MSD, Roche, Pierre Fabre. I. Svane: Financial Interests, Personal, Advisory Board: BMS, Pierre Fabre, Novartis; Financial Interests, Personal, Invited Speaker: MSD, Pierre Fabre, Novartis, Roche, BMS; Financial Interests, Institutional, Research Grant: Enara Bio, Adaptimmune, Lytix Biopharma, TILT Biotherapeutics; Financial Interests, Personal, Writing Engagement: MSD; Financial Interests, Personal, Stocks/Shares, Cofounder and Founder warrents: IO Biotech; Financial Interests, Institutional, Funding: Evaxion; Non-Financial Interests, Personal, Principal Investigator: BMS, Novartis, Roche, TILT Biotherapeutics, Lytix Biopharma. E. Muñoz Couselo: Financial Interests, Personal, Advisory Board: BMS, Novartis, Sanofi, Pierre Fabre, Sun Pharma, MSD; Financial Interests, Personal, Invited Speaker: Novartis, Roche, BMS, Sanofi, MSD, Pierre Fabre. J.J. Grob: Financial Interests, Personal, Speaker, Consultant, Advisor, Personal fees: Novartis, Bristol Myers Squibb; Financial Interests, Personal, Advisory Board: Novartis, Bristol Myers Squibb, MSD, Philogen, Pierre Fabre, Sanofi, Roche, Amgen. C.A. Gomez-Roca: Financial Interests, Personal, Invited Speaker: BMS, Roche/Genentech, Pierre Fabre; Financial Interests, Personal, Other, IDMC member: PharmaMar; Financial Interests, Personal, Advisory Board: Macomics; Financial Interests, Institutional, Research Grant: Roche/Genentech; Financial Interests, Institutional, Coordinating PI: BMS, Amunix, Hookipa, Kazia Therapeutics, IDEAYA; Financial Interests, Personal, Steering Committee Member: BMS, Genentech; Non-Financial Interests, Personal, Member of Board of Directors: FITC (Société française d'Immuno-Thérapies du Cancer); Non-Financial Interests, Personal, Officer: ESMO Membership Committee, ESMO - MCBS Extended Working Group; Non-Financial Interests, Personal, Officer, Young Investigators Committee at imCORE: inFLAME; Non-Financial Interests, Personal, Member of Board of Directors, Network of Early Phase Units: OncoDistinct; Non-Financial Interests, Personal, Leadership Role: FITC (Société française d'Immuno-Thérapies du Cancer). V. Chiarion Sileni: Financial Interests, Personal, Other, Travel, registration, and accommodation to attend ASCO annual meetings: Pierre Fabre; Financial Interests, Personal, Invited Speaker: Pierre Fabre; Financial Interests, Personal, Advisory Board: Merck Sharpe & Dohme; Non-Financial Interests, Personal, Other, Steering Committee: Roche. K. Peltola: Financial Interests, Personal, Advisory Board: MSD, Ipsen, Roche, BMS, Pfizer, Lilly, Novartis, Bayer; Financial Interests, Personal, Stocks/Shares: Faron Pharmaceuticals; Financial Interests, Institutional, Local PI, Conduct of sponsored clinical trial: Novartis; Financial Interests, Institutional, Local PI, Sponsored clinical trial: Exelixis; Financial Interests, Institutional, Local PI, Several clinical trials: BMS, MSD, Roche; Financial Interests, Institutional, Local PI, clinical trials: Incyte; Financial Interests, Institutional, Local PI, Conduct of clinical trials: Pfizer; Financial Interests, Institutional, Local PI, Conduct of clinical trial: Bayer. J. Larkin: Financial Interests, Personal, Invited Speaker: BMS, Pfizer, Roche, Pierre Fabre, AstraZeneca, Novartis, EUSA Pharma, MSD, Merck, GSK, Ipsen, Aptitude, Eisai, Calithera, Ultimovacs, Seagen, Goldman Sachs, eCancer, Inselgruppe, Agence Unik; Financial Interests, Personal, Other, Consultancy: Incyte, iOnctura, Apple Tree, Merck, BMS, Eisai, Debipharm; Financial Interests, Personal, Other, Honorarium: touchIME, touchEXPERTS, VJOncology, RGCP, Cambridge Healthcare Research, Royal College of Physicians; Financial Interests, Institutional, Funding: BMS, MSD, Novartis, Pfizer, Achilles, Roche, Nektar, Covance, Immunocore, Pharmacyclics, Aveo. I. Melero: Financial Interests, Personal, Advisory Board: Agneus, Alligator Bioscience, AstraZeneca, BMS, BioLineRx, Boehringer Ingelheim, Boston Pharma, CRISPR Therapeutics, CatalYm GmbH, Crescendo Biologics, Curon Biopharmaceutical, Highlight Therapeutics, EMD Serono, F-Star, Genentech, Genmab, Gossamer Bio, Hookipa Pharma, HotSpot Therapeutics, Inc., ImmuneSensor Therapeutics, Inc., Janssen, MSD, Merus N.V., Monopteros Therapeutics, Noxxon Pharma AG, Numab, Phenomic Bio, Pieris Pharmaceuticals GmbH, Pierre Fabre, Roche, Sanofi, Senti Biosciences, Servier, Shattuck Labs, Third Rock Ventures; Financial Interests, Personal, Other, Consultant: Pharma Mar; Financial Interests, Institutional, Coordinating PI: AstraZeneca, Roche, BMS, Genmab, Alligator. M. Callahan: Financial Interests, Institutional, Research Funding, Support that goes to MSK: Bristol Myers Squibb. R. Dummer: Financial Interests, Personal, Other, Consulting and/or advisory role: Novartis, Merck Sharp & Dohme (MSD), Bristol Myers Squibb (BMS), Roche, Amgen, Takeda, Pierre Fabre, Sun Pharma, Sanofi, Catalym, Second Genome, Regeneron, Alligator, MaviVAX SA, Simcere, touchIME, T3 Pharma, Pfizer. P. Djidel: Financial Interests, Personal, Stocks/Shares: Bristol Myers Squibb; Financial Interests, Personal, Full or part-time Employment: Bristo Myers Squibb. D. Warad: Financial Interests, Personal, Full or part-time Employment: Bristol Myers Squibb; Financial Interests, Personal, Funding, Support for travel, meetings, equipment, as part of employment: Bristol Myers Squibb; Financial Interests, Personal, Stocks/Shares: Bristol Myers Squibb. E.J. Lipson: Financial Interests, Personal, Advisory Board: Bristol Myers Squibb, Merck, Sanofi, Regeneron, Genentech, Eisai, Instil Bio, Natera, Nektar Therapeutics, Pfizer, Rain Therapeutics, CareDx, Immunocore, Novartis, Replimune, HUYA Bioscience International; Financial Interests, Personal, Other, Consultant: OncoSec; Financial Interests, Institutional, Coordinating PI: Bristol Myers Squibb; Financial Interests, Institutional, Local PI: Regeneron, Merck, Sanofi. C. Garnett-Benson: Financial Interests, Personal, Full or part-time Employment: Bristol Myers Squibb; Financial Interests, Personal, Funding, Support for attending meetings and/or travel; Receipt of equipment, materials, drugs, medical writing, gifts or other services, as part of employment: Bristol Myers Squibb; Financial Interests, Personal, Stocks/Shares: Bristol Myers Squibb.

Collapse
Poster Display (ID 34) Poster Display

34P - Heterogeneous response to Immune Checkpoint Inhibitors in metastatic melanoma patients - assessment of lesion-level response with 18F-FDG PET/CT

Presentation Number
34P
Lecture Time
12:00 - 12:00
Speakers
  • Katja Strasek (Ljubljana, Slovenia)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Existing therapy response assessments are based largely on summing changes in target lesion diameters assessed on CT or MRI (e.g., RECIST) and do not consider lesion response heterogeneity. Evaluating lesion-level response of all lesions enables identification of novel patterns, such as heterogeneous response (HeR). In our study, we use a lesion-level response model, based on 18F-FDG PET/CT analysis to correlate lesion response patterns and treatment outcomes in metastatic melanoma (MM) patients treated with immune checkpoint inhibitors (ICI).

Methods

We retrospectively assessed 27 MM patients treated with anti-PD-1 therapy in two academic centers. Lesions were manually segmented on baseline (PET1) and first follow-up (PET2, month 3-4) 18F-FDG PET/CT. Lesions were categorized based on relative change of SUVtotal from PET1 to PET2: complete response (iCR, disappeared on PET2), partial response (iPR, ΔSUVtotal<-30%), stable disease (iSD, |ΔSUVtotal | ≤ 30%), progressive disease (iPD, ΔSUVtotal>+30%), new disease (iND, new on PET2). HeR was defined as patients having at least one iCR or iPR and iPD or iND lesion. Survival analysis was done using Cox regression analysis. Patients were dichotomized into two groups based on their clinical status at the end of treatment (EOT): progressive disease (PD) and non-progressive disease patients (non-PD). Two-proportion z-test was used to compare proportion of patients with HeR in both groups.

Results

426 lesions were segmented and categorized as iCR (56%), iPR (10%), iSD (3%), iPD (8%), iND (23%). Patients with one or more iPD lesion (HR = 1.8, p= 0.003), two or more iND or iPD lesions (HR = 1.9, p=0.003), or identified with HeR (HR = 1.42, p=0.04) on PET2 had significantly shorter PFS. 37% (10/27) patients were identified to have HeR: 2/13 non-PD and 8/14 PD patients. Patients with HeR at PET2 have a significantly higher probability of progression at EOT (p=0.02).

Conclusions

Lesion-level analysis of 18F-FDG PET/CT response can recognize non-responding lesions. We found that patients with HeR had significantly shorter PFS. We anticipate a future interventional study where early identification of non-responding lesions may be amendable to local therapies.

Clinical trial identification

At University of Wisconsin Carbone Cancer Center, the study was approved by Institutional Review Board (UW14084). At Oncology Institute Ljubljana, the study was approved by the Ethics comitee of OIL and Comitee for assessment of Clinical protocols (ERDIKE-005/2020, ERID-KSOPKR-002/2020).

Legal entity responsible for the study

The authors.

Funding

University of Wisconsin Carbone Cancer Center (UWCCC), National Cancer Institute of the National Institutes of Health, The Slovenian Research Agency (ARRS).

Disclosure

R. Jeraj: Financial Interests, Personal and Institutional, Funding: AIQ Solutions. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

35P - Peripheral immunotype classification for monitoring Soft Tissue Sarcoma patients

Presentation Number
35P
Lecture Time
12:00 - 12:00
Speakers
  • Jani Sofia Almeida (Coimbra, Portugal)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Current immune research on soft tissue sarcomas (STS) primarily focuses on analyzing the tumor microenvironment and adjacent lymph nodes. However, alternative methods are necessary to monitor immune responses in non-surgical patients due to limited opportunities to study tumor-infiltrating lymphocytes. This study aimed to assess the peripheral immune profile of STS patients and its impact on patient survival.

Methods

Blood samples were collected from 55 STS patients and age-matched healthy individuals. Flow cytometry and qRT-PCR were conducted on whole blood, to proceed with deep immunophenotyping and gene expression quantification, respectively. Proteomic analysis was also performed on plasma samples by xMAP technology. Unsupervised clustering analysis was used to classify patients based on their immunotype.

Results

Significant differences were found between STS patients and healthy individuals. STS patients showed lymphopenia, particularly affecting B and CD4 T cells, with an expansion of myeloid cells such as granulocytes and monocytic-derived suppressor cells (M-MDSC). Gene expression analysis revealed decreased levels of activatory and cytotoxic-related factors, along with increased expression of ARG1, which may inhibit anti-tumoral activity. A tendency for increased levels of IL-10 and soluble checkpoint inhibitors VISTA and TIMD-4 were also observed in plasma samples. Furthermore, patients were classified into three distinct immunotypes: \"immune-high,\" \"immune-intermediate,\" and \"immune-low.\" These immunotypes correlated significantly with time after collection (TAC), which refers to the time from sample collection to the end of the study or occurrence of a death event. \"Immune-high\" patients had elevated levels of immune-related factors associated with cytotoxic/effector activity and longer TAC, while \"immune-low\" patients had increased levels of immune-related factors associated with inflammation and inhibition, and shorter TAC.

Conclusions

The correlation between immunotypes and survival times emphasizes the importance of studying peripheral blood samples in STS. Evaluating the peripheral immune response can be a valuable tool for monitoring and predicting outcomes in STS patients.

Legal entity responsible for the study

The authors.

Funding

FCT - Portuguese Foundation for Science and Technology.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

36P - Expression of germinal center B cell- and Th17 cell-related transcripts are prognostic of soft-tissue sarcoma patient outcomes

Presentation Number
36P
Lecture Time
12:00 - 12:00
Speakers
  • Giulia Petroni (Firenze, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Soft tissue sarcomas (STS) are a group of rare and heterogeneous tumors with different clinical and biological behaviors. Better prognosis has been associated to an inflamed tumor microenvironment (TME) characterized by the presence of tertiary lymphoid structures (TLSs) rich in B cells. However, studies are still needed to better characterize cellular and molecular components of the TME the that could favor or hamper B cell organization in STS-associated TLSs.

Methods

The transcriptomic immune profiling of STS TME was performed using the Human Immunology V2 Panel and the NanoString nCounter platform on 33 STS biopsies or surgical samples. Patients with primary or locally recurrent STS were enrolled and treated at the Careggi Hospital (Florence, Italy; RESEARCH study). Results were validated by retrieving RNA-sequencing and clinical data for 164 STS patients from The Cancer Genome Atlas (TCGA) SARC project.

Results

We investigated the impact of gene signatures discriminating for specific immune cells and we found a significant association with relapse-free survival (RFS) only for germinal center (GC) B cell- and T helper 17 (Th17) cell-related signatures (p < 0.05). We observed a correlation between GC-B and Th17 signatures (r = 0.77, p < 0.0001), and the combination of both GC B and Th17 signatures was associated with better RFS, while low expression of both GC B and Th17 signatures was associated with the worst survival outcome (p = 0.035). The prognostic value of the GC-B/Th17 signature was confirmed on data from TCGA SARC project for both RFS (p = 0.016) and overall survival (p = 0.014). Differential gene expression analysis showed a more inflamed landscape in GC-B/Th17-high tumors. Conversely, the upregulation of genes encoding immunosuppressive molecules and genes related to tumor-associated macrophages with an immunosuppressive phenotype, were observed in GC-B/Th17-low tumors.

Conclusions

Our data identified a novel prognostic signature composed of GC B cell- and Th17 cell-related genes for STS patients, and suggest a potential collaboration between GC B cells and Th17 cells in mediating antitumor immune responses and potentially TLS formation in STS.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

38P - Machine learning-based pathomics model to predict the infiltration of Treg and prognosis in IDH-wt GBM

Presentation Number
38P
Lecture Time
12:00 - 12:00
Speakers
  • Shaoli Peng (Fuzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Regulatory T cells (Treg) play a crucial role as prognostic factors and intervention targets for isocitrate dehydrogenase (IDH)-wild type (wt) glioblastoma (GBM). The study aimed to construct a model predicting the Treg infiltration in IDH-wt GBM patients using pathomics techniques and explore related biological processes.

Methods

Flow cytometry was used to detect the proportion of Treg in orthotopic mouse glioblastoma brain tissue. Clinical data of IDH-wt GBM patients from the TCGA database were analyzed retrospectively. Features were extracted from HE-stained biopsy sections using the Pyradiomics package. The pathomics model was constructed using the Gradient Boosting Machine algorithm after performing feature selection with mRMR and Relief algorithms. Cox proportional hazard regression analysis was employed to access the association between pathomics score (PS) and overall survival (OS). Transcriptomic data were analyzed through GSEA set enrichment, differential gene expression, and correlation analyses.

Results

The study established a pathomics model with 3 pathomics features that effectively predicted Treg infiltration (Training set: AUC=0.807; Validation set: AUC=0.735). PS positively correlated with high Treg expression. Survival analysis indicated that patients with high PS had significantly lower OS than the low PS group (median survival time: 12.0 vs 14.4 months; p=0.009). Multivariate COX analysis revealed that high PS expression independently served as a prognostic risk factor for IDH-wt GBM patients (HR, 2.16; 95% CI, 1.269-3.677; p=0.005). Subgroup analysis showed that high PS was a risk factor for OS in patients receiving chemotherapy (HR, 2.232; 95% CI, 1.309-3.905; p=0.003) or radiotherapy (HR, 1.882; 95% CI, 1.161-3.049; p=0.01).GSEA enrichment analysis revealed significant associations of PS with the NOTCH, IL-6/JAK/STAT3 signaling pathways. High PS significantly correlated with elevated RAD50 expression.

Conclusions

The developed pathomics model, based on machine learning algorithms, can noninvasively predict Treg infiltration and prognosis in IDH-wt GBM patients. The biological processes related to the model may involve RAD50 and pathways, including NOTCH, IL-6/JAK/STAT3.

Legal entity responsible for the study

The authors.

Funding

The Joint Funds for the Innovation of Science and Technology, Fujian Province (No.2021Y9301).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

39P - Patient selection based on hyperprogressive disease risk nearly doubles survival benefit of immune checkpoint blockade: validation of a pan-tumor tissue agnostic combined biomarker approach

Presentation Number
39P
Lecture Time
12:00 - 12:00
Speakers
  • Partha S. Ray (Lincolnwood, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

FOXC1, MKI67 and PDL1 expression that tracks plasticity, proliferation and immune evasion accurately predicts efficacy of Immune Checkpoint Blockade (ICB) with PD1/PDL1 inhibitor drugs. We sought to validate whether patient selection based on predicted Hyperprogressive Disease (HPD) risk using such an approach can improve Progression-free Survival (PFS) and/or Overall Survival (OS) in clinical trial and real world cohorts.

Methods

Pre-treatment tumor RNA-Seq data obtained from patients diagnosed with advanced/metastatic (a/m) melanoma (n=154,121), non-small cell lung cancer (NSCLC, n=82,140) and renal cell carcinoma (RCC, n=250,120) were analyzed for FOXC1, MKI67 and PDL1 expression, and correlated with overall response rate (ORR), PFS, OS and HPD, the latter defined as time-to-treatment-failure (TTF) <=2 months post-treatment initiation. Optimized biomarker cutoff values based on model AUC were leave-one-out cross validated and cancer-type specific (CTS) prediction algorithms derived. The unmodified strategy was then validated in the independent, CTS validation datasets.

Results

Predicted Clinical Responders (CR) displayed marked improvement in PFS and OS compared to Predicted Non-Responders with standard Progressive Disease (PD) or Hyperprogressive Disease (HPD). [a/m Melanoma OS HR=0.35 (0.187-0.666)95%CI, p=0.0004; a/m NSCLC OS HR=0.24 (0.119-0.499)95%CI, p<0.0001; a/m RCC OS HR= 0.45 (0.255-0.810)95%CI, p=0.008].

Patient stratification by HPD risk and median overall survival benefit

Cancer type Sample size ICB median OS HPD median OS PD median OS CR median OS
Melanoma Liu et al 121 22.5 mos 5 mos 17 mos 37.5 mos
NSCLC SU2C-mark cohort Ravi et al 140 17 mos 13 mos 19 mos 35.5 mos
RCC CM-025 cohort Motzer et al 120 27.5 mos 17.5 mos 22.5 mos 57 mos

Conclusions

Compared to ICB administered in a non-patient stratified manner, restricting ICB treatment only to those patients who are predicted CRs results in nearly a two-fold or greater improvement in the median OS survival benefit in all cancer types examined. Patients predicted to have elevated HPD risk should be considered for treatment with non-ICB regimens or offered enrollment in clinical trials that combine ICB with other drugs.

Clinical trial identification

NCT01668784; Nov 5, 2015.

Legal entity responsible for the study

P.S. Ray.

Funding

Has not received any funding.

Disclosure

P.S. Ray: Non-Financial Interests, Institutional, Advisory Role: Onconostic Technologies: Practicing surgical oncologist and clinical investigator, identified as the presenting author on the submitted abstract. PSR is not an employee of the company (Onconostic Tehcnologies), but is founder and Chairman of the Scientific Advisory Board of the company which is an unpaid position. T. Ray, R. Hussa: Financial Interests, Personal, Full or part-time Employment: Onconostic Technologies, Inc.; Financial Interests, Personal, Stocks/Shares: Onconostic Technologies, Inc. C. Taylor: Financial Interests, Personal, Advisory Board: Onconostic Technologies, Inc.; Financial Interests, Personal, Stocks/Shares: Onconostic Technologies, Inc.

Collapse
Poster Display (ID 34) Poster Display

37P - Association between blood cell composition and survival outcome in glioma patients: A topic modeling approach

Presentation Number
37P
Lecture Time
12:00 - 12:00
Speakers
  • Sara Hadad (San Francisco, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

40P - The role of low avidity tumour-specific CD8+ T cells in immunotherapeutic response to anti-PD-1

Presentation Number
40P
Lecture Time
12:00 - 12:00
Speakers
  • Doreen Lau (Oxford, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

T cell avidity plays a crucial role in antigen presentation and influences the quality of TCR signaling and T cell metabolic fitness. It is crucial in chronic inflammation e.g. cancer where persistent antigen exposure and chronic T cell stimulation may lead to exhaustion. Thus, mechanistic insights on the roles of CD8+ specificities and T cell avidity of naturally arising tumour-specific T cells where both high (Tethi) and low (Tetlo) avidity T cells recognising the same pMHC co-exist in the same tumour, are crucial for understanding resistance to PD-1 immunotherapy.

Methods

CT26 models were treated with anti-PD-1 on days 3, 6 and 9 following tumour implantation generating variable responses during early tumour development. Tetramer staining and T cell avidity measurement using acoustic force spectroscopy were conducted to determine the frequency and avidity of CD8+ T cells targeting the tumour-specific epitope GSW11. Tethi and Tetlo were functionally characterised using flow cytometry, RNA-seq, in vitro and in vivo cytotoxicity experiments.

Results

Treatment success with anti-PD-1 was associated with the preferential expansion of Tetlo. Tetlo were precursor exhausted with higher expression of Tcf-1 and T-bet, and lower expression of CD39, PD-1 and Eomes compared to Tethi. Pathways related to TCR signaling, cytotoxicity and oxidative phosphorylation were significantly upregulated in Tetlo found in both responding and non-responding tumours compared to Tethi. Interestingly, a small percentage of Tetlo found in the non-responding tumours were functional but metabolically challenged. In vitro studies showed that Tetlo exhibited higher cytotoxicity than Tethi. Curative response was achieved when Tetlo were adoptively transferred and in combination with anti-PD-1.

Conclusions

Targeting subdominant T cell responses with lower avidity against pMHC affinity neoepitopes showed potential for improving PD-1 immunotherapy. Future interventions may consider expanding low avidity populations via adoptive transfer or drugs targeting immunometabolism. These approaches may be combined with non-invasive tumour metabolism imaging and T cell tracking to understand the impact of immunometabolism on T cell dynamics at a system level.

Legal entity responsible for the study

The authors.

Funding

Worldwide Cancer Research Fund (20-0229), Cancer Research UK Programme Grant (A28279).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

41P - Contrasting drivers of response to immunotherapy across solid tumour types: results from analysis of >2500 cases

Presentation Number
41P
Lecture Time
12:00 - 12:00
Speakers
  • Danwen Qian (London, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Multiple genomic and transcriptomic biomarkers have been associated with response to immune checkpoint inhibitor (CPI) therapy. Our previously published meta-analysis of biomarkers included over 1000 patients across multiple cancer types, which demonstrated clonal TMB and CXCL9 expression as the strongest pan-cancer biomarkers of CPI response. However, over half of the variance in response to CPI treatment remains unexplained, for example 52% of patients who respond to CPI treatment were low-TMB and showed no evidence of an oncogenic viral infection.

Methods

To address the missing understanding in CPI response mechanisms, we have recently extended our dataset to 16 immunotherapy treated cohorts, across 10 solid tumour types, encompassing >2500 treated patients. Matched tumour/normal whole exome sequencing (WES) data and tumour transcriptomic data (RNAseq) was available. All data was accessed in raw sequencing read format and reprocessed through a standardized bioinformatics pipeline. Using pubmed key word search we completed a systematic literature review of ∼1,000 papers, which identified >200 unique biomarkers that could be quantified in exome or RNA-seq data. Data will be presented on these biomarkers to understand the full landscape of biomarkers responses to CPI therapy.

Results

Biomarkers with strongest effect size pan-cancer, and per cancer type will be presented, highlighting divergent mechanisms of CPI response across histology types. We will present data on emerging checkpoint targets in late-stage trials (e.g. LAG3, TIGIT, VISTA) across cancer types, to assess the potential of these new candidates to overcome resistance to current anti-PD-1 or anti-CTLA-4 treatment. We will additionally assess expression of tumor-associated antigen targets in solid tumour CAR T cell and vaccine trials (e.g. MAGE, NY-ESO-1, MUC1), to understand their role in CPI response. Lastly, we will discuss how these biomarkers can be combined into a multimodal test to accurately predict CPI response.

Conclusions

Results from this work have strongly highlighted the varying determinants of immunotherapy response across solid tumour types, offering unique insight into both tumour intrinsic and extrinsic drivers of immunogenicity.

Legal entity responsible for the study

The authors.

Funding

The Medical Research Council.

Disclosure

C. Swanton: Financial Interests, Personal, Invited Speaker, Activity took place in 2016: Pfizer, Celgene; Financial Interests, Personal, Invited Speaker, October 26th 2020: Novartis; Financial Interests, Personal, Invited Speaker: Roche/Ventana, BMS, AstraZeneca, MSD, Illumina, GSK; Financial Interests, Personal, Advisory Board, AdBoard - November 12th, 2020: Amgen; Financial Interests, Personal, Advisory Board, Current - since 2018: Genentech; Financial Interests, Personal, Advisory Board: Sarah Canon Research Institute; Financial Interests, Personal, Advisory Board, Joined October 2020. Also have stock options: Bicycle Therapeutics; Financial Interests, Personal, Other, Consultancy: Medicxi; Financial Interests, Personal, Advisory Board, Member of the Science Advisory Board. Also had stock options until June 2021: GRAIL; Financial Interests, Personal, Other, Consultancy agreement: Roche Innovation Centre Shanghai; Financial Interests, Personal, Advisory Board, 29 November - 1 December 2022: Novartis; Financial Interests, Personal, Invited Speaker, Oncology Collective - 2nd Nov - 4 Nov 2022 - Atlanta, USA: Roche; Financial Interests, Personal, Advisory Board, ctDNA advisory Board - 24th March 2023: AstraZeneca; Financial Interests, Personal, Invited Speaker, Pfizer Oncology 'Leading the revolution for the future: Pfizer; Financial Interests, Personal, Full or part-time Employment, Chief Clinician since October 2017: Cancer Research UK; Financial Interests, Personal, Ownership Interest, Co-Founder of Achilles Therapeutics. Also, have stock options in this company: Achilles Therapeutics; Financial Interests, Personal, Stocks/Shares, Stocks owned until June 2021: GRAIL, Apogen Biotechnologies; Financial Interests, Personal, Stocks/Shares: Epic Biosciences, Bicycle Therapeutics; Financial Interests, Institutional, Research Grant, Funded RUBICON grant - October 2018 - April 2021: Bristol Myers Squibb; Financial Interests, Institutional, Research Grant, Collaboration in minimal residual disease sequencing technologies: Archer Dx Inc; Financial Interests, Institutional, Research Grant: Pfizer, Boehringer Ingelheim; Financial Interests, Institutional, Trial Chair, Chief Investigator for the MeRmaiD 1and 2 clinical trials and chair of the steering committee: AstraZeneca; Financial Interests, Institutional, Research Grant, Research grant from Oct 2019 - July 2023 - Genetics of CIN and SCNAs for Targeted Discovery (SCEPTRE): Ono Pharmaceutical; Financial Interests, Institutional, Research Grant, Research Grants from 2015: Roche; Financial Interests, Personal, Other, Co-chief investigator: NHS-Galleri Clinical Trial; Financial Interests, Institutional, Research Grant, from October 2022: Personalis; Non-Financial Interests, Personal, Principal Investigator, Chief Investigator for MeRmaiD 1 and 2 clinical trials: AstraZeneca; Non-Financial Interests, Personal, Member of Board of Directors, From 2019-2022: AACR; Non-Financial Interests, Personal, Other, Board of Directors: AACR; Non-Financial Interests, Personal, Advisory Role, EACR Advisory Council member: EACR. K.R. Litchfield: Financial Interests, Personal, Invited Speaker: Roche Tissue Diagnostics; Financial Interests, Personal, Other, Consulting work: Kynos Therapeutics, Monopteros Therapeutics, Tempus; Financial Interests, Personal, Invited Speaker, Invited speaker: Ellipses Pharma; Financial Interests, Institutional, Research Grant: Ono/LifeArc; Financial Interests, Institutional, Research Grant, Research funding: Genesis Therapeutics; Non-Financial Interests, Institutional, Proprietary Information, Collaboration on data analysis: BMS. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

42P - TCCIA: A Comprehensive Resource for Exploring CircRNA in Cancer Immunotherapy

Presentation Number
42P
Lecture Time
12:00 - 12:00
Speakers
  • Jian-Guo Zhou (Zunyi, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immunotherapies targeting immune checkpoints have gained increasing attention in cancer treatment, emphasizing the need for predictive biomarkers. Circular RNAs have emerged as critical regulators of tumor immunity, particularly in the PD-(L)1 pathway, and have shown potential in predicting the efficacy of immunotherapies. Therefore, the development of a comprehensive resource that integrates circRNA profiles, immunotherapy response data, and clinical benefits is crucial for advancing our understanding of circRNA-mediated tumor-immune interactions and developing effective immunotherapy biomarkers.

Methods

To address these gaps, we constructed the Cancer CircRNA Immunome Atlas (TCCIA), the first database that combines circRNA profiles, immunotherapy response data, and clinical outcomes across multi-cancer types. The construction of TCCIA involved applying standardized preprocessing to the raw sequencing FASTQ files, characterizing circRNA profiles using CIRCexplorer2, analyzing tumor immunophenotypes through IOBR, and compiling immunotherapy response data from diverse cohorts treated with ICBs.

Results

TCCIA encompasses over 3,700 clinical samples obtained from 18 cohorts treated with ICBs, including PD-1/PD-L1 and CTLA-4 inhibitors, along with other treatment modalities. The database provides researchers and clinicians with a cloud-based platform that enables interactive exploration of circRNA data in the context of ICB. The platform offers a range of analytical tools, including visualization of circRNA abundance and correlation, association analysis between circRNAs and clinical variables, assessment of the tumor immune microenvironment, exploration of tumor molecular signatures, evaluation of treatment response or prognosis, and identification of altered circRNAs in immunotherapy-sensitive and resistant tumors. To illustrate the utility, we performed a re-analysis on a melanoma cohort with TCCIA and found that an isoform of circTMTC3 played a significant role in predicting unfavorable survival outcomes and treatment nonresponse.

Conclusions

TCCIA represents a significant advancement over existing resources, providing a comprehensive platform to investigate the role of circRNAs in immune oncology.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

43P - Immune and tumor cells expression of VISTA in a panel of cancer indications: A strategy to inform selection of patients treated with anti-VISTA

Presentation Number
43P
Lecture Time
12:00 - 12:00
Speakers
  • Pierre Launay (Toulouse, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

V-domain Ig suppressor of T-cell activation (VISTA) is a B7 family member that acts as an immune checkpoint regulator. VISTA is expressed in various cell subtypes such as tumor, myeloid, lymphoid, and endothelial cells. Within the tumor microenvironment (TME), its expression on myeloid cells favors immunosuppressive conditions and prevents T cell recruitment into tumors. In patients, high VISTA expression is associated with poor prognosis in multiple tumor indications, and act as a mediator of resistance to immune checkpoint therapies. Thus, VISTA appears as a potential therapeutic target in oncology requiring a more detailed characterization of its expression pattern across tumor indications to eventually inform future patient selection.

Methods

In tissue microarrays (TMAs) covering 23 solid tumor indications and representing more than 1000 patients, we investigated the expression of VISTA in combination with different immune and tumor cells markers. For this purpose, we developed and validated a 7-color multiplex immunofluorescence protocol to quantify VISTA expression on tumor cells (Cytokeratin/Melan A), cytotoxic CD8 T cells (CD8), myeloid cells (CD33), M2 macrophages (CD163), and PD-L1 positive cells (PD-L1).

Results

Consistent with the literature, we have observed that VISTA is not only predominantly expressed in the TME but also frequently and highly expressed in tumor cells in some indications such as sarcoma, mesothelioma, kidney, and gastric cancer for example. In the TME, VISTA is found to be highly expressed in CD33 and CD163 populations, particularly in kidney cancer and sarcoma. Moreover, we found that tumor infiltrating CD8 T-cells often expressed VISTA, notably in sarcoma, kidney cancer, and hepatocellular carcinoma.

Conclusions

Finally, we propose indications of interest that may support patient selection strategy for the development of anti-VISTA treatments.

Legal entity responsible for the study

Pierre Fabre Laboratories.

Funding

Pierre Fabre Laboratories.

Disclosure

P. Launay, A. Pillon, R. Bonnet, D. Vinet, A. Alloy, L. Lacastaigneratte, C. Welsch, C. Larzabal, G. Zorza, G. Gueguen Dorbes, O. Geneste, F. Hofmann, M-O. Roy: Financial Interests, Personal, Full or part-time Employment: Pierre Fabre.

Collapse
Poster Display (ID 34) Poster Display

44P - Exploratory Analysis of Peripheral Pharmacodynamic (PD) Biomarkers After Sitravatinib (Sitra) and Tislelizumab (TIS) in Advanced Solid Tumors: SAFFRON-103

Presentation Number
44P
Lecture Time
12:00 - 12:00
Speakers
  • Yi-Long Wu (Guangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Sitra is a spectrum-selective tyrosine kinase inhibitor targeting multiple receptors, including VEGFR2. Here, we present an exploratory analysis of PD biomarkers in SAFFRON-103, a phase 1b study investigating Sitra with TIS, an anti-PD-1 antibody, in patients (pts) with solid tumors including advanced non-squamous-non-small cell lung cancer (NSCLC), squamous-NSCLC, melanoma, or ovarian cancer.

Methods

Peripheral blood samples were collected at Cycle (C) 1 Day (D) 1, C2D1, and C3D1 prior to Sitra dosing, to investigate changes in cytokines (Meso Scale Discovery [MSD] multiplexing), plasma proteins (ELISA), and immune cell populations (fluorescence-activated cell sorting [FACS]). Generalized linear mixed models were used to estimate fold change and analyze biomarker changes; Wald tests were used to generate P-values.

Results

Baseline characteristics were balanced across pts with evaluable biomarker results (n=186 cytokines/plasma proteins, n=113 immune cell populations) and in the overall population (N=216). For all pts, changes in individual biomarker levels were consistent from C1D1 to both C2D1 and C3D1, with significant increases in VEGFA (P<0.0001; both) and CXCL10 (P<0.0001; both) and significant decreases across soluble (s) VEGFR2 (P<0.0001; both), peripheral G-MDSCs (P=0.0005; P=0.0002), and monocytes (P<0.0001; both). Estimated fold changes of PD biomarkers across tumor types are shown (Table). Changes in VEGFA (increased) and monocytes (decreased) after treatment (C2D1/C1D1) were associated with improved objective response rates (odds ratio [OR] 4.67, P=0.0005; OR 5.82, P<0.0001).

Estimated fold change from C1D1 Non-squamous-NSCLC Squamous-NSCLC Melanoma Ovarian cancer
C2D1 C3D1 C2D1 C3D1 C2D1 C3D1 C2D1 C3D1
ELISA/MSD eligible n=60 n=48 n=22 n=54
VEFGA 2.77 2.65 2.53 2.53 2.89 2.38 2.16 2.14
sVEFGR2 0.67 0.63 0.61 0.59 0.65 0.66 0.70 0.69
CXCL10 1.70 1.97 1.65 1.43 1.75 1.44 2.21 1.90
FACS eligible n=44 n=21 n=10 n=28
G-MDSCs 0.74 0.71 0.59 0.60 0.83 0.74 0.81 0.84
Monocytes 0.70 0.72 0.72 0.64 0.73 0.72 0.81 0.85

Abbreviations: G-MDSC, granulocyte-like myeloid derived suppressor cells; NSCLC, non-small cell lung cancer; VEFGA, vascular endothelial growth factor A; sVERGR2, soluble vascular endothelial growth factor receptor 2

Conclusions

VEGFA increased and sVEGFR2 decreased consistently and significantly after Sitra plus TIS therapy, demonstrating the on-target anti-angiogenesis effect of Sitra. Decrease of G-MDSCs and monocytes in peripheral blood indicates a potential immune-modulating role for Sitra with TIS.

Clinical trial identification

NCT03666143.

Editorial acknowledgement

This study was sponsored by BeiGene, Ltd. Medical writing support, under the direction of the authors, was provided by Gemma Walker, BSc, of Ashfield MedComms, an Inizio company, and was funded by BeiGene, Ltd.

Legal entity responsible for the study

BeiGene.

Funding

BeiGene.

Disclosure

Y. Wu: Non-Financial Interests, Personal, Advisory Role: AstraZeneca; Non-Financial Interests, Personal and Institutional, Coordinating PI: AstraZeneca, Roche, Merck; Financial Interests, Personal, Invited Speaker: AstraZeneca, Boehringer Ingelheim, Eli Lilly, Hengrui, MSD, Pfizer, Roche; Non-Financial Interests, Personal, Leadership Role: Chinese Thoracic Oncology Group (CTONG); Non-Financial Interests, Personal, Local PI: BeiGene, Innoven; Non-Financial Interests, Personal, Member: ASCO, ESMO, IASLC, CSCO; Financial Interests, Institutional, Research Grant: BMS, Pfizer. J.C. Goh: Non-Financial Interests, Institutional, Invited Speaker: Brisbane Cancer Conference; Financial Interests, Personal, Invited Speaker: GSK, MSD; Financial Interests, Personal, Speaker’s Bureau: MSD, GSK, Ipsen, Janssen, Eisai, AstraZeneca; Financial Interests, Personal, Advisory Board: Janssen, GSK, BMS, MSD, AstraZeneca; Financial Interests, Personal and Institutional, Full or part-time Employment, Part-time employment: Royal Brisbane & Women’s Hospital; Financial Interests, Personal and Institutional, Full or part-time Employment: ICON Chermside & Greenslopes; Financial Interests, Personal, Stocks/Shares: ICON Cancer Centres, Immutep; Non-Financial Interests, Personal and Institutional, Project Lead, Ceased; Collaborative Group: ITTACc trial; Non-Financial Interests, Institutional, Principal Investigator: MSD, BMS, Janssen, AstraZeneca, BeiGene, Sutro Biopharma, Exelixis, Pfizer, Alloplex, Alkermes, Mersana; Non-Financial Interests, Personal and Institutional, Member, Corporative trials group in ANZ, USA & Europe: ASCO, ANZGOG, ANZUP, ESMO; Non-Financial Interests, Institutional, Leadership Role: Member of ICON Cancer Centre MAC (Medical Advisory Committee). J. Zhao: Non-Financial Interests, Institutional, Coordinating PI: AstraZeneca; Non-Financial Interests, Personal, Invited Speaker: AstraZeneca, Roche, Merck, BeiGene, Pfizer; Non-Financial Interests, Personal, Local PI: Merck, Roche; Non-Financial Interests, Personal, Steering Committee Member: Pfizer, Pierre Fabre, BeiGene. Q. Zhou: Financial Interests, Personal, Speaker, Consultant, Advisor: Lecture and presentations fees to myself from AstraZeneca; Non-Financial Interests, Personal, Speaker, Consultant, Advisor: Boehringer Ingelheim, BMS, Eli Lilly, MSD, Pfizer, Roche, Sanofi outside the submitted work. M. Millward: Financial Interests, Personal, Advisory Board: BeiGene Australia Pty Ltd, Bristol Myers Squibb Australia Pty Ltd, AstraZeneca Australia Pty Ltd, The Limbic, Eli Lilly Australia Pty Ltd, IQVIA Australia Pty Ltd, Amgen Australia Pty Ltd, Merck Pte Ltd, Pfizer Australia Pty Ltd, Guardant Health, Roche Products Pty Ltd; Financial Interests, Personal, Full or part-time Employment, Employee: University of Western Australia; Financial Interests, Personal, Other, Consultant: Linear Clinical Research; Financial Interests, Institutional, Local PI, Trial payments to Institution: Bristol Myers Squibb, Genentech/Roche, BeiGene, Eli Lilly, Albion Laboratories, Akeso Biopharma, AbbVie, Five Prime Therapeutics, Dizal Pharma, Maxinovel, Amgen, Atridia, INXMED, Alpine Immune Sciences, Turning Point Therapeutics, IMPACT Therapeutics, Kinnate Biopharma, Rely Therapeutics, GenFleet Therapeutics, Vivace Therapeutics, Eucure Biopahrma, InventisBio, Cullinan Oncology, Tyra Biosciences, Axelia Oncology; Non-Financial Interests, Personal, Other, Scientific Advisory Committee member: Thoracic Oncology Group Australasia. T. Tian: Other, Institutional, Full or part-time Employment: GSDS, BeiGene USA, Inc. 1840 Gateway Drive, 3rd Floor, San Mateo, CA 94404; Financial Interests, Institutional, Stocks/Shares: GSDS, BeiGene USA, Inc. 1840 Gateway Drive, 3rd Floor, San Mateo, CA 94404. J. Guo: Financial Interests, Institutional, Speaker, Consultant, Advisor: Merck Sharp & Dohme, Roche, Bayer, Novartis, Simcere Pharmaceutical Group, Shanghai Junshi Biosciences, Oriengene. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

45P - Protein biomarkers associated with organ-specific immune-related toxicity and response to management identified by proteome analysis of extracellular vesicles from plasma

Presentation Number
45P
Lecture Time
12:00 - 12:00
Speakers
  • Anders H. Kverneland (Herlev, Denmark)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immune related adverse events (IrAEs) are a growing challenge in oncology affecting an increasing number of patients and putting a large strain on patient health and hospital resources. The current availability of biomarkers is highly dependent on the affected organ and the management strategy based on consensus or experiences with non-CPI induced autoimmune disease. In this study, we apply proteome analysis of extracellular vesicles from plasma to assess and discover biomarkers with the potential to aid clinical staging and care of patients suffering from IrAEs.

Methods

Thirty-six patients with grade 3-4 IrAEs (colitis, hepatitis or nephritis) were included in the study. Blood samples were collected at hospital admittance before or early in management (At tox), after initiation of high dose immunosuppressive therapy (At management) and after management (After tox). The collected plasma samples were enriched for extracellular vesicles using a novel workflow and analyzed using liquid-chromatography tandem mass spectrometry (LC-MS/MS). The data analysis was performed in R using the Limma package.

Results

The sample workflow allowed median quantification of 2015 proteins in the plasma samples. In patients responding to prednisolone, we found significant decreases in CRP, SAA1 and SAA2 and a significant increase in MMP9 At management compared to At tox. CRP, SAA1 and SAA2 together with EPS8 and CFP was also significantly lowered After tox. By comparing the IrAE subtypes, definite proteins associated to specific toxicities could be identified including ALDOB, ASL and DXCR in hepatitis, SLC47A2 and PRPF40A in colitis and NCAPH in nephritis.

Conclusions

Biomarkers for monitoring IrAE management are needed to improve the clinical care of patients. Our study identifies several proteins that correlate to specific types of IrAEs and to response to toxicity management, thus, constituting relevant IrAE biomarker candidates.

Clinical trial identification

Regional Ethical Commitee reference number: H-21027448.

Legal entity responsible for the study

Capital Region of Denmark.

Funding

Novo Nordisk Foundation.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

46P - Immunoprofiling of Peripheral Blood Cells as a Potential Predictor of Immune-Related Toxicity of PD-1 Inhibitors

Presentation Number
46P
Lecture Time
12:00 - 12:00
Speakers
  • Jan Podhorec (Brno, Czech Republic)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Checkpoint inhibitors have revolutionized treatment outcomes of several types of cancer. However, their administration is associated with unpredictable immune-related adverse events (irAEs). The objective of this study is to assess the potential predictive value of immune cells in peripheral blood for anticipating irAEs.

Methods

We prospectively enrolled patients with metastatic cancer treated with PD-1 inhibitors (nivolumab or pembrolizumab). Patients were recruited between 2017 and 2021 at the Masaryk Memorial Cancer Institute (Czech Republic). Before starting immunotherapy we performed immunoprofiling of key regulators and effectors of the immune system from peripheral blood cells using flow cytometry. We compared differences between patients who did not develop any or only mild grade (G) 1 irAE and patients who experienced clinically relevant irAE, specifically G2 and higher.

Results

We consecutively enrolled 63 patients. The median age was 66 years, with 19 (30.2 %) being female. Histological tumor types included 31 patients with melanoma, 22 with non-small cell lung cancer, 8 with renal carcinoma, and 5 with other malignancies. The majority of patients initiated treatment as first-line therapy (35 patients, 53 %). Out of these patients, 39 (61.9%) did not experience any irAEs, while 14 patients had G1 toxicity (22.2%), 10 patients (15.9%) had G2 toxicity, and 1 patient (1.6%) had G3 toxicity. G4 or G5 toxicity was not observed. Using the Mann-Whitney test, we determined that patients who did not experience any or mild (G1) immunologically mediated toxicity during treatment had significantly lower level of T helper lymphocytes CD4+ in peripheral blood before the initiation of treatment (P=0.034) compared to patients who experienced G2 or G3 toxicity during treatment. Additionally, they had lower level of naive T lymphocytes CD4+RO-CD27+ (P=0.012), lower level myeloid dendritic cells CD4-HLADR+ (P=0.040), and higher level of memory lymphocytes CD4+RO+CD27+ (P=0.006).

Conclusions

Our results support the hypothesis that initial flow cytometry parameters detectable in peripheral blood could significantly predict irAE. These findings need to be validated in a larger patient population.

Legal entity responsible for the study

The authors.

Funding

Ministry of Health of the Czech Republic, grant nr. NV18-03-00339.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

47TiP - A real-world study of multiparametric prediction of the efficacy of immune checkpoint inhibitors in combination with chemotherapy for advanced non-small cell lung cancer

Presentation Number
47TiP
Lecture Time
12:00 - 16:15
Speakers
  • Zihan Zhou (Nanjing, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immune checkpoint inhibitors (ICIs) combined with chemotherapy has become the first-line treatment option for non-small cell lung cancer (NSCLC) patients without driver mutation genes. However, a large amount of clinical data has confirmed that only part of NSCLC patients can achieve durable clinical benefits, while the incidence of adverse reactions is high. So, there is still an urgent need for reliable biomarkers to construct models for accurate prediction and assessment of treatment outcomes. Recently, ctDNA and tumor mutation burden (TMB) have been proposed as biomarkers for predicting the outcome of immunotherapy, but no consensus has been reached on the cutoff value of TMB, ctDNA clearance threshold and optimal sampling time point. Therefore, this study aims to develop a prediction model for the efficacy of chemo-immunotherapy based on the clinical multiparameter data, and explore the ctDNA change threshold and the optimal time of collection with superior predictive value.

Trial design

This study was a single-center, real-world study to collect an estimated 56 patients from the Department of Respiratory and Critical Care Medicine, Southeast University Zhongda Hospital, who were diagnosed with inoperable stage IIIB to IV NSCLC and treated with standard chemo-immunotherapy from August 2022 to January 2024. Blood samples at baseline, within 2 days before medication in the 2nd and 3rd treatment cycles, and the time of progression were collected respectively for genetic testing. At the same time, clinical data such as blood counts, blood biochemical indexes, lymphocyte subpopulations, etc. were recorded, and patients were subsequently followed up to record anti-tumor efficacy such as objective responds rate (ORR), progression-free survival (PFS), overall survival (OS), and the occurrence of adverse events. Patients were categorized into durable clinical benefit group (DCB group) and non-durable benefit group (NDB group) according to whether the PFS was more than 8 months, and the predictive model was proposed to be constructed using the support vector mechanism.

Clinical trial identification

NCT05725915.

Legal entity responsible for the study

The authors.

Funding

Beijing Red Clove Public Welfare Development Center.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

51P - Phase 1B (Ph1b), MESOVAX clinical trial of pembrolizumab (P) and dendritic cell vaccine (DCvax) in advanced pleural and peritoneal mesothelioma (M): preliminary results

Presentation Number
51P
Lecture Time
16:15 - 16:15
Speakers
  • Laura Ridolfi (Meldola, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

M is a rare cancer with limited terapeutic options. Immunotherpy has shown hints of activity and, despite combination of immunotherapic agents is the new standard of care for non epithelioid tumors, further study are requested to improve therapeutic strategies. DCvax has shown activity in M with a good safety profile. Our data showed that DCvax induces the expression of PD-L1 on tumor cells. The addition of DC to P may sensitize patients to the effects of PD-1 blockade.

Methods

MESOVAX is a Ph1b study evaluating safety of P 200 mg Q3W and an autologous DCvax administered subcutaneously Q3W for a maximum of 6 cycles, in pretreated metastatic M patients (pts). Primary endpoint was safety; secondary were PD-L1 expression variations defined by immunohistochemistry (IHC) and efficacy (objective response rate [ORR], duration of response [DOR], progression-free survival [PFS], overall survival [OS]).

Results

As of 30 Aug 2023, 6 pts (median follow-up 20.1 mo) were treated and evaluable for safety pre-planned interim analysis. Median age was 64 yrs, all pts were male, 17%/83% were ECOG PS 0/1. All pts had epithelioid M. Any grade treatment-related adverse events (TRAEs) occurred in 5 pts (83%); most common were local reaction to injection site (4 pts, 67%), asthenia (2 pts, 33%) and fever (2 pts, 33%). None of the patients experienced grade 3–4 TRAEs. Concerning the treatment, 4 pts (67%) received 6 cycles of P+DC; 3 received maintenance P. In a preliminary analysis of tumor response, at the first planned evaluation (median treatment duration 2.1 mo),2 had SD, 1 had PR and 3 had PD. PD-L1 expression at baseline and at week 12 is summarized on Table.

PD-L1 expression defined by IHC at screening and at week 12

PDL1 expression
PT.N Screening Week12
2 NEG NEG
4 NEG ND
5 NEG NEG
11 NEG 70%
22 5% ND
23 2% NEG

Legend: ND: not done; NE: not evaluable.

Conclusions

Preliminary results of MESOVAX trial of P combined with DC showed a manageable toxicity and demonstrated encouraging preliminary efficacy. The trial is actively recruiting.

Clinical trial identification

EudraCT 2018-000500-42.

Legal entity responsible for the study

The authors.

Funding

Merck MSD.

Disclosure

F. De Rosa: Financial Interests, Personal, Speaker, Consultant, Advisor: MSD, Pierre Fabre, Novartis, Sun Pharma. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

52P - The dependence of TKI-resistant lung cancer cells on EGFR increases sensitivity to EGFR-CAR NK.

Presentation Number
52P
Lecture Time
16:15 - 16:15
Speakers
  • Sumei Chen (Hangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Lung cancer's multiple resistance to tyrosine kinase inhibitors is inexorable and often unexplained in many NSLCs with EGFR mutations or other oncogenes and epigenetic changes. This suggests the need for new treatment strategies.

Methods

We generated second-generation CD28-costimulated EGFR-specific CAR constructs using lentiviral vectors to modify NK92 cells. We evaluated the utility of NK92 cells expressing a CAR-EGFR against TKI-resistant lung cancer cells at a low effector-to-target ratio to allow possible escape or evolution of select lung cancer cells. EGFR expression was analyzed by flow cytometry.

Results

We evaluated the utility of NK92 cells expressing a CAR-EGFR against TKI-resistant lung cancer cells. Our approach used a low effector-to-target ratio to allow possible escape or evolution of select lung cancer cells. We found that Gefitinib-resistant lung cancer cells HCC827GR6 and TKI-sensitive HCC827 are equally attacked by NK92 cells. However, TKI-resistant HCC827GR6 were more sensitive to EGFR-CAR NK cell attacks than parental HCC827. Predictably, EGFR-CAR NK cell attacks led to reduced EGFR expression in HCC827 and growth without increasing resistance to naïve NK92 attacks. However, the EGFR-CAR NK cell attack caused an increase in EGFR expression in gefitinib-resistant HCC827GR6 cells with a slight reduction in growth. This paradoxical increased EGFR protein expression suggests a positive selection of mutant EGFR, reducing their chance of escaping new EGFR-CAR NK cell attacks. Indeed, in co-culture experiments of TKI-sensitive and TKI-resistant cells, the latter were more efficiently eliminated, suggesting the preferential killing of TKI-resistant lung cancer cells by EGFR-CAR NK.

Conclusions

Our data indicate that TKI-resistant lung cancer cells, which evolved to depend on the expression of a mutated EGFR, are targeted more efficiently by EGFR-CAR NK cells than TKI-sensitive cells.

Legal entity responsible for the study

Sumei Chen and Youssef Jounaidi.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

53P - Integrin-_v_6 targeted CAR T-cells in an immunocompetent orthotopic model of pancreatic cancer

Presentation Number
53P
Lecture Time
16:15 - 16:15
Speakers
  • Nicholas F. Brown (London, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Pancreatic cancer confers a dismal prognosis and better therapies are needed. Integrin αvβ6 is expressed in >90% of human pancreatic cancers with minimal expression in healthy tissues. αvβ6-targeted CAR T-cells are efficacious in heterotopic immunodeficient pancreatic models. However, these models do not recapitulate the profoundly immunosuppressive tumour microenvironment (TME) that is a hallmark of pancreatic cancer, and a barrier to effective therapy. This study aimed to create and evaluated αvβ6-targeted CAR T-cell therapies in an orthotopic, immunocompetent, metastatic model of PDAC.

Methods

We created a 2nd generation murine CAR with the αvβ6-targeting A20FMDV2 peptide in a γ-retroviral backbone, and transduced splenocytes from C57BL/6 mice. Non-targeting (NT) CAR T-cells lacking the A20FMDV2 peptide were used as controls. In vitro cytotoxicity was evaluated with murine pancreatic cancer cells with minimal (TB32043) and ectopic expression of αvβ6 (TB32043mb6s2) using WST-1 viability assays, with T-cell activation measured by IFNγ assays. In vivo activity of the CAR T-cells were assessed in both immunocompetent C57BL/6 and immunodeficient NSG mice who had received orthotopic injections of TB32043mb6s2 cells, with cyclophosphamide preconditioning in C57BL/6 mice. The immune tumour microenvironment of TB32043mb6s2 tumours was evaluated using CyTOF.

Results

αvβ6-targeted murine CAR T-cells demonstrated dose-depending αvβ6-specific in vitro cytotoxicity and T-cell activation. αvβ6 CAR T-cells were tolerated in vivo in mice with metastatic pancreatic cancer, and led to improved survival in both immunocompetent mice (31 vs 21 days, p<0.05 vs NT CAR T-cells) and immunodeficient mice (39 vs 27 days, p<0.05). CyTOF evaluation of αvβ6-expressing TB32043mb6s2 tumours found the TME comprised ∼90% of the tumour, with immune cells comprising ∼ half of the TME.

Conclusions

Integrin-αvβ6 targeted CAR T-cell therapy was tolerated and improved survival in mice with an orthotopic immunocompetent model of pancreatic cancer. Further pre-clinical development is warranted.

Legal entity responsible for the study

Queen Mary University of London.

Funding

Pancreatic Cancer UK, Cancer Research UK, Medical Research Council.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

55P - DPP9 promotes renal cancer PD-L1 expression through SHMT2-BRISC complex

Presentation Number
55P
Lecture Time
16:15 - 16:15
Speakers
  • Wei Zhang (Shanghai, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

In recent years, immunotherapy has attracted much attention in the treatment of kidney cancer. Immune checkpoints, especially PD-L1, play an important role in the treatment of kidney cancer. Therefore, the treatment of immune-infiltration and immunosuppression through immune checkpoints has become an important direction in treating renal cancer.

Methods

Several high DPP9 expression and low DPP9 expression tumor tissues were selected for single-cell sequencing. Results told us that DPP9 regulates T-cell infiltrating and transcription of PD-L1. We used co-IP assay to discover the interaction between DPP9 and SHMT2-BRISC. Then we established the overexpression/knockout DPP9 renal cell lines to detect the upstream signal pathway of PD-L1, co-cultured with PBMC cells to observe the influence of immune escape. Meanwhile, we used organoids to confirm the above experiments. Finally, we used DPP9 inhibitor and PD-L1 monoclonal antibody in animal experiments.

Results

Single-cell sequencing showed that T-cell exhaustion signal was significantly up-regulated in the group with high DPP9 expression. IHC staining of DPP9, PD-L1, CD3, PD-1 in kidney cancer section was verified. Overexpression/knocked out of DPP9 in kidney cancer cells such as 786-O, 769P, caki-1 showed that DPP9 could regulate the transcription and translation of PD-L1. Then co-IP showed that DPP9 interacted with SHMT2, BRE, FAM175B proteins in the BRISC complex. The binding level of DPP9 and SHMT2-BRISC complex in the presence or absence of IFN-γ was detected, and dynamic binding was sought to determine whether more FAM175B and SHMT2 were involved in the formation of complex in the presence of DPP9, thus regulating the transcription of PD-L1 in the condition of IFN-γ. PD-L1 monoclonal antibody can inhibit the immune escape of renal cancer induced by DPP9, and the effect is better when combined with DPP9 inhibitors.

Conclusions

DPP9 can up-regulate the expression of PD-L1 in renal cancer cells through dynamically adjusting the stability of the BRISC complex via SHMT2. We provide the clinical principle and mechanism of DPP9 inhibitor combined with PD-L1 monoclonal antibody, and further determine the patient population suitable for immunotherapy.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

56P - FOXM1D in T cells promotes the transcription of PD-1 by interacting with HCFC1 and regulating the killing of renal cancer cells

Presentation Number
56P
Lecture Time
16:15 - 16:15
Speakers
  • Yue Wang (Shanghai, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

FOXM1D functions through interactions with proteins. We found that FOXM1D decreased in peripheral blood immune cells of renal cancer patients. It is speculated that FOXM1D is likely to regulate the expression level of immune checkpoint in immune cells through the way of interprotein interaction, and regulating the transcription of PD-1.

Methods

We detected the FD level in PBMC and CD3-positive T cells in clinical samples. PD-1 expression was detected. We performed mass spectrometry and found that HCFC1 function as a cotranscription factor. We examined the effect of FOXM1D on the HCFC1 protein. We detected the HCFC1 in cell lines with overexpression and knockdown of FOXM1D by karyoplasmic isolation. YY1 is predicted to be a transcription factor of PD-1, ChIP and Luciferase experiments to detect the binding sites to the PD-1 promoter. JKT co-culture tests with renal cancer cells in supernatant cytokine levels, observe the killer T cells. Finally, we proceed animal testing.

Results

The FD level in PBMC of renal cancer patients was significantly lower than that of normal people. The change of FOXM1D in CD3+T cells was the same as PBMC. The transcription level of PD-1 and the level of PD-1 on the surface of Jurkat-FOXM1D in cells overexpressing FOXM1D was significantly down-regulated. HCFC1 was found to function as a cotranscription factor. In immune cells, FOXM1D inhibits the entry of HCFC1 into the nucleus by interacting with the N-terminal of HCFC1, weakens the transcriptional activation of HCFC1 to YY1, and then inhibits the transcriptional regulation of PD-1 molecules. After co-culture with T cells, the cytokine in supernatant of 786O and 769P cells was changed. And the animal experiment was in progress.

Conclusions

HCFC1, as a co-transcription factor, can enhance its transcriptional function by interacting with YY1, and further regulate the transcription of immune checkpoint molecules PD-1. FOXM1D inhibits the nuclear entry of HCFC1 by interacting with the N-terminus of HCFC1, attenuates the transcriptional activation of YY1 by HCFC1, and then inhibits the transcription of various immune checkpoint molecules such as PD-1.

Legal entity responsible for the study

Fudan University Shanghai Cancer Center.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

57P - Anatomical location of metastasis and composition of the final infusion product in metastatic melanoma (MM) patients treated with tumor-infiltrating lymphocytes (TIL)

Presentation Number
57P
Lecture Time
16:15 - 16:15
Speakers
  • Joachim Stoltenborg Granhøj (Herlev, Denmark)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Adoptive cellular therapy (ACT) with TIL recently showed promising results in a multicenter phase 3 trial evaluating TIL in MM patients (NCT02278887). In this, 39 (49 %) of TIL-treated patients obtained a complete or partial response according to RECIST 1.1. The potential impact of the location of the resected metastasis for TIL production on the phenotype of the final TIL infusion product is essential knowledge for future development of TIL therapy.

Methods

Cryopreserved samples of TIL infusion product from all patients with unresectable stage IIIC-IV MM treated in the phase 3 trial were analyzed using multiparametric flow cytometry. Here, we investigated if the anatomical location of the metastasis used for TIL production impacted the composition of the final TIL infusion product. The Kruskal-Wallis test was used to test for statistical significance.

Results

In total, 80 patients were treated with TIL. Respectively, 37 (46.3 %), 29 (36.3 %), and 14 (17.5 %) underwent resection of a lymph node, (sub)cutaneous or a visceral metastasis for TIL production. Visceral metastases included lung (n=8), liver (n=2), spleen (n=1), small intestine (n=1), adrenal gland (n=1), and peritoneum (n=1). The location of the metastasis used for TIL production did not impact clinical response (p=0.84) or the composition of the final TIL infusion product, with no difference in the total number of TIL produced in major subsets including CD45+ CD3+ (p=0.42), CD8+ TCRab+ (p=0.37), CD4+ TCRab+ (p=0.09) or CD3+ TCRgd+ (p=0.36). For both CD4+ and CD8+ TIL, differentiation into naïve (CCR7+ CD45RO-), effector memory (CCR7- CD45RO+), central memory (CCR7+ CD45RO+), and terminally differentiated effector memory cells (CCR7- CD45RO-) was also not affected by the site of metastasectomy.

Conclusions

The anatomical location of the metastasis used for TIL production did not impact clinical response, the absolute number of TIL produced, or the overall phenotypic composition of the final TIL infusion product. Thus, easily accessible MM lesions can be preferred without the risk of impacting TIL phenotype or clinical outcome.

Clinical trial identification

NCT02278887, EudraCT 2013-005406-54.

Legal entity responsible for the study

National Center for Cancer Immune Therapy (CCIT-DK), Herlev Hospital (Denmark) and the Netherlands Cancer Institute (NKI), Amsterdam (the Netherlands).

Funding

This study was supported by the Dutch Cancer Society, the Netherlands Organization for Health Research and Development, the Dutch Ministry of Health, Stichting Avento, AVLFoundation, Copenhagen University Hospital, Herlev, the Danish Cancer Society and Capital Region of Denmark Research Foundation.

Disclosure

T.H. Borch: Other, Personal, Other, Speaker's honoraria: Bristol Myers Squibb. W. Van Houdt: Financial Interests, Institutional, Invited Speaker: Amgen; Financial Interests, Institutional, Advisory Board: Belpharma, Sanofi, MSD; Financial Interests, Personal, Other, travel grant: Novartis; Financial Interests, Institutional, Local PI: BMS. M. Donia: Financial Interests, Personal, Other, Advisor: Achilles Therapeutics; Non-Financial Interests, Personal, Other, Sub-investigator of clinical trial with connected translational research: Bristol Myers Squibb; Non-Financial Interests, Personal, Proprietary Information, Proprietary data access: Bristol Myers Squibb; Non-Financial Interests, Personal, Proprietary Information, Proprietary Data Access: Genentech; Other, Personal, Other, Chairman of the Melanoma and Non-melanoma Skin Cancer Scientific Committee: Danish Medicines Council (Medicinrådet). J.B.A.G. Haanen: Financial Interests, Institutional, Advisory Board: Bristol Myers Squipp, Achilles Therapeutics, Ipsen, Merck Sharpe & Dohme, Merck Serono, Pfizer, Molecular Partners, Novartis, Roche, Sanofi, Iovance Biotherapeutics, AstraZeneca; Financial Interests, Institutional, Advisory Board, SAB member: BioNTech, Immunocore, Gadeta, Instil Bio, PokeAcel, T-Knife; Financial Interests, Personal, Advisory Board, SAB member: Neogene Therapeutics, Scenic; Financial Interests, Personal, Advisory Board: Third Rock Venture, CureVac, Imcyse; Financial Interests, Personal, Stocks/Shares: Neogene Therapeutics; Financial Interests, Institutional, Research Grant: Bristol Myers Squibb, BioNTech US, Merck Sharpe & Dohme, Amgen, Novartis, Asher Bio, Sastra Cell Therapy; Non-Financial Interests, Personal, Member: ASCO, AACR, SITC; Other, Personal, Other, Editor-in-Chief IOTECH: ESMO; Other, Personal, Other, Editorial Board ESMO Open: ESMO; Other, Personal, Other, Editorial Board: Kidney Cancer. I. Svane: Financial Interests, Personal, Advisory Board: BMS, Pierre Fabre, Novartis; Financial Interests, Personal, Invited Speaker: MSD, Pierre Fabre, Novartis, Roche, BMS; Financial Interests, Institutional, Research Grant: Enara Bio, Adaptimmune, Lytix Biopharma, TILT Biotherapeutics; Financial Interests, Personal, Writing Engagement: MSD; Financial Interests, Personal, Stocks/Shares, Cofounder and Founder warrents: IO Biotech; Financial Interests, Institutional, Funding: Evaxion; Non-Financial Interests, Personal, Principal Investigator: BMS, Novartis, Roche, TILT Biotherapeutics, Lytix Biopharma. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

58P - Natural high-avidity T-cell receptor efficiently mediates regression of cancer/testis antigen 83 positive common solid cancers

Presentation Number
58P
Lecture Time
16:15 - 16:15
Speakers
  • Liangping Li (Guangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

T-cell receptor-engineered T cells (TCR-Ts) have achieved encouraging success in anticancer clinical trials. The antigenic targets, however, were primarily focused on a few cancer/testis antigens (CTAs) which are not widely expressed in common solid cancers; the tested T-cell receptors (TCRs) were frequently from tumor-infiltrating lymphocytes of old patients and were not assured to have higher avidity. Here, we propose the isolation of high-avidity TCRs against CTAs that are frequently expressed in common solid cancers.

Methods

CT83 protein, which is frequently expressed in common solid cancers, was as a model antigen for screening of its specific TCR. The predicted CT83 epitopes with binding to HLA-I molecules, popular in the Chinese population, were integrated into three synthetic long peptides. CT83 reactive CD8+ T cells were stimulated with peptide-loaded dendritic cells (DCs) and sorted using the CD137 biomarker for single-cell sequencing to obtain the paired TCRαβ sequence.

Results

CT83 reactive T cells from young healthy donors (YHDs) were generated by repeated stimulation with DCs and peptides. The single-cell TCR sequencing results of reactive T cells indicated that a single TCR clonotype dominated the paired TCRs. T cells engineered with this dominant TCR led to HLA-A*11:01-restricted recognition of the CT8314-22 epitope, with higher avidity. Functional assays showed powerful cytotoxicity in vitro against the targets of several CT83-positive solid cancer cell lines. Furthermore, TCR-Ts showed therapeutic efficacy in three xenograft solid tumor models. The meta-analysis of gene expression of 92 CTAs indicated that most CTAs did not or at low levels in the thymus, which suggested that those CTAs may experience incomplete thymic central tolerance.

Conclusions

High-avidity TCR against CT83 could be isolated from YHDs and efficiently mediate regression of well-established xenograft common solid tumors. The high-avidity TCR repertoire in the peripheral blood of some donors for CT83 and other CTAs provides the basis for the efficient isolation of high-avidity TCRs to target numerous solid cancers.

Legal entity responsible for the study

The author.

Funding

National Key Research & Development Projects of China.

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

59P - Revolutionizing cell therapy testing by co-culturing 3D patient derived cancer models and circulating immune cells on Organ-on-chip platform

Presentation Number
59P
Lecture Time
16:15 - 16:15
Speakers
  • Silvia Scaglione (Genova, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Cell therapy has emerged as a promising approach in cancer treatment, leveraging the potential of the patient's immune system to fight tumors. However, the assessment of the efficacy and safety of these therapies remains a significant challenge, since the current available approaches (i.e. 2D cell cultures and animals) have strong limitations in terms of predictability, reliability, and ability to fully mimic the human immune system. This study presents a groundbreaking technological approach in cell therapy testing by integrating 3D patient-derived cancer models with circulating immune cells co-cultured onto an Organ-on-Chip (OoC) platform.

Methods

Biologically relevant cancer samples have been optimized by using an alginate-based structure closely resembling the tumor extracellular matrix. Different cancer cell lines (i.e. MDA-MB-231, SKOV-3, HTLA-230) have been embedded in the matrix and cultured up to 2 months under fluid-dynamic conditions with a OOC chamber, simulating the bloodstream. Alternatively, patient derived tumor biopsies have been coated by a thin layer of alginate, to enhance their structural stability over time, and cultured ex vivo in the same OOC. At the same time, peripheral blood mononuclear cells have been injected in the circulatory OOC circuit and their extravasation and tumor infiltration analysed.

Results

While tumor cells are able to maintain a good viability, cytoskeleton reorganization and migration within the polymeric matrix up to 2 months of culture, patient derived biopsies displayed a challenging survival ex vivo, although the presence of the fluid flow was able to improve the tumor cells survival. The immune checkpoint ligands PD-L1 and PD-L2 were successfully upregulated by the presence of the IFN-gamma and 3% of PBMC derived natural killer cells were able to leave the circulatory flow, and infiltrate the tumor matrix where they induce apoptosis.

Conclusions

A novel fully humanized OOC based platform has been developed to co-culture clinically relevant human cancer model, while immune cells are in circulation, with the final aim to deepen insights into the crosstalk among immune /tumor cells and test cellular therapies in a reproducible and reliable way.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

S. Scaglione, M. Aiello: Financial Interests, Personal, Member of Board of Directors: React4life. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

60P - Adaptive NK cells as a therapeutic option for childhood leukaemia

Presentation Number
60P
Lecture Time
16:15 - 16:15
Speakers
  • Zoya Eskandarian (Hamburg, Germany)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Natural killer (NK) cells are promising tools for immunotherapy of different malignancies including childhood leukaemia and are shown to exhibit memory-like features. Epigenetic alterations including DNA methylation contribute to the generation of memory in these cells. Studying the DNA methylation profile of activated NK cells helps identify candidate genes of activation biomarkers and new approaches to enhance NK cytotoxicity by targeting intrinsic epigenetic regulators.

Methods

We investigated the DNA methylation profile in NK cells upon priming with activating cytokines. Basically, two types of peripheral blood NK cells were used in this study: Freshly antibody-bead isolated NK cells and expanded NK cells, generated by a 7-day coculture with irradiated K562 cells expressing membrane bound IL-21. Memory-like features were achieved by 16-h stimulation of NK cells with IL-12, IL-15, and IL-18, as previously published. The activation of NK cells was confirmed by higher production of Interferon-γ in preactivated cells. DNA was extracted from the FACS sorted NK cells, based on NK cells markers, in both experimental groups and were analysed by Illumina methylation bead array.

Results

Downstream bioinformatic analyses revealed differential methylation patterns with hypomethylated CpG sites predominantly in preactivated NK cells and in expanded NK cells, independent of cytokine pre-activation.

Conclusions

Our data suggest that the in vitro expansion of NK cells in coculture with K562 cells at least partially recapitulates the memory formation by cytokine-preactivation due to the encounter with K562 cell surface targets prior to being stimulated with cytokines resulting in DNA hypomethylation at a variety of loci in some genes encoding immune system ligands, receptors and regulators such as IL-5, C-C Motif Chemokine Receptor 5 (CCR5) and Programmed Cell Death Ligand 1 (PDL1). Targeted therapy of JAK-class Ph-like ALL with ruxolitinib might interfere with interleukin-signaling pathways in NK cells disrupting memory-formation and cytotoxic activities as shown by live cell imaging and multiparametric full spectrum flow cytometry.

Legal entity responsible for the study

The authors.

Funding

Research institute Children’s Cancer Center and Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

61P - Unlocking the Power of Natural Killer Cells: Precision Selection with Cutting-Edge Microfluidics

Presentation Number
61P
Lecture Time
16:15 - 16:15
Speakers
  • Neelima KC (London, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Natural killer (NK) cells may be particularly suited to cancer therapy. However, the heterogeneity of NK cell populations poses challenges in isolating highly functional subpopulations for therapeutic applications. This study presents a novel microfluidic platform that can select NK cells with enhanced avidity, aiming to improve the precision and efficacy of NK cell-based immunotherapies.

Methods

We used a microfluidics device that uses fluid shear stress to sort and isolate and sort NK cells based on their avidities to the target cells. Briefly, we sterilized our chip and coated it with fibronectin overnight and cultured lung cancer (A549) and ovarian cancer (SKOV3) cells for 24 hours until they achieved a monolayer. We then introduced NK cells on top of the monolayer and left these two types of cells to interact under static condition for 10 minutes. Using syringe pump, we applied fluid flow to induce shear ranging from 0.5Pa to 19.8 Pa in order to isolate the NK cells after each flow. To explore if avidity correlated with cytotoxicity, we cultured monolayer of A549 on our chip then introduced two types of NK cells: treated NK cells (adNK) and NK cells isolated from peripheral blood (cbNK). We followed the same protocol as described above to determine the NK cell type with high avidity.

Results

Our results show that at least 4.5 % and 4% cbNK cells remained attached to SKOV3 cells and A549 respectively after exposure to shear as high as 19.8 Pa. This avidity profile was different for adNK cells on A549. 30% of and NK cells remained attached to A549 cells after 19.8 Pa shear. This result also matched the cytotoxicity profile of adNK cells compared to that of cbNk cells for A549 cells.

Conclusions

In this study we used a novel method to sort and isolate NK cells based on their avidity to its target cancer cells. Although this device has been used to isolate and sort T cell population before, to our knowledge, for the first time we showed that NK cells can be isolated using their avidity to target cells. In addition, the higher avidity adNk cell aligned with its high cytotoxicity suggesting that selecting NK cells based on avidity may lead to selection of potent NK cells.

Legal entity responsible for the study

Imperial College London.

Funding

Imperial College London, Department of Bioengineering Imperial College London, Department of Life Sciences.

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

63TiP - A phase I study of tumor-infiltrating lymphocytes (TILs) in advanced solid tumors used an optimized regimen: MIZAR trial

Presentation Number
63TiP
Lecture Time
16:15 - 20:30
Speakers
  • Qing Xu (Shanghai, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The clinical usefulness of tumor-infiltrating lymphocyte (TIL) has been limited due to an intensive lymphodepletion regimen and high-dose intravenous interleukin-2 (IL-2) administration. We explore the low dose regimens of lymphodepletion and infusion without IL-2 administration in TIL therapy, which showed encouraging outcome in a case report. Thus, a phase I study to evaluate the safety and efficacy of optimized TIL-therapy regimen for the treatment of advanced solid tumors was conducted.

Trial design

The phase 1a part used a conventional 3+3 dose-escalation design. The primary endpoint in the phase 1a part was the frequency of dose-limiting toxicities (DLTs). Patients received three consecutive daily infusions of cyclophosphamide (25 mg/kg/day) from day -5 to day -3, and oral administration of hydroxychloroquine (600 mg once) on day -5. On day 0, patients received a single intravenous adoptive transfer of TILs following the administration of anti-PD-1 antibody (100mg per patient, sintilimab). Adverse events (AEs) were assessed based on Common Terminology Criteria for Adverse Events (CTCAE), version 5.0. Clinical responses were assessed according to Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1. Three patients were planned for DLT analysis at each dose level of TILs infusion: level 1 (5.0×109± 20% cells), level 2 (1.5×1010± 20% cells), level 3 (3.0×1010± 20% cells), and level 4 (4.5×1010± 20% cells). As of September 15, 2023, ten participants (9/10 with PD-1 antibody resistance) have been enrolled and underwent TIL infusion. None DLT was observed and the clinical responses were observed across different dose levels, suggesting the modified regimen is safe and the recommended dose (RD) of TILs can be defined within a broad range. The phase 1b part would be started in soon future.

Clinical trial identification

NCT05417750.

Legal entity responsible for the study

Shanghai Juncell Therapeutics Co., Ltd.

Funding

Shanghai Juncell Therapeutics Co., Ltd.

Disclosure

H. Jin: Financial Interests, Institutional, Funding: Shanghai Juncell Therapeutics Co., LTD. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

62P - Solupore® is a novel platform used for the delivery of gene editing tools and modalities, yielding healthier edited primary immune cells with superior function compared with electroporation.

Presentation Number
62P
Lecture Time
20:30 - 20:30
Speakers
  • Jamie Frankish (Maynooth, Ireland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

68P - Real-world (rw) outcomes in patients (pts) with metastatic (m) NSCLC and STK11, KEAP1 and/or KRAS mutations (mut) receiving PD-(L)1-based treatment (tx): CORRELATE

Presentation Number
68P
Lecture Time
20:30 - 20:30
Speakers
  • Solange Peters (Lausanne, Switzerland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

PD-(L)1 inhibitors ± chemotherapy (CT) have demonstrated survival benefit vs CT in mNSCLC RCTs. However, STK11mut and KEAP1mut tumours have been characterised as immunologically ‘cold’ and are frequently associated with a concomitant KRASmut. One aim of CORRELATE was to describe rw outcomes of pts with mNSCLC and STK11, KEAP1 and/or KRAS mut receiving tx with anti-PD-(L)1 ± CT.

Methods

This analysis included pts from the US Flatiron Clinico-Genomic Database who started 1L anti-PD-(L)1 tx for mNSCLC between 1 Nov 2016 and 31 May 2021, and met eligibility criteria based on 4 RCTs for US-approved tx (KEYNOTE [KN]-024, n=94; KN-189, n=462; KN-407, n=122; IMpower150, n=4). OS and rwPFS were estimated in subgroups based on STK11, KEAP1 and KRAS mut status by Kaplan-Meier analysis. Unadjusted HRs were calculated by univariate Cox regression and adjusted HRs by multivariable Cox regression.

Results

Of 682 pts, 44% had ≥1 mut of interest. Among pts with ≥1 mut, there were more smokers, pts with ECOG PS 2 or ≥3, and pts with non-squamous tumours vs pts with no mut (each nominal p<0.05). There was a trend towards increased risk of death in pts with 1 mut vs no mut; pts with co-mut (≥2 mut) had almost 2x increased risk of death vs no mut, both with and without adjustment for pt characteristics (Table). Pts with STK11mut and pts with KRASmut (± co-mut) had a 46% and 28% increased risk of death vs STK11wt and KRASwt, respectively, after adjustment (Table). After adjusting for potential confounders, the 26% increased risk of death among pts with KEAP1mut vs KEAP1wt was not statistically significant. Trends were similar for rwPFS.

N Median OS; 95% CI (mo) Unadjusted HR; 95% CI Adjusted HR; 95% CId
No mut 382 18.2; 15.0–21.1
1 mut 214 15.3; 11.6–21.2 1.12; 0.90–1.39b 1.20; 0.95–1.51b
Co-mut 86 8.3; 6.0–9.9 1.96; 1.49–2.58b 1.97; 1.47–2.64b
STK11wt 579 17.6; 14.7–20.1
STK11muta 103 8.9; 6.5–12.5 1.78; 1.39–2.29c 1.46; 1.12–1.92c,e
KEAP1wt 596 16.3; 14.1–18.3
KEAP1muta 86 9.9; 7.1–14.5 1.37; 1.05–1.80c 1.26; 0.95–1.68c,f
KRASwt 470 16.7; 14.6–19.4
KRASmuta 212 11.6; 8.9–15.3 1.22; 1.00–1.49c 1.28; 1.03–1.59c,g

a± co-mut. bvs no mut. cvs wt. dAdjusted for age, sex, smoking status, ECOG PS, histology, PD-L1 status, no. of baseline metastases; also adjusted for: eKEAP1 & KRAS status, fSTK11 & KRAS status, gSTK11 & KEAP1 status

Conclusions

In US pts with mNSCLC receiving 1L anti-PD-(L)1 ± CT, those with mut in STK11, KEAP1, KRAS, and particularly co-mut, had worse OS and rwPFS. These results highlight the need for novel combination tx approaches in these pts that might further boost immune responses (e.g., incorporating anti-CTLA-4) and optimise outcomes.

Editorial acknowledgement

Medical writing support for the development of this abstract, under the direction of the authors, was provided by Samantha Holmes, DPhil, of Ashfield MedComms (Macclesfield, UK), an Inizio company.

Legal entity responsible for the study

AstraZeneca PLC.

Funding

AstraZeneca.

Disclosure

S. Peters: Financial Interests, Institutional, Invited Speaker: AiCME, AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, ecancer, Eli Lilly, Foundation Medicine, GSK, Illumina, Imedex, Ipsen, Medscape, Merck Sharp and Dohme, Mirati, Novartis, PER, Peerview, Pfizer, Roche/Genentech, RTP, Sanofi, Takeda; Financial Interests, Institutional, Advisory Board, Consultation/Advisory role: AbbVie, AiCME, Amgen, Arcus, AstraZeneca, Bayer, BeiGene, BerGenBio, Biocartis, BioInvent, Blueprint Medicines, Boehringer Ingelheim, Bristol Myers Squibb, Clovis, Daiichi Sankyo, Debiopharm, ecancer, Eli Lilly, Elsevier, F-Star, Fishawack; Financial Interests, Institutional, Member of Board of Directors: Galenica; Financial Interests, Institutional, Principal Investigator: Amgen, Arcus, AstraZeneca, BeiGene, Bristol Myers Squibb, GSK, iTeos, Merck Sharp and Dohme, Mirati, Pharma Mar, Promontory Therapeutics, Roche/Genentech, Seattle Genetics; Non-Financial Interests, Personal, Member: ESMO, ASCO, AACR, IASLC, SSOM, SAKK, ETOP; Non-Financial Interests, Personal, Advisory Role: Cf. advisory boards; Non-Financial Interests, Personal, Leadership Role: Vice President Swiss Cancer League, past President ESMO, Strategic Advisory board SPCC (Paris Saclay) Chair, ETOP Scientific Chair; Financial Interests, Institutional, Advisory Board: Foundation Medicine, Genzyme, Gilead, GSK, Hutchmed, Illumina, Imedex, IQVIA, Incyte, Ipsen, iTeos, Janssen, Medscape, Medtoday, Merck Sharp and Dohme, Merck Serono, Merrimack, Mirati, Novartis, Novocure, OncologyEducation, Pharma Mar, Promontory Therapeutics, PER, Peerview, Pfizer, Regeneron, RMEI, Roche/Genentech, RTP, Sanofi, Seattle Genetics, Takeda, Vaccibody. R.J. Salomonsen: Financial Interests, Personal, Full or part-time Employment: AstraZeneca; Financial Interests, Personal, Stocks/Shares: AstraZeneca; Non-Financial Interests, Personal, Project Lead: AstraZeneca. F. Skoulidis: Financial Interests, Personal, Invited Speaker: ESMO, Japanese Lung Cancer Society, Medscape Llc, Intellisphere Llc, VSPO McGill Universite de Montreal, RV Mais Promocao Events LTDS, MJH Life Sciences, IDEOlogy Health, MI&T, PER Llc, CURIO Llc, DAVA Oncology, American Association for Cancer Research, IASLC; Financial Interests, Personal, Advisory Board: AstraZeneca, Amgen Inc, Revolution Medicines, Novartis, BridgeBio, BeiGene, BergenBio, Guardant Health, Tango Therapeutics, Calithera Bioscieces, Hookipa Pharma, Novocure, Merck & Co; Financial Interests, Personal, Stocks/Shares: BioNTech SE, Moderna Inc; Financial Interests, Institutional, Research Grant: Revolution Medicines, Mirati Therapeutics, Amgen Inc, Novartis, Merck & Co; Financial Interests, Personal, Principal Investigator: Amgen Inc, AstraZeneca, Revolution Medicines, Novartis, Merck & Co, Tango Therapeutics, Genentech/Roche; Financial Interests, Personal, Advisory Role: AstraZeneca. I. Diaz Perez: Financial Interests, Personal, Full or part-time Employment: AstraZeneca; Financial Interests, Personal, Stocks/Shares: AstraZeneca. A. Wang: Financial Interests, Personal, Full or part-time Employment, Contracted to AstraZeneca: AstraZeneca. M. Cooper: Financial Interests, Personal, Full or part-time Employment: AstraZeneca; Financial Interests, Personal, Stocks/Shares: AstraZeneca. S.V. Liu: Financial Interests, Personal, Advisory Board: AbbVie, Amgen, AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Catalyst, Daiichi Sankyo, Eisai, Elevation Oncology, Genentech/Roche, Gilead, Guardant Health, Janssen, Jazz Pharmaceuticals, Merck, Merus, Mirati, Novartis, Regeneron, Sanofi, Takeda, Turning Point Therapeutics; Financial Interests, Institutional, Research Grant: AbbVie, Alkermes, Elevation Oncology, Ellipses, Genentech, Gilead, Merck, Merus, Nuvalent, RAPT, Turning Point Therapeutics.

Collapse
Poster Display (ID 34) Poster Display

70P - LIST (Lung Initiative on Sequence Therapy), a real-world study of nivolumab for advanced NSCLC in France: first effectiveness, safety, and IO-rechallenge results

Presentation Number
70P
Lecture Time
20:30 - 20:30
Speakers
  • Benoît GODBERT (Vantoux, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

NSCLC treatment has evolved after approval of the 1st immunotherapy agent (IO) in 2015. The real-world LIST study of nivolumab for advanced NSCLC aims to describe patient/treatment characteristics, effectiveness, safety and rechallenge outcomes.

Methods

LIST is an ongoing longitudinal, prospective, observational study in patients (pts) with advanced NSCLC receiving nivolumab after one prior chemotherapy-based treatment alone or in combination with IO. Patient cohorts were: Cohort 1, IO-naïve patients; Cohort 2, IO-experienced patients who discontinued for non-IO-toxicity reasons; Cohort 3, IO-experienced patients who discontinued due to IO-toxicity. Descriptive interim analyses (follow-up ≥6 months) are presented.

Results

428 pts were enrolled between Sept 2020 and May 2022 at 99 centres – 278, 128 and 22 pts in cohorts 1, 2 and 3. Baseline patient characteristics were representative of advanced NSCLC: median age 65–70 years, male sex >65%, current/former smoker 22–27%/65–73%, non-squamous histology 60–75%. 41%/ 71%/ 76% of pts in cohorts 1/2/3 had PD-L1 expression (≥1%). At 6 months, respective overall survival (OS) rates were 70.0% [95% confidence interval 64.2–75.1%]/ 61.1% [51.6–69.3%]/ 61.2% [37.1–78.4%] and progression-free survival (PFS) were 33.4% [27.6–39.2%]/ 25.1% [17.2–33.8%]/ 51.0% [28.1–70.0%]. Current/former smokers and pts with tumour PD-L1 expression seem to have improved outcomes (table). All adverse events (AEs) occurred in 86%/ 78%/ 73% of pts and treatment-related grade 3/4 AEs in 11%/ 5%/ 18%, in cohorts 1/ 2/ 3.

Interim survival data by cohort and smoking and PD-L1 status

Cohort 1 (n=278) Cohort 2 (n=127) Cohort 3 (n=22)
Median, months Rate at 6 months, % Median, months Rate at 6 months, % Median, months Rate at 6 months, %
OS
Overall 12.2 70.0 8.3 61.1 10.4 61.2
Never smoker 8.1 62.9 9.5 77.8
Former smoker 12.2 72.4 6.7 54.6 10.4 58.7
Current smoker 12.6 66.5 11.0 76.0
PD-L1 <1%
PD-L1 1–49% 12.2 63.6 6.7 53.5
PD-L1 ≥50% 7.3 61.1 9.6 60.7
PFS
Overall 3.2 33.4 2.9 25.1 7.6 51.0
Never smoker 3.0 24.0 - -
Former smoker 3.9 35.6 2.2 20.6 3.2 46.7
Current smoker 2.7 30.9 3.9 36.6
PD-L1 <1%
PD-L1 1–49% 3.5 42.3 2.2 16.0
PD-L1 ≥50% 1.8 16.7 2.1 24.8
– = n<15

Conclusions

LIST interim results show nivolumab for IO-naïve pts in clinical practice has similar activity and safety as in the registrational CheckMate 017/057 trials. It shows encouraging results of rechallenge with nivolumab in pts with previous IO discontinuation, including for IO-toxicity.

Clinical trial identification

NCT04500535.

Editorial acknowledgement

We thank Sheridan Henness who provided medical writing assistance prior to submission on behalf of Springer Healthcare Communications.

Legal entity responsible for the study

The authors.

Funding

Bristol Myers Squibb.

Disclosure

B. Godbert: Financial Interests, Personal, Speaker, Consultant, Advisor: Bristol Myers Squibb, AstraZeneca, Sanofi, MSD; Financial Interests, Personal, Advisory Board: Bristol Myers Squibb, AstraZeneca, MSD, Sanofi; Financial Interests, Personal, Expert Testimony: Bristol Myers Squibb, AstraZeneca, Sanofi, MSD. M. Zysman: Financial Interests, Institutional, Other: AVAD; Financial Interests, Personal, Speaker, Consultant, Advisor: CSL Behring, GSK, Boehringer Ingelheim, AstraZeneca, Chiesi, Sanofi; Financial Interests, Personal, Other: Chiesi, AstraZeneca, GSK. E. Gobbini: Financial Interests, Institutional, Other: BMS; Financial Interests, Personal, Other: AstraZeneca, Roche, Pfizer, Merck Sharpe and Dohm, Bristol Myers Squibb, Takeda, Janssen, Sanofi. C. Decroisette: Financial Interests, Personal, Speaker, Consultant, Advisor: MSD, BMS, Takeda, Pfizer, Sanofi, Janssen, AstraZeneca, Amgen; Financial Interests, Personal, Other: Amgen, Janssen, Roche, MSD. H. Lena: Financial Interests, Personal, Speaker, Consultant, Advisor: Roche AstraZeneca, MSD, Novartis, Takeda, BMS, Roche, MSD, Pfizer, Lilly, Amgen; Financial Interests, Personal, Other: BMS, Takeda, Pfizer. D. Moro-Sibilot: Financial Interests, Personal, Speaker, Consultant, Advisor: BMS, MSD, Roche, AstraZeneca; Financial Interests, Personal, Other: Roche, MSD; Financial Interests, Personal, Advisory Board: BMS, Roche, MSD; Financial Interests, Institutional, Other: BMS, Roche, MSD. F. Guisier: Financial Interests, Personal, Advisory Board: Amgen, AstraZeneca, BMS, MSD, Roche, Sanofi, Janssen, Pfizer, Takeda; Financial Interests, Institutional, Research Grant: Pfizer, Roche, Takeda. S. Couraud: Other, Personal, Advisory Board: Adene; Financial Interests, Personal and Institutional, Advisory Board, Institution Funding: Amgen, BMS, MSD, Novartis, Pfizer, Sanofi; Financial Interests, Personal and Institutional, Advisory Board, Institution Funding, Research Funding: AstraZeneca; Financial Interests, Institutional, Research Funding: BD, Transdiag, Volition; Financial Interests, Institutional, Funding: Celgene, Chugai, Janssen, Laidet, Lilly, Roche, Takeda; Financial Interests, Personal, Speaker, Consultant, Advisor: Health EvenT; Financial Interests, Personal, Advisory Board: MaaT Pharma. A. Caraux: Financial Interests, Personal, Affiliate: Bristol Myers Squibb. D. Reynaud: Financial Interests, Personal Boehringer Ingelheim, Affiliate: Bristol Myers Squibb. F. Breliier: Financial Interests, Personal, Affiliate: Bristol Myers Squibb; Financial Interests, Personal, Stocks or ownership: Bristol Myers Squibb. N. Girard: Financial Interests, Personal, Invited Speaker: AstraZeneca, BMS, MSD, Roche, Pfizer, Mirati, Amgen, Novartis, Sanofi, Gilead; Financial Interests, Personal, Advisory Board: AstraZeneca, BMS, MSD, Roche, Pfizer, Janssen, Boehringer Ingelheim, Novartis, Sanofi, AbbVie, Amgen, Lilly, Grunenthal, Takeda, Owkin, Leo Pharma, Daiichi Sankyo, Ipsen; Financial Interests, Institutional, Research Grant, Local: Roche, Sivan, Janssen; Financial Interests, Institutional, Funding: BMS, Leo Pharma; Financial Interests, Institutional, Research Grant: MSD; Non-Financial Interests, Personal, Officer, International Thymic malignancy interest group, president: ITMIG; Other, Personal, Other, Family member is an employee: AstraZeneca. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

72P - Camrelizumab plus apatinib after chemoradiotherapy in unresectable stage III non-small-cell lung cancer?A multi-center, single-arm, phase 2 study

Presentation Number
72P
Lecture Time
20:30 - 20:30
Speakers
  • Hui Zhouguang (Beijing, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The PACIFIC trial established consolidation therapy with durvalumab as the standard of care for patients (pts) with unresectable stage III non-small cell lung cancer (NSCLC) without progression after chemoradiotherapy (CRT), however, additional studies are still needed to further explore the optimal therapeutic strategies. Here, we reported the preliminary results of the phase 2 study of camrelizumab (an anti-PD-1 antibody) plus apatinib (an anti-angiogenic inhibitor targeting VEGFR2) as consolidation therapy after CRT in unresectable stage III NSCLC.

Methods

This was a multi-center, single-arm, phase 2 study done at seven sites in China. Adult pts with pathologically confirmed unresectable stage III NSCLC, an ECOG PS of 0 or 1, and no disease progression following platinum-based concurrent or sequential CRT received camrelizumab (200 mg, i.v., q3w) and apatinib (250 mg, orally, qd) until disease progression, unacceptable toxicity or for up to 12 months. The primary endpoint was progression-free survival (PFS).

Results

Between March 17, 2021 to July 17, 2023, 42 pts were enrolled. The median age was 61 years (range 26-78), with 39 pts (92.9%) being male and 31 (73.8%) having squamous NSCLC. According to RECIST 1.1, three (7.1%) of the 42 pts had a complete response and eight (19.0%) had a partial response, with the confirmed objective response rate of 26.2% (95% CI 13.9-42.0). Twenty-three pts (54.8%) had stable disease, with the disease control rate of 81.0 % (95% CI 65.9-91.4). As of August 28, 2023, the median follow-up was 11.4 months (95% CI 5.8-15.6). The median PFS was not reached (NR; 95% CI 11.4-NR), with the 6- and 12-month PFS rates of 88.8% (95% CI 72.8-95.7) and 68.6% (95% CI 47.4-82.7), respectively. The median overall survival was immature, with six deaths at data cutoff. Treatment-related adverse events (TRAEs) of grade 3-4 occurred in 18 pts (42.8%), with the most common being hypertension (21.4%), AST increased (7.1%) and hand-foot syndrome (4.8%). No grade 5 TRAEs occurred.

Conclusions

Camrelizumab plus apatinib as consolidation therapy showed preliminary activity in unresectable stage III NSCLC pts without disease progression after CRT, with no new safety signals.

Clinical trial identification

NCT04749394; February 11, 2021.

Legal entity responsible for the study

Cancer Hospital Chinese Academy of Medical Sciences.

Funding

Jiangsu Hengrui Pharmaceuticals Co., Ltd.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

69P - Impact of pembrolizumab duration and comedications on overall survival: analysis from 45 000 patients with advanced NSCLC

Presentation Number
69P
Lecture Time
20:30 - 20:30
Speakers
  • Adrien Rousseau (Villejuif, Cedex, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

73P - Camrelizumab plus famitinib as first-line treatment in patient with locally advanced or metastatic pulmonary sarcomatoid carcinomas (CAPSTONE)?A multi-center, single-arm, phase II study

Presentation Number
73P
Lecture Time
20:30 - 20:30
Speakers
  • Qian Chu (Wuhan, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Pulmonary sarcomatoid carcinoma (PSC) is a rare, poorly differentiated, highly invasive subtype of non-small cell lung cancer (NSCLC), with extremely poor prognosis. Immunotherapy and targeted therapy have become the mainstay of management of advanced NSCLC, however, have been scarcely reported in PSC. Herein, we conducted a multi-center, single-arm, phase II study to assess the efficacy and safety of camrelizumab plus famitinib as first-line treatment in pts with locally advanced or metastatic PSC.

Methods

In this study, treatment-naïve pts with histologically confirmed stage IIIB-IV PSC received camrelizumab (200 mg, i.v., q3w) plus famitinib (20 mg, orally, qd) until disease progression or intolerable toxicity. Simon's two-stage design was adopted, and 15 pts were planned to be enrolled in the first stage, with 3 or more responses observed to progress to the second stage. The primary endpoint was objective response rate (ORR) as per RECIST 1.1. Here, we reported the first-stage results.

Results

From August 4, 2021 to April 24, 2023, 15 pts were enrolled, with a median age of 64 years (range 45-72) and all being male (100%). Of the 15 pts, seven (46.7%) achieved a partial response and six (40.0%) had stable disease, with the confirmed ORR of 46.7% (95% CI 21.3-73.4) and disease control rate of 86.7% (95% CI 59.5-98.3). As of August 16, 2023, the median follow-up was 10.0 months (IQR 3.9-14.5). The median duration of response was 7.1 months (95% CI 5.0-NR), median progression-free survival was 7.8 months (95% CI 1.6-NR), and median overall survival was 18.2 months (95% CI 18.0-NR). The median number of camrelizumab cycles was 7.0 (range 1-19). Treatment-related adverse events (TRAEs) of any grade occurred in 15 pts (100.0%), with seven (46.7%) developing grade ≥3 TRAEs. The most common TRAEs were proteinuria (53.3%), hypertension, neutrophil count decreased, and platelet count decreased (46.7% each). Two pts had grade 5 AEs (unknown cause).

Conclusions

Camrelizumab plus famitinib as first-line therapy for PSC showed promising activity and acceptable safety during the first stage of this study. Enrollment for the second stage is ongoing.

Clinical trial identification

NCT04888429; May 17, 2021.

Legal entity responsible for the study

Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

74P - A single-center, Phase II study of surufatinib combined with toripalimab, pemetrexed(A), and platinum (P) in patients with advanced non-squamous non-small cell lung cancer (nsq-NSCLC)

Presentation Number
74P
Lecture Time
20:30 - 20:30
Speakers
  • Wen Feng Fang (Guangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Surufatinib (S, a small-molecule inhibitor of VEGFR1-3, FGFR1 and CSF-1R) has exhibited encouraging efficacy in NSCLC patients (pts) as 3rd line or futher therapy in a phase II trial (NCT04922658). The purpose of this study is to evaluate the efficacy and safety of S + Toripalimab (T, an anti-PD-1 antibody) + chemotherapy (AP) in nsq-NSCLC pts. Here, we report the preliminary results.

Methods

This single-arm, phase Ⅱ study (NCT05003037) has two cohorts. Naive advanced nsq-NSCLC pts without driver gene mutation were enrolled in cohort 1and pts with mutated driver genes failing with tyrosine kinase inhibitors (TKIs) entered cohort 2. Both cohorts received S (250mg, qd, po, adjusted by DLTs in 1st cycle) plus T (240mg, iv, d1, q3w, fixed dose) and AP (q3w). After 4 cycles followed by maintenance therapy with S plus T and A, q3w. Primary endpoint is PFS. Secondary endpoints include ORR, DCR, OS, and safety.

Results

Until Sep 5, 2023, 40 pts were enrolled in cohort 1 (median age 60 years, male 89.7%, TNM stage IV 100%, brain metastases (BMs) 48.7%). 25 pts were assigned to cohort 2 (median age 58 years, male 48%, TNM stage IV 96%). The most common mutated genes were EGFR (64%), HER2 (16%), and MET (8%). Most commonly used TKIs included osimertinib (36%), gefitinib (24%), and almonertinib (24%). RP2D of S was 250mg, po, qd, q3w in both cohorts (1/0 DLT occurred in 6 pts in cohort 1/2). Among pts with at least one post-baseline tumor assessment (n=38 in cohort 1, 24 in cohort 2), ORRs were 57.9% and 54.2%, DCRs were 94.7% and 95.8%. Median PFS was 10.2 months (95%CI 6.8, 13.4; BMs: 7.2m; non-BMs: not reached) in cohort 1, was not reached in cohort 2. The most common treatment-emergent adverse events (Total; Grade ≥3) in cohort 1 were diarrhea (67.5%; 2.5%), proteinuria (57.5%; 5.0%), and anemia (50.0%; 2.5%); in cohort 2 were decreased platelet count (56.0%; 32.0%), fatigue (48.0%; 0) and diarrhea (40.0%; 0).

Conclusions

Surufatinib plus toripalimab and AP showed promising anti-tumor activity and acceptable toxicity for the treatment of advanced nsq-NSCLC, whether the driver gene is mutated or not. The combination of the 4 agents might be a novel therapeutic option for advanced nsq-NSCLC.

Clinical trial identification

NCT05003037; Release date: December 8, 2021.

Legal entity responsible for the study

Sun Yat-sen University Cancer Center.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

71P - Real-world analysis of first-line maintenance (1LM) immunotherapy (IO) for nonsquamous (NSQ) advanced/metastatic non-small lung cancer (a/mNSCLC) without targetable mutations

Presentation Number
71P
Lecture Time
20:30 - 20:30
Speakers
  • Umit Tapan (Boston, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

75P - Patient-reported outcomes (PROs) of cemiplimab + chemotherapy in advanced non-small cell lung cancer (NSCLC): EMPOWER-lung 3 liver metastases subpopulation

Presentation Number
75P
Lecture Time
20:30 - 20:30
Speakers
  • Ana Baramidze (Tbilisi, Georgia)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

As part of key subgroup analyses of EMPOWER-lung 3 (NCT03409614), a double-blinded, randomized (2:1) phase 3 trial, an improvement in overall survival (OS) was observed with cemiplimab + chemotherapy (CEMI+CHEMO, n=47) vs placebo + chemotherapy (CHEMO, n=23) in patients with baseline liver metastases from advanced NSCLC (median OS: 14.4 vs 8.9 months; hazard ratio (HR): 0.61 (95% confidence interval (CI): [0.31, 1.20]). The safety results of patients with baseline liver metastases were generally similar to those of the overall study population. We conducted exploratory analyses to evaluate PROs within this subgroup of patients.

Methods

PROs were assessed at day 1 (baseline), the start of each treatment cycle (every 3 weeks) for the first 6 doses, and then at start of every third cycle, using the EORTC-QLQ-C30 and -QLQ-LC13 questionnaires. A mixed-effect model for repeated measures analysis was performed to compare CEMI+CHEMO vs CHEMO for all scales. Time to definitive clinically meaningful deterioration (TTD) was evaluated using Kaplan-Meier analysis and between-arm TTD comparisons were made using a log-rank test and proportional hazards model.

Results

Statistically significant delay in TTD was observed favouring CEMI+CHEMO over CHEMO for role functioning (HR: 0.28, 95% CI [0.10, 0.79]; p=0.011), cognitive functioning (0.18, [0.06, 0.55]; p=0.001), haemoptysis (<0.01, [<0.01, not calculable]); p=0.021), and alopecia (0.25, [0.09, 0.72]; p=0.007). When comparing between arms, no statistically significant differences between CEMI+CHEMO vs CHEMO in overall change from baseline across all C30 or LC13 scales were observed. No analyses yielded statistically significant PRO results favouring CHEMO vs CEMI+CHEMO for any C30 or LC13 scales.

Conclusions

Among patients with baseline liver metastases from advanced NSCLC, CEMI+CHEMO resulted in significant delay in TTD in role functioning, cognitive functioning, haemoptysis, and alopecia when compared with CHEMO. Overall change from baseline across all PROs were maintained. These PRO results further support the favourable benefit–risk profile of CEMI+CHEMO vs CHEMO in advanced NSCLC with baseline liver metastases.

Clinical trial identification

NCT03409614.

Editorial acknowledgement

Editorial support was provided by John G Facciponte, PhD, of Prime, Knutsford, UK.

Legal entity responsible for the study

Regeneron Pharmaceuticals, Inc.

Funding

Regeneron Pharmaceuticals, Inc., and Sanofi.

Disclosure

A. Baramidze: Other, Personal, Other, Travel support: Regeneron Pharmaceuticals, Inc. C. Gessner: Financial Interests, Personal, Advisory Board: AstraZeneca, Berlin-Chemie, Boehringer Ingelheim, Bristol Myers Squibb, Chiesi, GSK, Novartis, Merck Sharp & Dohme, Pfizer, Roche, Sanofi. A. Sezer: Financial Interests, Institutional, Funding: Roche, Merck Sharp & Dohme, Merck Serono, AstraZeneca, Lilly, Novartis, Johnson & Johnson, Regeneron Pharmaceuticals, Inc., Sanofi. M. Gumus: Financial Interests, Personal, Other, Honoraria: Roche, Merck Sharp & Dohme, Gen İlaç, Novartis. X. He, G. Gullo, P. Rietschel, R. Quek: Financial Interests, Personal, Full or part-time Employment: Regeneron Pharmaceuticals, Inc. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

76P - Immune checkpoint inhibitor (ICI) retreatment patterns and outcomes in patients with locally advanced or metastatic non-small cell lung cancer (NSCLC) who received second-line or later (2L+) nivolumab (nivo): A French I-O Optimise analysis

Presentation Number
76P
Lecture Time
20:30 - 20:30
Speakers
  • Gregoire Justeaux (Angers, Cedex 9, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

While ICIs demonstrate improved clinical trial outcomes in patients with NSCLC, little is known about real-world ICI retreatment patterns and outcomes after 2L+ nivo therapy. This analysis aimed to understand these endpoints in a real-world patient group in France.

Methods

Patients with locally advanced/metastatic NSCLC who initiated 2L+ nivo from 2015–2020 were included from ESME-AMLC, a retrospective observational cohort involving 39 centres in France (NCT03848052). Patient demographics, clinical and retreatment characteristics and outcomes were determined. Patients were followed from 2L+ nivo start date to last medical information date. Retreatment patterns were defined as: rechallenge (any ICI following an intervening non-ICI anticancer treatment after 2L+ nivo discontinuation); resumption (any ICI post-treatment break >6 weeks), ICI switch (any non-nivo ICI post-treatment break <6 weeks).

Results

After a median follow-up of 24 weeks, 2985/4001 (74.6%) patients discontinued 2L+ nivo treatment, 1604 received chemotherapy (CT) or targeted therapy (40.1%), 800 (20%) had no further treatment, and 226 (7.6%) were retreated with ICI. Of these: 110 (48.7%) were rechallenged (median treatment duration 2.1 months), 102 (45.1%) resumed (median treatment duration 3.3 months), and 14 (6.2%) switched ICI. Most patients retreated with ICI had non-squamous histology (80 [72.7%] patients with rechallenge, 76 [74.5%] with resumption, and 12 [85.7%] with ICI switch). Median duration of initial nivo treatment was 5.5 months for rechallenge, 7 months for resumption and 7 months for ICI switch. Median overall survival (OS) from retreatment date was 16.6 months vs 8.3 months for patients who resumed or were rechallenged. Patients who received initial 2L+ nivo for >26 weeks had higher ICI retreatment rates (15.8% overall) than patients who received 2L+ nivo for <13 weeks (3.6% overall).

Conclusions

Following 2L+ nivo discontinuation, most patients received CT. Few received ICI retreatment, although this increased with 2L+ nivo duration. Patients resuming ICI had longer OS than those with rechallenge.

Clinical trial identification

NCT03848052.

Editorial acknowledgement

Medical writing support was provided by Latitude (Powered by AXON).

Legal entity responsible for the study

Bristol Myers Squibb.

Funding

Bristol Myers Squibb.

Disclosure

G. Justeau: Non-Financial Interests, Personal, Advisory Role: BMS. M. Pérol: Financial Interests, Personal, Advisory Board: Roche, AstraZeneca, MSD, BMS, Lilly, Novartis, Takeda, Gritstone, Sanofi, Pfizer, Amgen, Janssen, GSK, Eisai, Ipsen; Financial Interests, Personal, Invited Speaker: Roche, AstraZeneca, MSD, BMS, Boehringer Ingelheim, Takeda, Illumina, Pfizer, Medscape; Financial Interests, Institutional, Research Grant: AstraZeneca, Roche, Takeda, Boehringer Ingelheim; Financial Interests, Personal, Steering Committee Member: Roche; Financial Interests, Personal, Other, DMSB: Roche. C. Audigier Valette: Financial Interests, Personal, Advisory Board: Roche, BMS, MSD, AstraZeneca, Sanofi, Janssen; Financial Interests, Personal, Invited Speaker: Pfizer. D. Debieuvre: Financial Interests, Personal, Advisory Board: AstraZeneca, BMS, MSD, Janssen, Pfizer, OSE Immunotherapeutics, Novartis, SanofiAventis, Amgen, Roche, Ipsen; Financial Interests, Personal, Invited Speaker: Gilead, Takeda; Financial Interests, Personal, Coordinating PI: Pfizer; Financial Interests, Institutional, Funding: Roche, AstraZeneca, Janssen, MSD, Pfizer, BMS, Lilly, Boehringer Ingelheim, GSK, Chugaï, Chiesi, Novartis, Takeda, Bayer, SanofiAventis. X. Quantin: Financial Interests, Institutional, Invited Speaker: Sanofi; Financial Interests, Institutional, Advisory Board: BMS; Financial Interests, Institutional, Other, Educational support: AstraZeneca. H. Lena: Non-Financial Interests, Personal, Research Funding: Bristol Myers Squibb; Non-Financial Interests, Personal, Coordinating PI: Regeneron Pharmaceuticals, Sanofi Aventis, GSK, Pfizer; Financial Interests, Personal, Advisory Board: Sanofi Aventis, Pfizer, Takeda, Roche, Amgen. P. Macouillard: Financial Interests, Institutional, Full or part-time Employment: Unicancer. L. Bosquet: Financial Interests, Institutional, Full or part-time Employment, In charge of scientific projects at Unicancer, Health Data and Partnership Department: Unicancer. M.J. Schoemaker, M. Mella, B. Correia: Financial Interests, Personal, Full or part-time Employment: IQVIA. C. Rault: Financial Interests, Personal, Speaker, Consultant, Advisor: Bristol Myers Squibb; Financial Interests, Personal, Full or part-time Employment: Data Gnosis. M.J. Daumont: Financial Interests, Personal, Full or part-time Employment: Bristol Myers Squibb; Financial Interests, Personal, Stocks or ownership: Bristol Myers Squibb. J. Penrod: Financial Interests, Personal, Full or part-time Employment: Bristol Myers Squibb; Financial Interests, Personal, Stocks or ownership: Bristol Myers Squibb. C. Chouaid: Financial Interests, Personal, Advisory Board: AZ, BI, GSK, Roche, Sanofi Aventis, BMS, MSD, Lilly, Novartis, Pfizer, Takeda, Bayer, Janssen and Amgen; Financial Interests, Institutional, Funding: AZ, BI, GSK, Roche, Sanofi Aventis, BMS, MSD, Lilly, Novartis, Pfizer, Takeda, Bayer, Janssen and Amgen.

Collapse
Poster Display (ID 34) Poster Display

78P - Final Analysis of the French Real-World Study EVIDENS: Effectiveness, Safety & Quality of Life At 36 Months of Nivolumab in Advanced Non-Small Cell Lung Cancer (NSCLC)

Presentation Number
78P
Lecture Time
20:30 - 20:30
Speakers
  • Fabrice Barlesi (Villejuif, Cedex, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

EVIDENS was a prospective, non-interventional, multicenter study evaluating nivolumab in metastatic NSCLC, initiated before immunotherapy was available in first-line. This final analysis presents results at 36-months’ follow-up.

Methods

Adults with pathologically confirmed lung cancer initiating nivolumab between October 2016–November 2017 were included and followed up until December 2020. Patient characteristics were recorded at diagnosis & nivolumab initiation. Survival was evaluated with the Kaplan-Meier method & quality of life (QoL) with the EuroQol-5D-3 Level (EQ-5D-3L) questionnaire and visual analog scale (VAS).

Results

A total of 1423 patients with NSCLC (69% male, median age 66 years) were included in this analysis. At nivolumab initiation, most patients had an Eastern Cooperative Oncology Group performance status (ECOG-PS) of 0–1 (83.0%), non-squamous histology (69.1%), stage IV disease (91.5%) & were current/former smokers (89.8%); 19.9% had brain metastases. Most patients (99.7%) had previously received chemotherapy; nivolumab was second line therapy in 73.5% of patients with a median treatment duration of 73.0 days. Median overall survival (OS) was 11.0 months (95%CI: 9.8–12.2) & progression-free survival (PFS) 3.0 months (95% CI: 2.6–3.2). In multivariate analyses, patients with squamous NSCLC, liver metastasis, ECOG-PS ≥2 or who never smoked had significantly higher risk of shorter OS & PFS. Age, brain metastasis, or corticosteroid therapy at nivolumab initiation had no effect on OS & PFS. The overall response rate was 12.1%; median duration of response was 13 months. Treatment-related adverse events occurred in 35.7% (any grade), 8.0% (grade 3) and 0.8% (grade 4) of patients. Patients continuously treated with nivolumab had better EQ-5D-3L VAS scores than those who discontinued.

Conclusions

EVIDENS final analysis confirm the effectiveness, safety & effect on QoL of nivolumab reported in clinical trials. They help inform clinical practice by virtue of a long-term follow-up & subgroup analyses suggesting greater benefit in certain patients. This conclusion might be explored further in the LIST study.

Clinical trial identification

NCT03382496.

Editorial acknowledgement

Tracy Harrison of Springer Healthcare Communications provided editing of the abstract prior to submission. Yaacoub Khalife of Bristol Myers Squibb France Medical Oncology Department provided editing of the abstract prior to submission.

Legal entity responsible for the study

The authors.

Funding

Bristol Myers Squibb.

Disclosure

F. Barlesi: Financial Interests, Institutional, Advisory Board: AstraZeneca, Bayer, Bristol Myers Squibb, Boehringer Ingelheim, Eli Lilly Oncology, F. Hoffmann-La Roche Ltd, Novartis, Merck, Mirati, MSD, Pierre Fabre, Pfizer, Sanofi Aventis, Seattle Genetics, Takeda, AbbVie, ACEA, Amgen, Eisai, Ignyta; Non-Financial Interests, Personal, Principal Investigator: AstraZeneca, BMS, Merck, Pierre Fabre, F. Hoffmann-La Roche Ltd., Innate Pharma, Mirati. D. Debieuvre: Financial Interests, Personal, Advisory Board: AstraZeneca, BMS, MSD, Janssen, Pfizer, OSE Immunotherapeutics, Novartis, Sanofi Aventis, Amgen, Roche, Ipsen; Financial Interests, Personal, Invited Speaker: Gilead, Takeda; Financial Interests, Personal, Coordinating PI: Pfizer; Financial Interests, Institutional, Funding: Roche, AstraZeneca, Janssen, MSD, Pfizer, BMS, Lilly, Boehringer Ingelheim, GSK, Chugaï, Chiesi, Novartis, Takeda, Bayer, Sanofi Aventis. J.B. Auliac: Financial Interests, Personal, Speaker, Consultant, Advisor: Sanofi, Janssen, AstraZeneca, Takeda, BMS, MSD. D. Moro-Sibilot: Financial Interests, Personal, Advisory Board: Lilly, Roche, BMS, MSD, AbbVie, Becton Dickinson, Pfizer, Takeda, AstraZeneca, Boehringer Ingelheim, Novartis, GSK, Sanofi; Financial Interests, Personal, Invited Speaker: Amgen; Financial Interests, Institutional, Funding, IFCT clinical trials: Pfizer; Financial Interests, Institutional, Funding: Roche, AbbVie, AstraZeneca. B. Asselain: Financial Interests, Personal, Other: BMS, AstraZeneca, Pierre Fabre, Servier, Daiichi Sankyo, Gilead. C. Audigier-Valette: Financial Interests, Personal, Advisory Board: Roche, BMS, MSD, AstraZeneca, Sanofi, Janssen; Financial Interests, Personal, Speaker, Consultant, Advisor: Pfizer. F. Breliier, E. Gauthier, D. Reynaud: Financial Interests, Personal, Full or part-time Employment: BMS. F. Cotté: Financial Interests, Personal, Full or part-time Employment: BMS; Financial Interests, Personal, Stocks/Shares: BMS. M. Pérol: Financial Interests, Personal, Advisory Board: Roche, AstraZeneca, MSD, BMS, Lilly, Novartis, Takeda, Gritstone, Sanofi, Pfizer, Amgen, Janssen, GSK, Eisai, Ipsen; Financial Interests, Personal, Invited Speaker: Roche, AstraZeneca, MSD, BMS, Boehringer Ingelheim, Takeda, Illumina, Pfizer, Medscape; Financial Interests, Institutional, Research Grant: AstraZeneca, Roche, Takeda, Boehringer Ingelheim; Financial Interests, Personal, Steering Committee Member: Roche; Financial Interests, Personal, Other, DMSB: Roche. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

79P - A prospective, single-arm, phase II study to evaluate the efficacy and safety of Tislelizumab plus chemotherapy in resectable NSCLC

Presentation Number
79P
Lecture Time
20:30 - 20:30
Speakers
  • Daqiang Sun (Tianjin, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Perioperative immunotherapy has been shown to be promising option for resectable non-small cell lung cancer(NSCLC). This study aimed to investigate the efficacy and safety of perioperative treatment with tislelizumab (PD-1 inhibitor) plus chemotherapy in resectable stage II-III NSCLC.

Methods

This open-label, single-arm, phase 2 trial planned to enroll 20 patients(pts) with resectable II-IIIB(N2) (AJCC 8th) NSCLC. Pts received neoadjuvant treatment with intravenous tislelizumab combined with chemotherapy Q3W for 2-3 cycles before surgery and 1-2 cycles after surgery (up to 4 cycles perioperative chemotherapy), followed by tislelizumab Q3W for 1 year. Primary endpoints were major pathological response (MPR) rate, the secondary endpoints included pathologic complete response (pCR) rate, objective response rate (ORR), event-free survival (EFS) and overall survival (OS). This study is registered with chictr.org.cn, ChiCTR2300068140.

Results

Between February 2023 and August 2023, 15 pts (median age: 67; male:80%) were enrolled, of whom 66.7% pts had stage III disease, 93.3% pts had squamous cell lung cancer. Among 15 enrolled pts, 5 pts are on neoadjuvant period; 10 pts completed neoadjuvant treatment, and 9 of them underwent surgery with 100% R0 resection. Of 9 pts who underwent resection, 5 pts (55.6%) achieved MPR and pCR. ORR was 63.6%, DFS and OS data was immature. The most common TRAEs were anemia (n=8; 53.3%), and thrombocytopenia (n =5; 33.3%). Most of the TRAEs were grade 1or 2. 3 pts experienced Grade 3 TRAE of WBC count decreased, increased creatine kinase and pneumonia, respectively. No grade 4-5 adverse events were reported.

Conclusions

Tislelizumab combined platinum-based chemotherapy demonstrated high MPR and pCR rate, feasible surgical resection and manageable toxicity in stage II-IIIB NSCLC. Pts will be followed for long-term survival.

Clinical trial identification

ChiCTR2300068140.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

77P - A single-arm trial of BACE plus tislelizumab in patients with NSCLC: updated analysis

Presentation Number
77P
Lecture Time
20:30 - 20:30
Speakers
  • Xuhua Duan (Zhengzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

80P - Efficacy and Safety of Tislelizumab Combined with Anlotinib and 2-cycles Chemotherapy as First-line Treatment for Advanced NSCLC(TISAL-FE-01)

Presentation Number
80P
Lecture Time
20:30 - 20:30
Speakers
  • Jun W. Tang (Haikou City, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Although IMpower150 demonstrated atezolizumab combined with bevacizumab plus chemotherapy was a standard of care for advanced NSCLC, the efficacy and safety remained to be improved. This study was aimed to evaluate the efficacy and safety of tislelizumab (tis)plus anlotinib (anlo) combined with 2-cycles chemotherapy as first-line treatment for advanced NSCLC.

Methods

This study was a single-arm, one set and phase II study. Eligible patients were histologically confirmed locally advanced or metastatic (Stage IIIB-IV) NSCLC and untreated, excluding driver mutations. Twenty patients received 2 cycles of tis (200mg, iv, Q3W) plus anlo (12mg, QD for 2W, Q3W) and platinum-based dual agent chemotherapy, followed by tis plus anlo maintenance therapy. The primary endpoint was progression free survival. The second endpoints included over survival, object response rate, during of response, disease control rate and safety. The number of planned enrollment patients was 30.

Results

As of the data cutoff of August 31, 2023, the median follow-up time was 8 months (95%CI 4-12).There were 20 pts eligible for inclusion,18/20 pts were stage IV and 14/20 pts were non-squamous NSCLC. The median PFS was 8.5months(95%CI:4.5-NA), the 6m-PFS% and 12m-PFS% were 75.9% (95%CI 0.477-0.902) and 36.1% (95%CI 0.103-0.634) respectively. The median OS has not arrived, the 12m-OS% was 77.9% (95%CI 0.440-0.927). The ORR was 75% and DCR was 100%, the median DOR was 5.2months (95%CI 1.68-NA). The safety was manageable and none ≥3TRAE and ≥3irAE occurred.

AEs, n (%) N=20
Any TEAEs 20(100)
TEAEs ≥15% Grade1-2 Grade 3
WBC decreased 6 (30) 2 (10)
Neutrophil count decreased 6 (30) 2 (10)
Canker sores 5 (25) 1 (5)
Rash 5 (25) 1 (5)
Anemia 5 (25)
Hand-foot-syndrome 5 (25) 2 (10)
Decreased platelet count 5 (25) 2 (10)
Pulmonary infection 4 (20)
Hypertension 4 (20) 1 (5)
GGT increased 4 (20)
Hypertriglyceridemia 3 (15)
Hyperuricemia 3 (15)
Vomiting 3 (15)

Conclusions

The preliminary result of this study indicated that tislelizumab, anlotinib combined with 2-cycles chemotherapy as first-line treatment for advanced NSCLC showed promising efficacy and good safety.

Clinical trial identification

TISAL-FE-01.

Legal entity responsible for the study

The authors.

Funding

BeiGene and Nanjing Zhengda Tianqing Pharmaceutical Co., Ltd.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

81P - Real-World Experience with Docetaxel Regimens in Metastatic Non-Squamous (mNSq) Non-Small Cell Lung Cancer (NSCLC) Patients Previously Treated with Platinum-Based Chemotherapy (PCT) and an Immune Checkpoint Inhibitor (ICI) in the United States (US)

Presentation Number
81P
Lecture Time
20:30 - 20:30
Speakers
  • Marisa A. Bittoni (Columbus, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Significant unmet needs exist for patients with mNSq NSCLC who experience disease progression after treatment with PCT and an ICI. Docetaxel (DOC)-based regimens are commonly used to treat such patients; however, data on the characteristics of and outcomes for those receiving these therapies in the real world are lacking. The objective of this study was to provide a benchmark for survival with present standard-of-care therapies by conducting a retrospective cohort study of real-world patients treated with DOC or DOC plus ramucirumab (DOC+RAM) in the US.

Methods

The study utilized data from US patients included in the ConcertAI Patient 360 NSCLC database who initiated first-line treatment between 01-Jul-2016 and 31-Dec-2020. Patients who had mNSq NSCLC, an Eastern Cooperative Oncology Group performance status of 0 or 1, and received DOC or DOC+RAM following disease progression after PCT and an ICI (in combination or separately) were eligible. Patients’ demographics and clinical characteristics as well as outcomes (real-world overall survival [OS], progression-free survival [PFS], time to treatment discontinuation [TTD], and time to next treatment [TTNT]) were analyzed.

Results

For patients receiving DOC (N=41; mean age: 65 years) or DOC+RAM (N=87; mean age: 64 years), median OS was 6.0 months (95% confidence interval [CI]: 4.8, 11.6) and 5.9 months (95% CI: 4.2, 11.6), respectively. Median PFS was 2.9 months (95% CI: 1.9, 4.6) for DOC and 2.7 months (95% CI: 2.3, 4.6) for DOC+RAM. Median TTD and TTNT were 2.1 months (95% CI: 1.4, 3.4) and 7.4 months (95% CI: 5.7, not estimable [NE]) for DOC and 1.5 months (95% CI: 1.4, 2.1) and 15.2 months (95% CI: 12.3, NE) for DOC+RAM, respectively. Estimates for TTNT should be considered with caution, given that a large number of patients were censored because of death or end of follow-up.

Conclusions

This study highlights the poor prognosis for real-world mNSq NSCLC patients who experience disease progression after PCT and ICI. The results can be used to help evaluate novel therapies in this population.

Editorial acknowledgement

Hriticka Choudhurry and Pranshu Roy from Sanofi provided medical writing support.

Legal entity responsible for the study

Sanofi.

Funding

Sanofi.

Disclosure

M.A. Bittoni: Financial Interests, Institutional, Speaker, Consultant, Advisor, consultancy services: Sanofi. S. Keeping, J.E. Park, A.T.H. Nguyen, K. Toor: Financial Interests, Institutional, Full or part-time Employment: PRECISIONheor; Professional service fees for conducting this research: Sanofi. D.P. Carbone: Financial Interests, Institutional, Speaker, Consultant, Advisor, consultancy services Sanofi. N. Petruski-Ivleva, A.K. Vadanahalli Shankar, G. Konidaris: Financial Interests, Institutional, Full or part-time Employment: Sanofi; Financial Interests, Institutional, Stocks/Shares: Sanofi.

Collapse
Poster Display (ID 34) Poster Display

82P - The Role of Circadian Rhythms in NSCLC Immunotherapy Efficacy: A Focus on First Dose Timing

Presentation Number
82P
Lecture Time
20:30 - 20:30
Speakers
  • Martin Igor Gomez-Randulfe Rodriguez (Manchester, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Circadian rhythms are known to influence adaptive immune responses. A seminal study showed that immune checkpoint inhibitors (ICI) infusions administered in the evening (later than 16:30h) to stage IV melanoma patients led to significantly shorter progression-free survival (PFS) and overall survival (OS). Subsequent retrospective studies suggested worse outcomes for metastatic non-small cell lung cancer (NSCLC) patients who received over 20% of their ICI doses in the evening. We hypothesized that the timing of the initial dose could be responsible for this observed effect.

Methods

We conducted a retrospective review of patients diagnosed with advanced or metastatic NSCLC who initiated palliative first line pembrolizumab treatment between June 2017 and April 2023 at The Christie. Sociodemographic and clinical data were collected. Patients were stratified into two groups based on the timing of their first dose infusion: those before 16:30h (early group) and those after 16:30h (evening group).

Results

276 patients met the inclusion criteria: 237 in the early group and 39 in the evening group. Both cohorts were similar, with no significant differences in sex, ECOG, histology, brain metastases at diagnosis, or PD-L1 expression (≥ or <90%). Inflammatory blood biomarkers (NLR, PLR, PNI) were consistent between groups. The evening group had a significantly shorter median OS (16m vs. 19m; HR 1.5; p=0.047), corroborated by the multivariate analysis (Table). Although median PFS was comparable between groups, the 3-year PFS rates stood at 10% for the evening group and 22% for the early group.

Multivariate Cox regression model

HR (95%) P
ECOG PS 0.50 (0.26 - 0.94) 0.031
Histology (non-squamous) 0.76 (0.55 - 1.04) 0.083
PD-L1 <90% 1.58 (1.17 - 2.15) 0.003
NLR <5 0.61 (0.45 - 0.82) 0.001
Evening Group 1.63 (1.09 - 2.43) 0.018

Conclusions

Our findings add to the emerging body of evidence suggesting a link between circadian rhythms and immunotherapy efficacy. While this study indicates a difference in OS based on the timing of the first dose, further prospective research is imperative. Modifying infusion timings could provide a straightforward approach to potentially optimizing patient outcomes.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

F. Blackhall: Financial Interests, Personal, Invited Speaker, Educational Symposium lecture: AstraZeneca; Financial Interests, Personal, Advisory Board, NTRK Advisory Board and guidelines for diagnosis: Bayer; Financial Interests, Personal, Other, IDMC Chair: AstraZeneca; Financial Interests, Personal, Advisory Board, Small cell Advisory Board Oct 2020: Amgen; Financial Interests, Personal, Invited Speaker, ESMO Satellite Symposium November 2020: Takeda; Financial Interests, Personal, Other, Consultancy for RETinhibitor development: Blueprint; Financial Interests, Personal, Other, Real world evidence research study design and analysis (EGFR): Janssen; Financial Interests, Institutional, Coordinating PI, Institutional payment for clinical trial activities: Amgen, Pfizer; Financial Interests, Institutional, Coordinating PI, Payment for clinical trial activities: Mirati; Financial Interests, Institutional, Coordinating PI, Clinical trial activities: BMS; Financial Interests, Institutional, Funding, Real world evidence research programme: Roche; Non-Financial Interests, Personal, Advisory Role, Application of genotyping platforms in lung cancer: Guardant Health; Non-Financial Interests, Personal, Advisory Role, Clinical trials of IMPs in lung cancer and translational lung cancer biomarkers: AstraZeneca. R. Califano: Financial Interests, Personal, Advisory Board: AstraZeneca, Bayer, Lilly Oncology, Roche, Pfizer, MSD, Takeda, Amgen, Bayer, Janssen and Novartis, Sanofi, GSK and PharmaMar; Financial Interests, Personal, Invited Speaker: AstraZeneca, Lilly Oncology, Roche, Pfizer, MSD, Takeda, Amgen, GSK and Janssen; Financial Interests, Personal, Other, speaker in educational activities: Medscape, Touch IME and PeerVoice; Financial Interests, Personal, Ownership Interest, partnership: The Christie Private Care - LOC; Financial Interests, Personal, Stocks/Shares: Supportive care UK; Financial Interests, Institutional, Local PI, principal investigator for clinical trial: Roche, AstraZeneca, Pfizer, Clovis, Lilly Oncology, MSD, BMS, AbbVie, Takeda, Janssen, and Novartis; Financial Interests, Institutional, Local PI: PharmaMar and GSK; Non-Financial Interests, Personal, Member, member of Lung Cancer Group: EORTC. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

83P - Association of immune-related adverse events and the efficacy of anti-PD-(L)1 monotherapy in patients with advanced NSCLC: a cohort study adjusting for immortal-time bias

Presentation Number
83P
Lecture Time
20:30 - 20:30
Speakers
  • Yun Fan (Hangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The association between immune-related adverse events (irAEs) and survival outcomes in non-small cell lung cancer (NSCLC) patients treated with programmed-death-(ligand)1 [PD-(L)1] inhibitors remains controversial, partly due to variations in dealing with immortal-time bias (ITB).

Methods

We retrospectively enrolled 425 patients with advanced NSCLC who received anti-PD-(L)1 monotherapy between January 2016 and June 2021, stratifying them into irAEs (n=127) and non-irAEs (n=298) groups. The primary endpoint was to assess the impact of irAEs on progression-free survival (PFS) and overall survival (OS). Landmark and time-dependent Cox analyses were performed in addition to conventional analysis to eliminate ITB.

Results

127 patients (29.9%) experienced 186 irAEs, with 89 single-organ and 38 multi-organ. 42 skin irAEs, 39 thyroid irAEs, 27 pneumonitis, 25 hepatic irAEs, and some less frequent irAEs et.al. With a median follow-up of 38.8 months, the occurrence of overall irAEs was significantly associated with superior PFS (11.2 vs 3.4 months, P<0.0001) and OS (31.4 vs 14.0 months, P<0.0001), and this positive association persisted in landmark (3-, 6-, and 9-month; all P<0.05) and time-dependent Cox analyses (adjusted HR for PFS=0.55, 95% CI 0.43-0.71; for OS=0.53, 95% CI 0.41-0.70). For the main organ-specific irAEs, skin, thyroid, and hepatic irAEs, respectively, showed significantly improved survival compared to the non-irAE group, whereas pneumonitis did not. Single-organ irAEs had the best outcomes compared with multi-organ or no irAE, which also held across subgroups of skin, thyroid and hepatic irAEs. Moreover, severe grade irAEs and immunotherapy discontinuation had a detrimental effect on survival, systemic steroid therapy showed little effect, while immunotherapy resumption had tolerable safety and a trend of improved survival.

Conclusions

After adequately adjusting ITB, the occurrence of overall irAEs predicts for favourable efficacy of anti-PD-(L)1 monotherapy in NSCLC, with better outcomes observed in patients with skin, thyroid, or hepatic irAEs, particularly those with single-organ involvement.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

84P - Adebrelimab plus chemotherapy (chemo) as first-line treatment for extensive-stage small-cell lung cancer (ES-SCLC): 3-year update of the phase 3 CAPSTONE-1 study

Presentation Number
84P
Lecture Time
20:30 - 20:30
Speakers
  • Ying Cheng (Changchun, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

In the phase 3 CAPSTONE-1 study, adebrelimab, a humanized IgG4 anti-PD-L1 monoclonal antibody, plus chemo as first-line treatment for ES-SCLC significantly improved overall survival (OS) vs placebo plus chemo. Here we reported updated outcomes ∼3 years after the enrollment of the last patient.

Methods

462 patients with systemically untreated ES-SCLC were randomized 1:1 to receive 4-6 cycles of adebrelimab (20 mg/kg, iv, d1, q3w) or placebo, with carboplatin (AUC 5, d1, q3w) plus etoposide (100 mg/m2, d1, d2, d3, q3w), followed by maintenance therapy with adebrelimab or placebo. The primary endpoint was OS.

Results

As of data cutoff of Aug. 11, 2023, median follow-up was 15.1 mo for all patients in the adebrelimab group and 12.8 mo in the placebo group; median follow-up for censored patients was 45.8 mo and 42.5 mo, respectively. OS events were recorded in 188 (81.7%) patients in the adebrelimab group and 212 (91.4%) in the placebo group. Median OS was 15.3 mo (95% CI 13.2-17.3) with adebrelimab plus chemo vs 12.8 mo (95% CI 11.3-13.9) with placebo plus chemo (HR 0.73, 95% CI 0.60-0.89; one-sided p=0.0008). Over 10% improvement in OS rate was seen at 12, 24 and 36 mo in the adebrelimab group vs placebo group (Table 1). The OS benefits with adebrelimab were generally consistent across clinically relevant subgroups. No new safety signals were observed.

OS outcomes

Adebrelimab + chemo (N=230) Placebo + chemo (N=232)
Median OS*, mo (95% CI) 15.3 (13.2-17.3) 12.8 (11.3-13.9)
HR† (95% CI); p-value‡ 0.73 (0.60-0.89); p=0.0008
OS rate*, % (95% CI)
12 mo 62.9 (56.3-68.8) 52.0 (45.4-58.2)
24 mo 30.9 (25.0-37.0) 17.8 (13.1-23.0)
36 mo 21.1 (16.1-26.7) 10.5 (6.9-14.9)

* Kaplan-Meier method. † Stratified Cox proportional-hazards model stratified by liver metastases (yes/no), brain metastases (yes/no), and lactate dehydrogenase concentration (normal/elevated) at baseline. ‡ One-sided p-value was calculated based on stratified log-rank test.

Conclusions

After prolonged follow-up, the addition of adebrelimab to chemo continued to demonstrate OS benefits with manageable toxicities, further supporting this regimen as a new first-line treatment option for ES-SCLC.

Clinical trial identification

NCT03711305.

Legal entity responsible for the study

Jiangsu Hengrui Pharmaceuticals.

Funding

Jiangsu Hengrui Pharmaceuticals.

Disclosure

C. Zhou: Financial Interests, Personal, Speaker’s Bureau: Lily China, Sanofi, BI, Roche, MSD, Qilu, Hengrui, Innovent Biologics, C-Stone, Luye Pharma, TopAlliance Biosciences Inc., Amoy Diagnositics, Anheart Therapeutics; Financial Interests, Personal, Advisory Role: Innovent Biologics; Financial Interests, Personal, Advisory Board: Hengrui, Qilu, TopAlliance Biosciences Inc. X. Li, K. Ma: Financial Interests, Personal, Full or part-time Employment: Hengrui. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

85P - Phase II trial of tislelizumab plus sitravatinib as maintenance therapy in extensive-stage small-cell lung cancer (ES-SCLC)

Presentation Number
85P
Lecture Time
20:30 - 20:30
Speakers
  • Yun Fan (Hangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

First-line chemo-immunotherapy has shown significant survival improvement in ES-SCLC, however, rapid drug resistance frequently occurs. Sitravatinib (Sitra), a selective tyrosine kinase inhibitor targeting TAM, VEGFR2 and KIT, can alter immune landscape to favor immunotherapy and overcome resistance. This single-arm, phase Ⅱ trial evaluated the efficacy and safety of tislelizumab (TIS, a PD-1 inhibitor) plus Sitra as maintenance therapy in ES-SCLC patients (pts) after induction therapy with TIS plus chemotherapy (chemo), aiming to postpones disease progression.

Methods

Eligible pts with untreated ES-SCLC received TIS (200 mg) plus platinum–based chemo Q3W for 4 cycles (induction), followed by maintenance TIS 200 mg Q3W and Sitra 70 mg daily. Maintenance analysis set (MAS) included pts who received ≥1 dose maintenance therapy. Efficacy outcomes in MAS were calculated from the start of maintenance TIS+Sitra. The primary endpoint was 1-year PFS rate per RECIST v1.1 in MAS.

Results

Between March 2022 and August 2022, 21 pts were enrolled and received induction therapy. Of these, 18 pts entered maintenance phase, with 27.8% having liver metastases, 27.8% bone metastases, 16.7% brain metastasis, and 94.4% ECOG PS 1. As of 21 Jul 2023, among the 18 pts in MAS, median PFS was 6.4 months and 1-year PFS rate was 27.8% from the start of maintenance therapy. Median OS was not reached. In MAS, 4 pts achieved further PR during maintenance therapy, yielding a confirmed ORR of 22.2%, and the DCR was 88.9%. In MAS, TRAEs and irAEs of grade 3-4 occurred in 10 (55.6%) and 2 (11.1%) pts, respectively. Serious TRAEs occurred in 4 (22.2%) pts. No TRAEs led to treatment discontinuation or death.

Maintenance analysis set, N=18 Induction analysis set, N=21
Efficacy
Confirmed ORR, % (95% CI) 22.2 (6.4, 47.6) 76.2 (52.8, 91.8)
DCR, % (95% CI) 88.9 (65.3, 98.6) 95.2 (76.2, 99.9)
Median DoR, months (95% CI) 7.6 (5.3, NR) 7.9 (6.0, 10.4)
Median PFS, months (95% CI) 6.4 (4.5, 9.0) 9.1 (7.3, 11.8)
1-year PFS rate, % (95% CI) 27.8 (10.1, 48.9) 26.3 (9.6, 46.8)
1-year OS rate, % (95% CI) 59.4 (27.9, 80.9) 65.3 (40.5, 81.8)
Safety
Grade 3-4 TRAEs, n (%) 10 (55.6) 18 (85.7)
Grade 3-4 irAEs, n (%) 2 (11.1) 2 (9.5)

Defined as pts who received ≥1 dose of induction therapy; efficacy outcomes in induction analysis set were calculated from the start of induction therapy. NR=not reached

Conclusions

TIS plus Sitra showed promising efficacy with manageable toxicities as maintenance therapy in ES-SCLC pts after induction TIS+chemo. It provides a new treatment strategy to delay drug recurrence and prolong the survival for ES-SCLC pts.

Clinical trial identification

NCT05228496.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

86P - Durvalumab plus Olaparib as maintenance therapy in extensive-stage small-cell lung cancer (TRIDENT): updated efficacy and safety analysis

Presentation Number
86P
Lecture Time
20:30 - 20:30
Speakers
  • Yan Huang (Guangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

First line durvalumab in extensive-stage small-cell lung cancer (ES-SCLC) demonstrated significant improvement of OS in CASPIAN trial. PARP inhibitors have the potential to confer antitumor activity, modify tumor immunogenicity, and sensitize tumors to anti-PD-1/PD-L1 therapy. Here, we investigated the efficacy and safety of durvalumab plus Olaparib as maintenance therapy in ES-SCLC pts after Durvalumab plus chemotherapy as first line treatment.

Methods

TRIDENT is a single arm, multicenter, phase 2 study. Treatment-naïve ES-SCLC aged ≥18 with ECOG PS 0-2 were eligible. Durvalumab (1500 mg) was concurrently administered with platinum–etoposide every 3 weeks for 4 cycles, followed by durvalumab 1500 mg every 4 weeks plus Olaparib 300 mg twice daily until disease progression or unacceptable toxicity. The primary endpoint was the rate of progression free survival at 12 months (APF12). Secondary endpoints included PFS, Overall Survival, Objective Response Rate (according to RECIST1.1.) and safety profile.

Results

60 patients were enrolled from 4 sites in China between August 2021 and August 2022. At the data cutoff on August 9, 2023, the median duration of follow-up was 13.0 months. 10 (16.7%) patients were still receiving study treatment. The primary endpoint of PFS at 12 months was 24.5% (95% CI, 14.3%-36.2%). Median PFS from first line treatment was 6.7 months (95% CI, 5.1-8.4), median overall survival was 14.6 months (95% CI 10.9–21.8); 2 (3.3%) patients received complete response (CR), 42 (70%) patients received partial response (PR) and the ORR was 73.3% (95%CI, 60.3-83.9%). Treatment-emergent adverse events (TEAE) occurred in 58 (96.7%) patients, with 22 (36.7%) of grade ≥3; 15 (25%) patients reported serious adverse events; TEAEs leading to death of any cause occurred in 2 (3.3%) patients.

Conclusions

Durvalumab plus Olaparib as maintenance therapy in ES-SCLC patients showed encouraging anti-tumor activity without new safety signal observed. Further exploration of ES-SCLC subpopulation who may benefit from durvalumab combined with Olaparib modality is needed.

Clinical trial identification

NCT05245994.

Legal entity responsible for the study

Sun Yat-sen University Cancer Center.

Funding

Sun Yat-sen University Cancer Center and AstraZeneca.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

88P - Adverse events (AEs) as potential predictive factors of activity in patients with advanced hepatocellular carcinoma (HCC) treated with atezolizumab plus bevacizumab (AB)

Presentation Number
88P
Lecture Time
20:30 - 20:30
Speakers
  • MARA PERSANO (Cagliari, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

This retrospective multicenter real-world study aims to investigate the potential prognostic value of AEs in HCC patients treated with AB in first-line setting.

Methods

The study’s population consisted of 823 HCC patients from five countries (Italy, Germany, Portugal, Japan, and the Republic of Korea) treated with AB between October 2018 and April 2022.

Results

Median overall survival (OS) was 15.9 months and median progression-free survival (PFS) was 7.6 months. 73.3% of patients presented at least one AE during the study period. The most common AEs were proteinuria (29.6%), arterial hypertension (27.2%), and fatigue (26.0%). 17.3% of the AEs were grade (G) 3. One death due to bleeding was reported. The multivariate analysis confirmed the appearance of decreased appetite G < 2 versus G ≥ 2 [hazard ratio (HR): 0.60; p < 0.01] and immunotoxicity G < 2 versus G ≥ 2 (HR: 0.70; p = 0.04) as independent prognostic factors for OS, and the appearance of decreased appetite G < 2 versus G ≥ 2 (HR: 0.73; p = 0.01), diarrhea of any G versus no diarrhea (HR: 0.57; p = 0.01), fatigue of any G versus no fatigue (HR: 0.82; p < 0.01), arterial hypertension G < 2 versus G ≥ 2 (HR: 0.68; p < 0.01), and proteinuria of any G versus no proteinuria (HR: 0.79; p = 0.03) as independent prognostic factors for PFS. The objective response rate (ORR) was 27.3%: complete responses (CR) were 35 (4.2%), partial response 190 (23.1%), stable disease 428 (52.0%), and progressive disease 170 (20.7%). The appearance of hypothyroidism of any G (odds ratio: 0.52; p = 0.04) and immunotoxicity of any G (odds ratio: 0.54; p < 0.01) were correlated with higher ORR, while the absence of fatigue of any G (odds ratio: 3.90; p = 0.02) and decreased appetite of any G (odds ratio: 9.71; p = 0.02) were correlated with more CR.

Conclusions

As also demonstrated for other therapies, also for the combination of AB there is a correlation between the occurrence of AEs and HCC patient 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
Poster Display (ID 34) Poster Display

89P - Hepatic arterial infusion chemotherapy sequential transarterial embolization combined with lenvatinib and tislelizumab in patients with high-risk unresectable hepatocellular carcinoma: a single arm phase 2 trial

Presentation Number
89P
Lecture Time
20:30 - 20:30
Speakers
  • Liang Ma (Nanning, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Hepatic arterial infusion chemotherapy (HAIC), transarterial embolization (TAE), and lenvatinib are main treatments in unresectable hepatocellular carcinoma (HCC). This study aims to evaluate the safety and efficacy of HAIC sequential TAE combined with lenvatinib and tislelizumab (an anti-PD-1) as a first-line treatment in patients with high-risk unresectable HCC.

Methods

This is a single-arm, open-label, phase 2 trial (NCT05532319), which is ongoing. Eligible patients with high-risk unresectable HCC (CNLC stage Ib, II and III), ECOG performance status of 0 or 1 and adequate liver function received at least one cycle of HAIC sequential TAE (every 4 weeks) combined with lenvatinib and tislelizumab (200 mg intravenously every 3 weeks). The primary endpoint was the progression-free survival (PFS) rate at six months. Secondary endpoints included objective response rate (ORR), the proportion of patients who underwent curative treatments, significant tumour necrosis, PFS, OS and safety.

Results

Between Nov 1, 2022 and Aug 12, 2023, 35 patients with high-risk unresectable HCC were enrolled. The tumour load of 14 (40%) patients exceeds 50% of the liver volume. The PFS rate at six months was 83.6%. The ORR after one cycle of HAIC sequential TAE was 31.9% (15/47, RECIST 1.1) and 63.8% (30/47, mRECIST). The ORR after two cycle of HAIC sequential TAE was 35.1% (26/40, RECIST 1.1) and 80.0% (32/40, mRECIST). 19 patients (54.3%) received curative treatment after reaching partial response, including 10 (28.6%) curative hepatic resection, 6 (17.1%) radiotherapy, 2 (5.7%) radiofrequency ablation and 1 (2.9%) liver transplantation. Significant tumour necrosis was observed in 100% tumours based on postoperative histopathology in patients who received completed hepatectomy. Median PFS and OS were not reached. Most adverse events were grade 1 or 2. The most common were thrombocytopenia (22.9%), alanine aminotransferase elevating (20%) and serum albumin decreasing (17.1%).

Conclusions

HAIC sequential TAE combined with lenvatinib and tislelizumab is a safe and effective treatment modality in patient with high-risk unresectable HCC.

Legal entity responsible for the study

The authors.

Funding

BGNE.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

90P - HAIC plus sintilimab and bevacizumab biosimilar as treatment for patients with advanced hepatocellular carcinoma (HCC): a phase II trial

Presentation Number
90P
Lecture Time
20:30 - 20:30
Speakers
  • HAIBIN ZHANG (Shanghai, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The combination treatment using sintilimab (a PD-1 antibody) and bevacizumab (bev) biosimilar demonstrated excellent efficacy and safety in ORIENT-32 study and HAIC performed impressive tumor response in researches. We assessed the efficacy and safety of the combination therapy as first-line treatment for initial unresectable HCC.

Methods

The study was an ongoing open-label, single-arm, phase II trial. Treatment-naïve HCC patients (pts) with initial unresectable and BCLC stage B or C were enrolled in this phase II trial. Eligible pts received sintilimab (200 mg, IV, D1) and bev (15 mg/kg, IV, D1) per 3 weeks, as well as FOLFOX-HAIC per 3-6 weeks. Pts eligible for resection were referred for hepatectomy after bev weaned over at least a 4-week period. Sintilimab and bev were given until disease progression or unacceptable toxicity or up to 24 months, and FOLFOX-HAIC were given 3-6 times. Primary endpoints were progression-free survival (PFS) per RECIST v1.1 and key secondary endpoints included objective response rate (ORR), disease control rate (DCR), surgical conversion rate, pathologic response, overall survival (OS) and safety.

Results

At data cutoff on July 25, 2023, 43 pts were enrolled, 41 of them underwent at least one course of treatment and 38 enrollees received at least one tumor assessment. The median follow-up time was 6.34 months (range 0.2–14.62). mPFS and mOS were not reached. According to RECIST v1.1, ORR and DCR were 68.42% and 100% (26 PR and 12 SD). Moreover, ORR and DCR were 86.84% and 100% (8 CR, 25 PR and 5 SD) based on mRECIST, respectively. 10 pts met the criteria for hepatectomy and 6 pts received liver resection. Among this 6 pts, 5 pts was in stage IIIA and 1 in IIIB before enrollment. No postoperative mortality was observed. Most treatment-related AEs (TRAE) were grade 1-2, and the incidence of grade 3 TRAEs was 14.63%. Grade 3 TRAEs includes decreased platelet count, hypertension, increased ALT, decreased white blood cell count, rash, hematencephalon and upper gastrointestinal hemorrhage.

Conclusions

Systemic therapy using HAIC plus Sintilimab and bev as first-line therapy for initial unresectable HCC was well tolerated and effective. Longer follow up is ongoing.

Clinical trial identification

ChiCTR2000034758.

Legal entity responsible for the study

Shanghai Eastern Hepatobiliary Surgery Hospital.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

91P - A real-world study of tislelizumab (Anti-PD-1) plus tyrosine kinase inhibitors for intermediate or advanced hepatocellular carcinoma

Presentation Number
91P
Lecture Time
20:30 - 20:30
Speakers
  • Wei Zhang (Nantong, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Data on elucidating the efficacy and safety of tislelizumab combined with tyrosine kinase inhibitors (TKIs) in hepatocellular carcinoma (HCC) are lacking. Herein, we explored the regimen of tislelizumab plus TKIs for intermediate or advanced HCC patients (pts) in a real-world setting.

Methods

In this study, 73 pts were included between Jul. 2021 and Jul. 2023. Pts were assigned either with tislelizumab plus TKIs (n=33), or tislelizumab plus TKIs plus TACE (n=40), where TKIs included lenvatinib (49.3%), regorafenib (23.3%), donafenib (21.9%), apatinib (2.7%) and anlotinib (2.7%). The primary endpoints were objective response rate (ORR, mRECIST criteria) and progression-free survival (PFS). The secondary endpoints included 6-month and 12-month PFS rates, disease control rate (DCR) and safety.

Results

Among the 73 pts, 80.8% were male, 53.4% had BCLC stage B, 75.3% were Child-Pugh A, 69.9% were first-line recipients and the average age was 62.8 years. The mean follow-up period was 13.3 (95% CI: 11.9-14.8) months and the mean number of TACE sessions were 0.8 (range 0-3) for all patients. The ORR and DCR were 42.5% and 79.5% with a median PFS of 12.6 months (95% CI: 8.71-16.49) and the 6-month, 12-month PFS rates were 69.6% and 50.4%. Compared to the tislelizumab plus TKIs group, the tislelizumab plus TKIs plus TACE group achieved longer median PFS (12.7 months (95% CI 9.17-16.23) vs. 8.4 months (95% CI 4.83-11.97), P=0.132), higher ORR (55.0% vs 27.3%, P = 0.017) and DCR (87.5% vs 69.7%, P = 0.061). Any-grade TRAEs were reported in 59 (80.8%) pts, and grade 3 TRAEs were reported in 16 (21.9%) pts. Most frequent TEAEs (≥10%) included hand-foot syndrome (24.7%), increased transaminase (15.1%), hyperbilirubinemia (13.7%), diarrhea (13.7%) and fever (11.0%). G3 TRAEs occurred in more 5% pts included hand-foot syndrome (8.2%), increased transaminase (6.8%). No grade 4/5 TRAE and new safety signal were identified.

Conclusions

Tislelizumab plus TKIs with TACE appeared to deliver encouraging efficacy and acceptable safety for pts with intermediate or advanced HCC. Further patient recruitment and longer follow up are warranted.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

92P - TAE-HAIC plus lenvatinib and PD-1 inhibitors versus TAE-HAIC plus atezolizumab and bevacizumab for unresectable hepatocellular carcinoma: A propensity score matching study

Presentation Number
92P
Lecture Time
20:30 - 20:30
Speakers
  • Hongjie Cai (Guangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Systemic therapy combined with transarterial-based therapy has demonstrated promising results for unresectable HCC. Atezolizumab plus bevacizumab (“T+A”) is the standard first-line therapeutic regimen for advanced HCC, while lenvatinb combined programmed death-1 (PD-1) inhibitors shows synergistic anti-tumor effect as well. This study aims to compare the efficacy and safety of TAE-HAIC plus lenvatinib and PD-1 inhibitors versus TAE-HAIC plus “T+A” for unresectable HCC.

Methods

In this retrospective study, treatment-naïve unresectable HCC patients who were treated with TAE-HAIC plus lenvatinib and PD-1 inhibitors (THLP group) or TAE-HAIC plus “T+A” (THTA group) were included. The primary endpoint was overall survival (OS). The secondary outcomes included progression-free survival (PFS) and tumor response according to modified RECIST, and adverse events (AEs). We performed propensity score matching (PSM) approaches to reduce bias between two groups.

Results

From June 2020 to June 2023, 339 patients were enrolled in this study: 233 in the THLP group and 106 in the THTA group. After PSM with a ratio of 3:1, 153 and 51 patients were assigned to the THLP and THAT group, respectively. The THLP group showed a longer median OS (21.3 versus 18.2 months; P = 0.486), while median PFS was longer in the THTA group (6.8 versus 6.3 months; P = 0.552), both without statistical differences. There were no statistical differences in objective response rate (ORR) (73.3% versus 68.3%; P =0.635) and disease control rate (DCR) (91.7% versus 89.3%; P =0.716) neither. No significant difference in the rate of the grade 3/4 AEs was observed between the two groups, and all AEs were controllable. No treatment-related grade 5 AE took place in the two groups.

Conclusions

TAE-HAIC plus lenvatinib and PD-1 inhibitors or TAE-HAIC plus “T+A” demonstrated similar outcomes for unresectable HCC with acceptable toxic effects, which needs to be validated with larger-scale randomized clinical trials.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

93P - The survival impact of the addition of durvalumab to cisplatin/gemcitabine in advanced biliary tract cancer: a real-world, retrospective, multicentric study.

Presentation Number
93P
Lecture Time
20:30 - 20:30
Speakers
  • Margherita Rimini (Milan, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The TOPAZ-1 phase III trial reported a survival benefit with the anti-programmed death cell ligand 1 (anti-PD-L1) durvalumab in combination with gemcitabine and cisplatin in patients with advanced biliary tract cancer (BTC). The present study investigated for the first time the survival impact resulted from the addition of durvalumab to cisplatin/gemcitabine in a real-world setting.

Methods

The analyzed population included patients with unresectable, locally advanced, or metastatic BTC treated with durvalumab in combination with cisplatin/gemcitabine or cisplatin/gemcitabine alone. The impact of the addition of durvalumab to chemotherapy in terms of both overall survival (OS) and progression free survival (PFS) was investigated with uni- and multivariate analysis.

Results

Overall, 358 patients were included in the analysis: 213 received cisplatin/gemcitabine alone, 145 received cisplatin/gemcitabine plus durvalumab. At the univariate analysis, the addition of durvalumab resulted to have a survival impact, since the median OS was 11.2 Vs 12.9 months (HR 1.8, 95% CI 1.3-2.5, p=0.0005) in patients who received cisplatin/gemcitabine alone compared to those who received cisplatin/gemcitabine plus durvalumab. Moreover, patients who received cisplatin/gemcitabine alone showed worse PFS compared to those who received cisplatin/gemcitabine plus durvalumab (mPFS 6.0 Vs 8.9 months, HR 1.8, 95% CI 1.4-2.3, p<0.0001). The multivariate analysis confirmed that the addition of durvalumab to cisplatin/gemcitabine is a independent prognostic factor for both OS and PFS. Finally, an exploratory analysis of the prognostic factors in the cohort of patients who received durvalumab was performed: NLR>3 and ECOG PS>0 resulted to be independent prognostic factors in terms of both OS and PFS in this cohort of patients. The interaction test highlighted NLR>3 and ECOG>1 as predictive factors of response to cisplatin/gemcitabine plus durvalumab.

Conclusions

Accordingly to the results of the TOPAZ-1, the addition of durvalumab to cisplatin/gemcitabine has been confirmed to confer a survival benefit in terms of both OS and PFS in a real-world setting of advanced BTC patients.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

94P - First-line chemotherapy plus immunotherapy versus chemotherapy alone for advanced gallbladder carcinoma

Presentation Number
94P
Lecture Time
20:30 - 20:30
Speakers
  • Qin-qin Liu (Guangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Gallbladder carcinoma (GBC) is a highly malignant tumor of the biliary tract. The prognosis for advanced GBC is generally very poor with scarce evidence on effective treatment options. In light of the limited therapeutic strategies, this study aims to shed light on the outcomes of using chemotherapy alone or in combination with PD-1 inhibitor as the first-line therapy.

Methods

All the patients who received systematic treatment for unresectable or recurrent GBC were included in this study from January 2018 to December 2022. The systematic treatment involved either chemotherapy alone or combination therapy. Clinical data were collected, including baseline characteristics, therapeutic response to the treatment, overall survival, and progression-free survival. Additionally, adverse events (AEs) during the treatment period were carefully documented to access the safety of the treatment.

Results

The eligible patients were divided into two groups: one group received combination therapy consisting of chemotherapy and PD-1 inhibitor (n=31), while the other group received mono-chemotherapy (n=19). The results of the study showed that the overall survival (OS) and progression-free survival (PFS) were longer in the combination therapy group compared to the mono-chemotherapy group. The patients who received combination therapy had a median OS of 18.2 months, whereas those who received mono-chemotherapy had a median OS of 6.1 months (P=0.016). The median PFS for combination therapy was 11.3 months, compared to 5.5 months for mono-chemotherapy, with a p-value of 0.008. The overall response rate (ORR) was 35.5% in the combination therapy group and 5.3% in the mono-chemotherapy group (P=0.037). Despite these favorable outcomes, the combination therapy did not demonstrate a significant advantage over mono-chemotherapy in terms of disease control rate (DCR). The DCR was 74.2% in the combination therapy group and 57.9% in the mono-chemotherapy group (P=0.230).

Conclusions

The present findings imply that combining chemotherapy and PD-1 inhibitor may be an effective and safe first-line treatment option for patients with unresectable or recurrent GBC, offering improved OS, PFS and ORR compared to chemotherapy alone.

Clinical trial identification

SYSKY-2022-282-01.

Legal entity responsible for the study

Sun Yat-sen Memorial Hospital.

Funding

This work was supported by the Special Research Foundation of the National Nature ScienceFoundation of China (82273476, 81972262, 81972255, 82173195); Grant [2013]163 from Key Laboratory of Malignant Tumor Molecular Mechanism and Translational Medicine of Guangzhou Bureau of Science and Information Technology; Grant KLB09001 from the Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes; Grant from Guangdong Science and Technology Department (2015B050501004, 2017B030314026); Sun Yat-sen University Clinical Research 5010 Program (2018008).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

95P - A single-arm, multicenter phase ? trial evaluating TQB2450 plus anlotinib combined with paclitaxel and cisplatin in first-line treatment of advanced esophageal squamous cell carcinoma (ESCC)

Presentation Number
95P
Lecture Time
20:30 - 20:30
Speakers
  • Junsheng Wang (Anyang, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Anlotinib is a multi-target TKI that has been proved to have good efficacy and tolerable toxicity whether as first-line treatment when combined with TP in advanced ESCC. TQB2450 is a novel humanized anti-PD-L1 monoclonal antibody. We conducted a phase II trial to evaluate the efficacy and safety of alotinib combined with TQB2450, cisplatin, and paclitaxel as first-line treatment for advanced ESCC. Here is the update results.

Methods

Eligible patients (pts) with previously untreated unresectable locally advanced or metastatic ESCC received TQB2450 (1200mg, iv, d1, q3w) plus anlotinib (10mg, po, d1∼14, q3w) combined with paclitaxel (135mg/m2 , iv, d1, q3w) and cisplatin (60∼75mg/m2 , iv, d1∼3, q3w) for 4 - 6 cycles as initial therapy. Patients without progressive disease (PD) continued to receive same dose of anlotinib plus TQB2450 as maintenance therapy until PD or unacceptable toxicity. The primary endpoint was PFS (RECIST version 1.1). Secondary endpoints included iPFS (iRECIST), ORR (RECIST version 1.1), DCR, DOR and safety.

Results

At the data cutoff date of April 15, 2023, 50 pts were enrolled with a median age of 64 years (range 41-74), male (38/50, 76%) and ECOG PS 1 (39/50, 78%). Among 45 tumor response evaluable pts, the ORR was 82.2% (95% CI: 68.3%, 91.7%) and the DCR was 100.0% (95% CI: 92.1%, 100.0%). The preliminary median PFS was not reached. The incidence of grade 3-4 treatment emergent adverse events was 66% (33/50), there was no grade 5 TRAE. 20 pts (40%, 20/50) occurred treatment related serious AEs. 25 patients received maintenance treatment for more than 10 cycles, the median DOT was 12.19m(9.53m-18.53m). Only 5 patients experienced grade 3-4 AE, mainly include leukopenia, hypertension, hyponatremia and hypokalemia.

Conclusions

TQB2450 plus anlotinib with paclitaxel and cisplatin showed promising activity with well-tolerated toxicities in pts with advanced ESCC as first-line treatment and the maintenance treatment also showed manageable safety.

Clinical trial identification

NCT05013697.

Legal entity responsible for the study

The authors.

Funding

Chia Tai Tianqing Pharmaceutical Group Co., Ltd.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

96P - Cadonilimab combined with taxane and cisplatin as the first-line treatment of advanced esophageal squamous cell carcinoma (ESCC): an open-label, multicenter phase ? trial (AK104-IIT-014)

Presentation Number
96P
Lecture Time
20:30 - 20:30
Speakers
  • Wang Qu (Bejing, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Chemotherapy combined with anti-PD-1 monoclonal antibody has already become the first-line treatment in advanced ESCC, while the benefits in objective responses and survival were limited. Cadonilimab (AK104), a bispecific antibody simultaneously targeting PD-1 and CTLA-4, was designed to boost anti-tumour activity with an improved safety profile. Here, we first evaluated the safety and efficacy of AK104 combination therapy as the first-line treatment in advanced ESCC.

Methods

Eligible pts with previously untreated unresectable locally advanced or metastatic ESCC received AK104 (10mg/kg, iv, d1, q3w) combined with paclitaxel or nab-paclitaxel (175 mg/m2, iv, d1, q3w) and cisplatin (65 to 75 mg/m2, iv, d1, q3w) for up to 6 cycles, then continued AK104 (10mg/kg, iv, d1, q3w) treatment as maintenance until progressive disease or unacceptable toxicity, with a maximum of 24 months. The primary endpoint was objective response rate (ORR). Secondary endpoints were progression-free survival (PFS), overall survival (OS), disease control rate (DCR) and safety.

Results

As of data cut-off (September 21, 2023), 22 pts were enrolled with a median age of 61 years (range 44-75), 50.0% had PD-L1 CPS<10. In the efficacy-evaluable population (n=15), 13 pts reached partial response and 2 pts had stable disease. The ORR was 86.7% (95%CI: 58.4%-97.7%) and the DCR was 100.0% (95%CI: 74.7%-100%). Both median PFS and OS were not reached. Among evaluable pts with PD-L1 CPS≥10 and PD-L1 CPS<10, the ORR were 83.3% (5/6) and 88.9% (8/9), respectively. Seven (31.8%) pts experienced grade 3-4 TRAEs, which mainly included neutropenia (22.7%), leukopenia (9.1%) and hyponatremia (9.1%). Immune-related AEs were observed in 3pts (13.6%). 3 pts (13.6%) suffered from serious AEs, and no grade 5 TRAE was observed. AEs led to discontinuation of AK104 in 2 (9.1%) of enrolled pts.

Conclusions

Bispecific antibody AK104 combined with taxane and cisplatin showed better ORR and manageable safety as first-line treatment in advanced ESCC, regardless of PD-L1 expression. Survival outcomes will be reported in the future and single AK104 cohort in pts with PD-L1 CPS≥5 ESCC is being explored.

Clinical trial identification

NCT05522894.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

97P - ICI for patients with MSS metastatic colorectal cancer

Presentation Number
97P
Lecture Time
20:30 - 20:30
Speakers
  • Zayana Sangadzhieva (Moscow, Russian Federation)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Colorectal cancer (CRC) is the third most often-diagnosed cancer in both men and women, with more than 1.9 million new cases worldwide in 2020. Immune-checkpoint inhibitors (ICI) show modest activity and efficacy in microsatellite stable (MSS) metastatic colorectal cancer (mCRC) patients harbouring a proficient mismatch repair system (pMMR). However, even the fact that MSS tumours are infiltrated by T cells was proved, they will not respond to therapy with anti-PD-1 mAbs as they do not express ICI but escape from immunosurveillance, for example, by downregulating HLA expression. Importantly, there is a hypothesis that a particular fraction of non-hypermutated MSS tumours might in theory respond to PD-1/PD-L1 blockade therapy. Some trials demonstrated benefits for surveillance for MSS tumours treated by intensive combination of chemotherapy with ICI, but fragile and older patients cannot receive these regimens. Treatment with less toxicity could be the option for some specific group of patients with MSS tumours.

Methods

We performed a retrospective analysis of the efficacy and safety of pembrolizumab and lenvatinib for late-stage colorectal cancer as a salvage line. The Kaplan-Meier method was used to estimate survival distribution and response rate (RR).

Results

There were 20 cases (male:13, female:7), and the median average age was 63 (32-75) years old. The median progression-free survival (PFS) and overall survival (OS) were 4.5 (95%CI;1.4-14) months and 10 (95%CI;2.5-26.4) months, respectively. Response rate (RR) was 16% and disease control rate (DCR) was 25.6%. Also in analysis, Grade 2 or more severe diarrhea was an independent prognostic factor for PFS (p = 0.004) and OS (p < 0.0001). The common treatment-related adverse events were gastrointestinal toxicity and hepatic dysfunction, but they were not severe and no treatment related death was observed.

Conclusions

Even though LEAP-017 was not effective, we suggest that some populations of patients could have benefits from this regimen as a salvage therapy for MSS colorectal cancer. Also, our findings assume that the possibility that Grade 2 or more severe diarrhea may be a predictive factor. In conclusion, we think that further investigations in this field are needed.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

99P - Efficacy and safety of toripalimab plus metronomic chemotherapy in HER2 negative metastatic breast cancer

Presentation Number
99P
Lecture Time
20:30 - 20:30
Speakers
  • Hongnan Mo (Beijng, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Combining immune checkpoint blockade (ICB) with chemotherapy may significantly improve efficacy in patients with breast cancer. Whether metronomic chemotherapy is more suitable for ICB than conventional chemotherapy is still unclear.

Methods

This study is a multicenter randomized phase 2 trial using a multi-arm design with Bayesian adaptive randomization and efficacy monitoring. Eligible patients have advanced HER2-negative breast cancer, with no more than one prior line of standard chemotherapy. Patients were randomized to 5 groups: 1) metronomic vinorelbine (NVB, 40 mg/day, TIW) monotherapy (the control cohort); 2) NVB + toripalimab (anti-PD1 antibody, 240 mg Q3W); 3) bevacizumab (5 mg/kg Q3W) + NVB+ toripalimab (the BEV cohort); 4) cisplatin (50mg/m Q3W) + NVB + toripalimab (the DDP cohort); 5) cyclophosphamide (50mg/day, QD) + capecitabine (500 mg, TID) + NVB+ toripalimab (the VEX cohort). The primary endpoint was disease control rate (DCR). Mass cytometry time-of-flight analyses of paired blood samples were performed to demonstrate dynamic changes in systemic immune profile.

Results

A total of 103 patients were randomized. The rate of nausea was significantly higher in the cisplatin cohort than in the others (P< 0.001). Among the five treatment cohorts, the VEX cohort and the cisplatin cohort had the highest DCR, 69.7% (95% CI 51.7–85.9%) and 73.7% (95% CI 56.1–88.7%), respectively. It is worth noting that the PFS of patients in the VEX cohort was the longest, reaching 6.6 months (95% CI 4.0-5.9). The PFS of patients in the DDP cohort was relatively short, only 3.5 months (95% CI 2.2-5.3). In the TNBC subgroup, again, patients in the VEX cohort had the highest DCR (74.1%, 95%CI 47.9%-95.4%) and longest PFS (9.8 months, 95%CI 3.8-21.9). We clustered CD45+ immune cells into 32 clusters. Only the change of cluster 30 differed between responders and non-responders. This is a group of intermediate monocytes with a high expression of CD38. Meanwhile, the overall expression of CD38 in monocytes was significantly increased by DDP and BEV treatment compared with baseline, but not in the VEX groups.

Conclusions

These data suggest promising clinical efficacy and evidence of cooperativity between metronomic VEX chemotherapy and PD-1 blockade.

Clinical trial identification

NCT04389073.

Legal entity responsible for the study

The authors.

Funding

CAMS Innovation Fund for Medical Sciences (CIFMS 2022-I2M-C&T-B-067).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

98P - Short-course Radiotherapy based Total Neoadjuvant Therapy combined with Tislelizumab in the Treatment of Locally Advanced Rectal Cancer: Mid-term Report of a Phase II, Open-Label, Randomized Controlled Trial

Presentation Number
98P
Lecture Time
20:30 - 20:30
Speakers
  • Wu Fengpeng (Shijiazhuang, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

100P - Phase II trial of tislelizumab plus bevacizumab and chemotherapy as the first-line therapy for persistent, recurrent, or metastatic cervical cancer: updated efficacy and safety results

Presentation Number
100P
Lecture Time
20:30 - 20:30
Speakers
  • Jianqing Zhu (Hangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Patients (pts) with persistent, recurrent, or metastatic cervical cancer are at high risk of progression after first-line standard treatment with platinum-doublet chemotherapy ± bevacizumab. Adding a PD-1 inhibitor to this standard treatment improves the survival of PD-L1+ pts. The preliminary results showed that tislelizumab plus bevacizumab and platinum-based chemotherapy had a promising efficacy with unconfirmed ORR of 78.4% (29/37, 95% CI, 61.8%-90.2%), irrespective of PD-L1 expression status (J Zhu, et al., JCO 2023 41:16_suppl). Here, we presented the updated results based on the full efficacy analysis set.

Methods

Eligible pts with persistent, recurrent, or metastatic cervical cancer received tislelizumab (200 mg, Q3W) plus bevacizumab (7.5 mg/kg, Q3W) and platinum-based chemotherapy. PD-L1 expression was accessed using VENTANA PD-L1(SP263) assay. The primary endpoint was PFS. Secondary endpoints were ORR, DCR, DOR and safety.

Results

A total of 50 pts were enrolled, with 46 (92.0%) having squamous cell carcinoma. 41 pts were detected for PD-L1 expression; 35 (85.4%) pts were PD-L1+, 6 (14.6%) were PD-L1. As of 10 Aug 2023, the median follow-up was 7.8 months (range 0.8-14.6). Among 47 pts in the efficacy analysis set, the confirmed ORR was 72.3% (34/47, 95% CI 57.4%-84.4%), with CR rate of 14.9% (6/47). Subgroup analysis showed that ORR for pts with squamous cell carcinoma and adenocarcinoma were 72.7% (32/44) and 66.7% (2/3), respectively; ORR for pts with PD-L1+ and PD-L1 tumors were 70.6% (24/34) and 100% (5/5), respectively. DCR was 100% (95% CI 92.4%-100.0%). Median DOR and PFS were not reached. After a median treatment duration of 6.8 months, grade ≥3 treatment-related adverse events occurred in 48.0% of pts. Immune-related adverse events (irAE) were reported in 32.0% of pts, with no grade ≥3 irAE reported. Serious adverse events occurred in 20.0% of pts.

Conclusions

The updated results further demonstrated the efficacy and tolerability of tislelizumab plus bevacizumab and chemotherapy, with increased CR rate observed during extended treatment duration.

Clinical trial identification

NCT05247619.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

101P - Progression-Free Survival is an acceptable surrogate endpoint for chemo-immunotherapy combinations in Cervical Carcinoma, an EORTC Young GCG study

Presentation Number
101P
Lecture Time
20:30 - 20:30
Speakers
  • Ramon Yarza (Sutton, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immunotherapy has become a standard of care for recurrent and metastatic cervical carcinoma resulting in the need to carefully assess for surrogacy focusing on new therapeutic scenarios. This study delves into a systematic review and meta-analysis to unravel the potential of progression-free survival (PFS) as a surrogate endpoint in cervical cancer.

Methods

We performed a comprehensive search of randomized phase 2 and 3 clinical trials in advanced and recurrent cervical cancer. PRISMA guidelines were followed, and the study was registered in PROSPERO (CRD42023405604). Inclusion criteria required RCTs with mature PFS and OS data. Trials with adjuvant treatment or disease-free survival as primary endpoint were excluded. Subgroup analysis was performed by type of treatment (i.e. chemotherapy-based, chemo-immunotherapy combinations).

Results

11,348 articles were screened, and 31 included in the final analysis. In the overall population, a robust correlation between PFS and OS was observed (Spearman's rho = 0.66, p-value < 0.001). We calculated the Surrogate Threshold Effect (STE) at the PFS intersection with null OS outcome (p = 0.05), resulting in a threshold for PFS z-score (z-PFS) of 2.23. When examining the results by treatment modality, chemotherapy trials exhibited a notably weak and non-significant correlation (Spearman 0.34, p = 0.31) with a significantly higher STE (z-PFS = 2.92). Conversely, in chemo-immunotherapy combinations, the correlation was notably robust and statistically significant (Spearman 0.94, p = 0.017), associated with a considerably lower STE (z-PFS = 2.1).

Conclusions

PFS may serve as a surrogate endpoint in cervical cancer. Notably, PFS was a more promising surrogate marker in the context of chemo-immunotherapy when compared to chemotherapy alone. This aligns with the hypothesis that chemo-immunotherapy induces a response, which is further sustained through maintenance immunotherapy, thereby enabling PFS to better capture the impact on OS. Conversely, in chemotherapy-based treatments, the expected survival benefit may be more limited, and PFS may not as strongly capture the effect of chemotherapy alone.

Legal entity responsible for the study

European Organisation for Research and Treatment of Cancer (EORTC), Young Gynaecological Cancer Group.

Funding

Has not received any funding.

Disclosure

F. Herrera: Financial Interests, Institutional, Research Grant, Grant/Research Support: Accuracy Inc, Bioprotext, BMS, Roche-ImFlame/ImCore, Nanobiotix, AstraZeneca, Eisai, MSD, Seagen; Financial Interests, Institutional, Research Grant, Grant/Research Support Foundations: Prostate Cancer Foundation, San Salvatore Foundation; Financial Interests, Personal, Advisory Board, Consultation, fees, travel expenses: Johnson & Johnson, Seagen, MSD, BMS, AstraZeneca, Eisai; Non-Financial Interests, Institutional, Non-financial benefits, Academic Collaborations: EORTC chairman GCG, ESMO Scientific Committee member for drug development, ASTRO Scientific Committee Annual Meeting. A. Madariaga Urrutia: Financial Interests, Personal, Invited Speaker: GSK, AstraZeneca, Clovis, MSD; Financial Interests, Personal, Advisory Board: GSK, AstraZeneca, PharmaMar. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

102P - Interim safety analysis of a phase 2 trial of cisplatin-sensitized radiation therapy and pembrolizumab for unresectable vulvar cancer

Presentation Number
102P
Lecture Time
20:30 - 20:30
Speakers
  • Oladapo O. Yeku (Boston, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Vulvar cancer is a rare disease with increasing incidence. Treatment can involve surgery and chemoradiation for local and regional disease, while systemic chemotherapy and immunotherapy are reserved for patients with distant metastases. Patients with unresectable or metastatic disease have relatively poor outcomes. Cisplatin and radiation (cis-RT) have been reported to have anti-tumor immunomodulatory properties in addition to their cytotoxic effects. We evaluated the addition of pembrolizumab (pembro) to cis-RT in vulvar cancer.

Methods

In this ongoing single-arm phase II trial (NCT04430699), patients with primary unresectable, incompletely resected, recurrent, or metastatic squamous cell carcinoma of the vulva undergoing RT were eligible. Patients who had received prior chemotherapy or immunotherapy were also eligible. Patients received cis 40 mg/m2 weekly concurrently with RT, and pembro 200 mg was administered every three weeks for a total of 12 cycles. The primary endpoint was ORR, and the secondary objective was 6-month RFS. Serum cytokines and HMGB-1 were assessed.

Results

At the time of the data cutoff, 15 patients were enrolled, and 14 were evaluable. Any grade adverse events (AE) occurred in all patients. Grade (G) 3 AEs occurred in 11 (78.6%) patients, most of which were related to cis. There were no G4 or higher AEs. There was one serious AE; a G3 acute kidney injury related to cis. G3 neutropenia and G3 ALT increase were observed in 4 (28.6%) and 1 (7.1%) patient respectively. The most common treatment-emergent adverse events (TEAE) were anemia (78.6%), diarrhea (78.6%), nausea (78.6%), fatigue (78.6%), and thrombocytopenia (50%). 18 AEs were related to pembro, and all were grade 1, except for G2 nephrotic syndrome in 1 patient (9%) and G2 hypothyroidism in 2 patients (14%). Patients with progressive disease were more likely to have elevated serum levels of CCL4, CCL11, and CXCL12 at cycle 3.

Conclusions

In this interim analysis, concurrent treatment with cis-RT and pembro did not lead to any unexpected AEs. Cytokine analysis might yield insight into the mechanisms of resistance. This study, the first to evaluate pembrolizumab with cis-RT, is ongoing, with a target enrollment of 24 patients.

Clinical trial identification

NCT04430699.

Legal entity responsible for the study

The authors.

Funding

Merck Sharp & Dohme Corporation.

Disclosure

O.O. Yeku: Financial Interests, Personal, Advisory Board: hC Bioscience; Financial Interests, Personal, Speaker, Consultant, Advisor: TigaTx Inc, GIMV NV; Other, Institutional, Principal Investigator: Merck Sharp & Dohme Corp, Avant Immunotherapeutics, Compugen, Ascendis Pharma A/S, ProfoundBio, Avenge Bio, Immunocore Limited, Duality Biologics; Financial Interests, Institutional, Principal Investigator: Department of Defense. S. Bouberhan: Financial Interests, Personal, Speaker, Consultant, Advisor: ImmunoGen. C.M. Castro: Financial Interests, Personal, Speaker, Consultant, Advisor: Qiagen, Teladoc, Aikili Biosystems, InfiniteMD. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

103P - Long-term survivorship rates among previously treated patients with advanced renal cell carcinoma (aRCC) achieving objective response with nivolumab

Presentation Number
103P
Lecture Time
20:30 - 20:30
Speakers
  • Saby George (Buffalo, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Durability of response with immune checkpoint inhibitors is often associated with long-term survival benefits. This analysis explored the relationship between achieving objective response (OR) with nivolumab (NIVO) and the possibility of previously treated patients with aRCC being long-term survivors (LTS) in the phase 3 CheckMate 025 trial.

Methods

Mixture cure models (MCMs) were applied to analyze overall survival (OS) and duration of response (DoR) data with ≥7 years of follow-up separately for patients achieving confirmed OR (n=94, 22.9%) with NIVO. In both analyses, LTS were only subject to risk of non–disease-related (NDR) mortality. In the DoR analysis, LTS were also assumed to remain in response until death. OS/DoR outcomes for non-LTS were subject to both disease-related and NDR risk, and modeled by parametric distributions. NDR mortality rates were derived via publicly available World Health Organization lifetable data matched to the study population’s demographic characteristics. The fraction of LTS and OS/DoR for non-LTS were estimated simultaneously through maximum likelihood methods. Candidate MCMs were evaluated based on statistical fit criteria, visual fits to the observed OS/DoR data, and corresponding hazard trends.

Results

Across all clinically plausible candidate models, log-normal MCMs provided the best fit to the observed data for both endpoints, and estimated the fractions of LTS (95% CI) among responders as 31.5% (12.1%–60.5%) and 7% (1.1%–33.4%) from the OS and DoR data, respectively. The range of estimated fractions of LTS across all plausible models was narrower in the DoR analysis (5.7%–11.2%) than in the OS analysis (28.6%–41.1%). Projected 30-year mean OS and DoR for responders from the best-fitting MCMs were 9.35 and 3.30 years, respectively.

Conclusions

MCMs estimated the subgroup achieving OR with NIVO in the CheckMate 025 trial to have a higher fraction of LTS than those previously reported for the entire NIVO arm and a modest proportion of LTS to be in response until death. Even with long-term follow-up data, estimated fractions of LTS were subject to uncertainty due to the limited number of patients contributing to the formation of OS and DoR plateaus.

Clinical trial identification

NCT01668784.

Legal entity responsible for the study

Bristol Myers Squibb.

Funding

Bristol Myers Squibb.

Disclosure

S. George: Financial Interests, Personal, Advisory Board, advisor/consultant: BMS, Bayer, Pfizer, Exelixis, Sanofi/Genzyme, Seattle Genetics, EMD Serono, Eisai, Merck, Aveo, QED therapeutics; Financial Interests, Personal, Advisory Board, Advisor/consultant: Novartis, AstraZeneca; Financial Interests, Institutional, Local PI: Pfizer, Merck, Agensys, Novartis, BMS, Bayer, Eisai, Seattle Genetics, Surface Oncology, Exelixis, Aravive, Aveo, Gilead. J. Larkin: Financial Interests, Personal, Invited Speaker: BMS, Pfizer, Roche, Pierre Fabre, AstraZeneca, Novartis, EUSA Pharma, MSD, Merck, GSK, Ipsen, Aptitude, Eisai, Calithera, Ultimovacs, Seagen, Goldman Sachs, eCancer, Inselgruppe, Agence Unik; Financial Interests, Personal, Other, Consultancy: Incyte, iOnctura, Apple Tree, Merck, BMS, Eisai, Debipharm; Financial Interests, Personal, Other, Honorarium: touchIME, touchEXPERTS, VJOncology, RGCP, Cambridge Healthcare Research, Royal College of Physicians; Financial Interests, Institutional, Funding: BMS, MSD, Novartis, Pfizer, Achilles, Roche, Nektar, Covance, Immunocore, Pharmacyclics, Aveo. L.M. Garcia Fernandez: Financial Interests, Institutional, Advisory Role: Parexel International. J. May, M. Dyer, M. Kurt: Financial Interests, Personal, Full or part-time Employment: Bristol Myers Squibb; Financial Interests, Personal, Stocks or ownership: Bristol Myers Squibb. M. Patel: Financial Interests, Personal, Full or part-time Employment: Bristol Myers Squibb/Medarex, LEO Pharma; Other, Personal, Other, An Immediate Family Member: Pfizer, CVS Health; Financial Interests, Personal, Stocks or ownership: Sanofi, Bristol Myers Squibb/Medarex; Financial Interests, Personal, Stocks or ownership, An Immediate Family Member: CVS Health. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

104P - Bladder-Sparing Treatment for muscle-invasive bladder carcinoma (MIBC): A single-center, single-arm clinical study of sequential neoadjuvant chemotherapy followed by tislelizumab neoadjuvant immunotherapy.

Presentation Number
104P
Lecture Time
20:30 - 20:30
Speakers
  • Yu Zeng (Shenyang, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Bladder-sparing treatment has emerged as an alternative choice for patients who are concerned about the life quality after radical cystectomy (RC). The optimal strategy for bladder-sparing treatment such as trimodal therapy (TMT) showed similar effects with RC. Since neoadjuvant immunotherapy has illuminated considerable response in MIBC, we aim to study whether neoadjuvant chemotherapy plus immunotherapy can improve the bladder-sparing rate in MIBC patients.

Methods

30 planned patients with MIBC (T2-4a N0-1 M0) received cisplatin 70mg/m2 or carboplatin AUC 4.5 on day 1 every 3 weeks (Q3W) plus gemcitabine 1000 mg/m2 on the 1st and 8th day of each 21-day cycle x 4 cycles. Tislelizumab 200mg was administered on the 14th day of each 21-day cycle at the 3rd and 4th cycles. CT and cystoscopy imaging were carried out to evaluate disease progression. After the 4th treatment, radical cystectomy, partial cystectomy or TURBT were perform in accordance with the disease status. For continuous bladder-sparing treatment, 2 additional cycles of tislelizumab were performed. Bladder-sparing rate was settled as the exploratory endpoint based on 2-years of follow-up and predictive biomarker will be analyzed.

Results

To date, 21 of 30 pts have been enrolled and 17 pts have completed the regimen. 2pts were excluded because of complicating with other disease (cerebral infraction or rectal cancer), and 2pts voluntarily quit due to intolerance of chemotherapeutic adverse effect (fatigue and gastrointestinal symptoms). 2pts showed partial response to the therapy and received RC after disease progression. 1pt have no response to the therapy and received palliative care as unsuitable for surgery. The remaining 10 of 17 pts successfully preserved their bladder by achieving pT0 (58.8%) after treatment. Among them, 3 pts have been followed up over 1 year and no relapse was observed.

Conclusions

These data support neoadjuvant chemotherapy plus immunotherapy as a feasible bladder-sparing choice for MIBC patients. Further completed follow-up data and biomarker analysis will accurately identify patients who are suitable for this therapy.

Clinical trial identification

ChiCTR2100050763.

Legal entity responsible for the study

The author.

Funding

Has not received any funding.

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

105P - Preliminary efficacy and safety results from ‘ReBirth’, a phase II study of risk-based bladder-sparing therapy for MIBC.

Presentation Number
105P
Lecture Time
20:30 - 20:30
Speakers
  • Yijun Shen (Shanghai, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Trimodal therapy (TMT) has achieved long-term survival and persistent oncologic control in selected MIBC patients, however, tailored treatment using biomarkers based on chemotherapy plus PD-1 inhibitor responses is currently absent. Furthermore, the safety and efficacy of hypo-fractionated radiation in combination with PD-1 inhibitors and concurrent chemotherapy is worth exploring.

Methods

This is a two-stage, single-arm, phase II trial recruiting cT2-4aN0-1M0 MIBC pts. Based on results of cystoscopy, urine cytology, imaging and MRD detection after first stage (Tislelizumab (T) 200 mg on D1, Cisplatin (C) 70 mg/m2 on D1 and Gemcitabine (G) 1000 mg/m2 on D1 and D8 Q3W for 3-4 cycles), pts achieving cCR (cT0, cTa) are treated with T, while the other pts receive T and chemoradiotherapy (whole bladder 44Gy/16 fractionation combined with C as radiosensitizer). 1-year BIDFS rate is the primary endpoint. Secondary endpoints include 2-year MFS rate, 2-year BIDFS rate and safety. Tissue and urine samples will be obtained for genetic profiling and biomarker research.

Results

As of September 20, 2023 (follow up: 258 (49-415) days, 25 pts with a median age of 64 (36-77) yrs were enrolled and 96% are male. 19 pts with cT2 (57.14%), cT3 (33.33%) and cT4 (9.52%) tumors were evaluable. 2 pts were assessed as N1. 14/19 pts (73.68%) achieved cCR and maintained a sustained response. Based on positive urine cytology and MRD, 5 pts were classified as non-cCR (2 pts in cT3N0M0, 2 pts in cT4N0M0 and 1 pt in cT2N0M0). Due to RC (2 pts achieving ypT0N0 and 1 pt achieving ypT2N0) and incomplete treatment cycles, 6 pts were excluded from the efficacy analysis set. BFS rate at 1 yr is evaluated in 6 pts (2 pts received hypo-fractionated on the second stage) and the rate is 100%. TRAEs were found in 19 of 25 pts (76%, 8% are ≥ 3 grade). Hematological AEs (32%), renal insufficiency (24%), pruritus (20%), and fatigue (20%) are the most common AEs. During hypo-fractionated radiation, no new safety signs were discovered.

Conclusions

The preliminary findings indicate a potential efficacy and manageable toxicity during the two-stage treatment. Enrollment is still ongoing, and long-term efficacy will be proved.

Clinical trial identification

NCT05531123.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

106P - Treatment Sequencing in PD-L1-Positive Recurrent/Metastatic (R/M) Head and Neck Squamous Cell Carcinoma (HNSCC): Exploratory Analysis of the Phase 3 KEYNOTE-048 Study

Presentation Number
106P
Lecture Time
20:30 - 20:30
Speakers
  • Amanda Psyrri (Athens, Greece)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Pembrolizumab (pembro) and pembro + platinum + 5-FU (pembro + chemo) are standard-of-care options alongside cetuximab-based regimens such as EXTREME (cetuximab + platinum + 5-FU) for R/M HNSCC, depending on PD-L1 expression and clinical circumstances, but optimal sequencing has not been defined. We present OS in pts with PD-L1+ R/M HNSCC who received first-line (1L) pembro or pembro + chemo followed by cetuximab-based therapy, or EXTREME followed by immunotherapy (IO), in KEYNOTE-048 (NCT02358031).

Methods

Pts with locally incurable R/M HNSCC were randomly assigned 1:1:1 to 1L pembro, pembro + chemo, or EXTREME. This post hoc exploratory analysis included pts with PD-L1 CPS ≥1 who received second-line (2L) cetuximab-based therapy (cetux) after pembro or pembro + chemo, or 2L IO after EXTREME.

Results

Of 882 pts, 170 with PD-L1 CPS ≥1 who received 2L cetux or IO were included (1L pembro, n = 63; pembro + chemo, n = 37; EXTREME, n = 70). Median follow-up was 69.3 mo (range, 61.2-81.2). Median OS (95% CI) was 15.7 mo (12.3-22.6) for pts who received pembro followed by cetux vs 16.5 mo (13.5-19.8) for EXTREME followed by IO (HR, 1.0; 95% CI, 0.7-1.5); 24-mo OS was 32% vs 31%. Median OS (95% CI) was 22.1 mo (14.0-24.7) for pts who received pembro + chemo followed by cetux vs 16.5 mo (11.5-19.8) for EXTREME followed by IO (HR, 0.7; 95% CI, 0.4-1.0); 24-mo OS was 39% vs 30%. Median OS (95% CI) was 16.6 mo (13.8-23.2) for all pts who received pembro or pembro + chemo (n = 100) followed by cetux vs 16.5 mo (13.5-19.8) for EXTREME followed by IO (HR, 0.9; 95% CI, 0.6-1.2); 24-mo OS was 34% vs 31%. Of pts receiving pembro or pembro + chemo, 13 received subsequent cetux monotherapy and 87 received cetux + chemo.

Conclusions

In this post hoc exploratory analysis of KEYNOTE-048, OS was longer for pts who received pembro + chemo followed by cetux vs EXTREME followed by IO and was similar for pts who received pembro followed by cetux vs EXTREME followed by IO. Clinical factors may have contributed to different use of 2L therapies by treatment arm. These findings, in addition to the primary analysis of KEYNOTE-048, support 1L pembro and pembro + chemo in PD-L1–positive R/M HNSCC.

Clinical trial identification

NCT02358031.

Editorial acknowledgement

Medical writing and/or editorial assistance was provided by Matthew Grzywacz, PhD of ApotheCom (Yardley, PA, USA).

Legal entity responsible for the study

Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA.

Funding

Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA.

Disclosure

R. Greil: Financial Interests, Personal, Advisory Board: Celgene, Novartis, Roche, BMS, Takeda, AbbVie, AstraZeneca, Janssen, MSD, Merck, Gilead, Daiichi Sankyo, Sanofi; Financial Interests, Institutional, Stocks/Shares: Novo Nordisk, Lilly; Financial Interests, Personal and Institutional, Funding: Celgene, Roche, Merck, Takeda, AstraZeneca, Novartis, Amgen, BMS, MSD, Sandoz, AbbVie, Gilead, Daiichi Sankyo; Non-Financial Interests, Personal, Other, Travel, Accomodation, Expenses, Consulting, Adv Role: Roche, Amgen, Janssen, AstraZeneca Novartis, MSD, Celgene, Gilead, BMS, AbbVie, Daiichi Sankyo, Sanofi; Other, Personal, Other, Travel, Accomodation, Expenses, Consulting, Adv Role, Honoraria: Roche, Amgen, Janssen, AstraZeneca, Novartis, MSD, Celgene, Gilead, BMS, AbbVie, Daiichi Sankyo, Sanofi. B. Burtness: Financial Interests, Personal, Advisory Board: Merck, Vaccinex, Coherus; Financial Interests, Personal, Invited Speaker: Genentech. S. Laban: Financial Interests, Institutional, Advisory Board: Merck Sharp & Dohme, Bristol Myers Squibb, Sanofi Genzyme; Financial Interests, Institutional, Invited Speaker: Merck Sharp & Dohme, Bristol Myers Squibb; Financial Interests, Personal, Advisory Board: Merck Sharp & Dohme; Financial Interests, Institutional, Other, patent for an oropharyngeal cancer multi-peptide vaccine (pending): filed patent (patent pending); Non-Financial Interests, Personal, Principal Investigator: Immutep, Merck Sharp & Dohme, Bristol Myers Squibb, ISA-Pharmaceuticals. I. Braña: Financial Interests, Personal, Advisory Board: Achilles Therapeutics, Bristol Myers Squibb, Cancer Expert Now, eTheRNA Immunotherapies, Merck Serono, Merck Sharp & Dohme (MSD), Rakuten Pharma, Boehringer Ingelheim, PCI Biotech, Guidepoint; Financial Interests, Personal, Invited Speaker: Bristol Myers Squibb, Merck Serono, Merck Sharp & Dohme (MSD), Roche; Financial Interests, Institutional, Local PI: AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, GSK, Gliknik, Incyte, ISA Pharmaceuticals, Janssen Oncology, Kura, Merck Serono, Novartis, Debiopharm, Merck Sharp & Dohme (MSD), Nanobiotix, Northern Biologics, Regeneron, Pfizer, Seattle Genetics, Shattuck Labs, VCN Biosciences, Roche, Immutep, MacroGenics, Sanofi, PharmaMar, Odonate Therapeutics, Bicycle Therapeutics, Dragonfly therapeutics, Gilead; Non-Financial Interests, Personal, Principal Investigator, Basket of baskets: Cancer Core Europe; Non-Financial Interests, Personal, Member, Head and Neck Group: EORTC; Non-Financial Interests, Personal, Member: SEOM, ASCO. R. Mesia Nin: Financial Interests, Personal, Advisory Board: Merck, MSD, Bayer, Seattle Genetics, Nanobiotix, Boehringer, Segean; Financial Interests, Personal, Invited Speaker: Merck, MSD, BMS; Non-Financial Interests, Personal, Principal Investigator, Clinical Trial PI: BMS; Non-Financial Interests, Personal, Principal Investigator, Observational trial PI: Merck. T. Fuereder: Financial Interests, Personal, Advisory Board: MSD, Pfizer, Boehringer Ingelheim, Sanofi, Merck KGaA, Amgen, Bristol Myers Squibb, Janssen; Financial Interests, Personal, Invited Speaker: MSD, Pfizer, Boehringer Ingelheim, Sanofi, Merck KGaA, Amgen, Bristol Myers Squibb, Janssen; Financial Interests, Personal, Research Funding: Merck Sharp & Dohme; Financial Interests, Personal, Research Grant: Merck Sharp & Dohme; Financial Interests, Personal and Institutional, Principal Investigator: Merck Sharp & Dohme, Roche, Merck KGaA, Amgen, Bristol Myers Squibb; Financial Interests, Personal and Institutional, Research Funding: Roche, Merck KGaA, Amgen, Bristol Myers Squibb; Financial Interests, Personal and Institutional, Research Grant: Roche, Merck KGaA, Amgen, Bristol Myers Squibb. î Bratland: Financial Interests, Personal and Institutional, Invited Speaker: MSD, BMS, Sanofi; Financial Interests, Personal and Institutional, Advisory Board: MSD, Sanofi; Financial Interests, Personal and Institutional, Principal Investigator: MSD, BMS; Financial Interests, Personal and Institutional, Advisory Role: Sanofi. L.F.L. Licitra: Financial Interests, Personal, Advisory Board, for expert opinion in advisory boards: AstraZeneca, Bayer, BMS, Eisai, MSD, Boehringer Ingelheim, Hoffmann-La Roche Ltd, Novartis, Roche, Debiopharm International SA, Sobi, Incyte Biosciences Italy srl, Doxa Pharma srl, Amgen, Nanobiotics, GSK; Financial Interests, Institutional, Research Grant, Funds received by my institution for clinical studies and research activities in which I am involved: AstraZeneca, BMS, Boehringer Ingelheim, Celgene International, Eisai, Exelixis, Debiopharm International SA, Hoffmann-La roche ltd, IRX Therapeutics, Medpace, Merck Serono, Merck Healthcare KGaA, MSD, Novartis, Pfizer, Roche, Adlai Nortye. A. Psyrri: Financial Interests, Personal, Invited Speaker: MSD, Merck Serono; Financial Interests, Personal, Advisory Board: Pfizer, Sanofi, MSD, AstraZeneca, BMS, Leo, Rakuten, eTheRNA immunotherapies, Merck Serono; Financial Interests, Personal and Institutional, Local PI: AstraZeneca, Pfizer, GSK, Genesis, MSD, Incyte, Amgen, Debiopharm, Janssen, Lilly, Regeneron, Sanofi, BI, Roche, Peregrine, Oncolytics Biotech; Financial Interests, Personal, Coordinating PI: AstraZeneca; Financial Interests, Personal and Institutional, Steering Committee Member: Iovance, Pfizer, Roche; Financial Interests, Institutional, Steering Committee Member: Kura Oncology; Financial Interests, Personal, Steering Committee Member: Kura Oncology; Financial Interests, Personal and Institutional, Funding: Kura Oncology, BMS, Roche, DEMO, Amgen, BI, Genesis, BMS, Pfizer, Oncolytics Biotech; Financial Interests, Institutional, Funding: Merck Serono, Pfizer; Financial Interests, Personal, Other, Educational activity: Medscape, PrimeOncology; Financial Interests, Institutional, Local PI: Novartis, Replimmune; Non-Financial Interests, Personal, Project Lead, Medical Education with honoraria: Medscape. B. Hughes: Non-Financial Interests, Personal, Advisory Board: MSD, BMS, Sanofi, Eisai; Financial Interests, Institutional, Research Grant: Amgen. M. Tahara: Financial Interests, Personal, Advisory Board: Merck Biopharma, Ono Pharma, Bristol Myers Squibb, MSD, Pfizer, Bayer, Lilly, Rakuten Medical, Boehringer Ingelheim, AstraZeneca; Financial Interests, Personal, Invited Speaker: Eisai; Financial Interests, Institutional, Local PI: MSD, AstraZeneca, Ono Pharma, Novartis, Pfizer, Bristol Myers Squibb, Loxo, GSK, Lilly, Rakuten Medical, Bayer, Merckbiophama; Financial Interests, Institutional, Research Grant: Bayer. J. Lin, B. Gumuscu, N. Lerman: Financial Interests, Personal, Full or part-time Employment: Merck; Financial Interests, Personal, Stocks/Shares: Merck. K.J. Harrington: Financial Interests, Institutional, Advisory Board: Amgen, AstraZeneca, BMS, Boehringer Ingelheim, Merck, MSD, Pfizer, Replimune, Oncolys, Vyriad, Idera; Financial Interests, Institutional, Other, Honoraria for lectures: Amgen, AstraZeneca, BMS, Boehringer Ingelheim; Financial Interests, Institutional, Advisory Board, Advisory board for CD47 assets: Arch Oncology; Financial Interests, Institutional, Advisory Board, Development of DDR assets: ARTIOS; Financial Interests, Institutional, Advisory Board, Development of exosomal STING agonist: Codiak; Financial Interests, Institutional, Advisory Board, Development of oncolytic adenovirus: PsiVac; Financial Interests, Institutional, Funding, Research: AstraZeneca, Boehringer Ingelheim, MSD; Financial Interests, Institutional, Funding, Development of oncolytic HSV platform: Replimune; Non-Financial Interests, Personal, Leadership Role, Chair of Steering Committee: ART NET; Non-Financial Interests, Personal, Other, Member of Global Steering Committee: MR - Linac. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

108P - Real-world (RW) effectiveness and safety of adjuvant nivolumab (NIVO) in patients (pts) with melanoma in Belgium and Luxembourg: PRESERV MEL

Presentation Number
108P
Lecture Time
20:30 - 20:30
Speakers
  • Bart Neyns (Brussels, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Adjuvant NIVO is approved for pts with melanoma with lymph node involvement/metastatic disease who have undergone complete resection. PRESERV MEL is an observational study aiming to describe effectiveness, safety, and quality of life in pts treated with adjuvant NIVO in the RW. Updated results are shown.

Methods

Pts were enrolled prospectively/retrospectively over a 2-y period and are being followed for 5 y. Index date was first NIVO administration. Pts received NIVO for ≤ 12 mo per label. Recurrence-free survival (RFS), distant metastasis-free survival (DMFS; including baseline stage IV pts), time to discontinuation (TTD), subsequent treatment (tx), and adverse events (AEs)/tx-related AEs (TRAEs) were assessed.

Results

In total, 152 pts (125 prospective; 27 retrospective) were enrolled (Jan 2019–Dec 2020) at 15 sites in Belgium and Luxembourg. Median follow-up was 25 mo. Median TTD was 11.1 mo. All pts ended tx due to tx completion (56%), AE (23%), recurrence (15%), pt decision unrelated to AE (1%), or other reason (5%). Median RFS was 47.2 mo, 2-y RFS rate was 62%, median DMFS was not reached, and 2-y DMFS rate was 72% (Table). Sites of first recurrence in pts with distant recurrence (n = 41; 27%) were soft tissue (39%), lungs (34%), multiple organs (37%), skin (27%), liver (22%), lymph nodes (22%), bone (15%), brain (7%), intestines (5%), other visceral organs (17%), and other (2%). Subsequent tx was used in 51 pts (34%), including surgery (9%), radiotherapy (8%), and systemic tx (32% [adjuvant tx, 10%; unresectable/metastatic tx, 22%]). Median time from NIVO discontinuation (D/C) to subsequent tx was 12.6 mo. Grade 3/4 TRAEs occurred in 21 pts (14%). Any AEs led to tx D/C in 35 pts (23%). Late-emergent grade 3/4 TRAEs (100 d–2 y after tx) were reported in 3 pts (2%; 2 of 3 pts had prior TRAE occurrence).

Adjuvant NIVO (n = 152)
Median RFS, mo (95% CI) 47.2 (33.2–NR)
RFS rate, % (95% CI)
1 y 75 (67–81)
2 y 62 (54–70)
3 y 56 (46–65)
Recurrence, n (%) 59 (39)
Local 17 (11)
Regional 14 (9)
Distant 26 (17)
Died before recurrence, n (%) 3 (2)
Median DMFS, mo (95% CI) NR (45.7–NR)
DMFS rate, % (95% CI)
1 y 80 (73–86)
2 y 72 (64–79)
3 y 66 (57–74)
Distant recurrence, n (%) 41 (27)
Died before distant recurrence, n (%) 6 (4)

NR, not reached.

Conclusions

RW effectiveness and safety of adjuvant NIVO in pts with resected stage III/IV melanoma in PRESERV MEL were consistent with CheckMate 238 results.

Editorial acknowledgement

Medical writing support for the development of this abstract, under the direction of the authors, was provided by Mark Palangio of Ashfield MedComms, an Inizio company, and funded by Bristol Myers Squibb.

Legal entity responsible for the study

Bristol Myers Squibb.

Funding

Bristol Myers Squibb.

Disclosure

B. Neyns: Financial Interests, Institutional, Advisory Board: Novartis, Bristol Myers Squibb, Pierre Fabre, MSD (Merck Sharp & Dohme); Financial Interests, Institutional, Research Grant: Novartis, Pfizer; Non-Financial Interests, Institutional, Product Samples: Bayer. L. McDonald: Financial Interests, Personal, Full or part-time Employment: Bristol Myers Squibb; Financial Interests, Personal, Stocks or ownership: Bristol Myers Squibb. H. Van Campenhout: Financial Interests, Personal, Full or part-time Employment: Bristol Myers Squibb; Financial Interests, Personal, Stocks or ownership: Bristol Myers Squibb. C. Jacobs: Financial Interests, Institutional, Advisory Board: BMS, Novartis, Pierre Fabre, MSD; Financial Interests, Institutional, Invited Speaker: BMS. A. Rogiers: Financial Interests, Personal, Advisory Board: MSD, BMS. A. Rorive: Financial Interests, Personal, Advisory Board: BMS, Novartis, MSD, Pierre Fabre; Financial Interests, Personal, Invited Speaker: BMS, Novartis, MSD, Pierre Fabre. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

109P - Prognosis of patients with metastatic melanoma with initial stable disease during treatment with anti-PD-1 monotherapy

Presentation Number
109P
Lecture Time
20:30 - 20:30
Speakers
  • Inge M. Noringriis (Herlev, Denmark)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Stable disease (SD) upon treatment with immune checkpoint inhibitors is generally associated with poorer survival outcomes than partial (PR) or complete response (CR). Yet, in the KEYNOTE-001 and -006 cohort, approximately half of patients with initial SD subsequently developed an objective response (OR) with improved survival outcomes. We present a real-world analysis of the prognosis of unresectable melanoma patients with SD at first evaluation after initiation of anti-PD-1 monotherapy.

Methods

The Danish Metastatic Melanoma Database (DAMMED) is a nation-wide registry on all Danish patients with unresectable melanoma eligible for systemic therapy. All patients diagnosed with unresectable cutaneous or unknown primary melanoma treated with single agent anti-PD-1 therapy, regardless of treatment line, were selected for analysis.

Results

1052 patients, treated between July 2014 and April 2022, were found eligible for analysis. At the initial evaluation, approximately 12 weeks after first anti-PD-1 administration, 232 patients (22.1%) had SD. Of these, 66 (28.4%) patients were alive and progression-free after a median follow up of 38.0 months while 87 (37.5%) later achieved PR (45) or CR (42). Overall, patients with initial SD had a median progression free survival (PFS) of 13.7 months and a median overall survival (OS) of 43.0 months. Grouping patients with initial SD by best overall response (BOR) revealed significantly different survival curves (p<0.001 for SD vs PR vs CR for both PFS and OS). In contrast, no significant differences were found when comparing the survival curves of patients with initial SD and subsequent OR to those of patients with initial OR (PFS: p=0.46 and p=0.83 for PR and CR, respectively; OS: p=0.69 and p=0.81, respectively). OS of patients with BOR SD was significantly better than the OS of patients with immediate PD (p<0.001).

Conclusions

More than one-third of patients with initial SD later obtain an OR with a prognosis similar to patients having the corresponding OR on first evaluation. Durable disease control is a common outcome following initial SD to anti-PD-1 monotherapy in melanoma.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

I.M. Noringriis: Financial Interests, Institutional, Research Grant, Research granted to our institution in order to initiate a biobank containing blood samples from patients with immune related adverse events. I am the principle investigator of the protocol: Sygeforsikringen, Novo Nordisk Fonden. M. Donia: Financial Interests, Personal, Other, Advisor: Achilles Therapeutics; Non-Financial Interests, Personal, Other, Sub-investigator of clinical trial with connected translational research: Bristol Myers Squibb; Non-Financial Interests, Personal, Proprietary Information, Proprietary data access: Bristol Myers Squibb; Non-Financial Interests, Personal, Proprietary Information, Proprietary data access: Genentech; Other, Personal, Other, Chairman of the Melanoma and Non-melanoma Skin Cancer Scientific Committee: Danish Medicines Council (Medicinrådet). L. Bastholt: Non-Financial Interests, Personal, Advisory Role, Scientific committee under Danish Medicines Agency regarding new treatments of melanoma, skin cancer and thyroid cancer: Danish Medicines Agency. C.A. Haslund: Financial Interests, Personal, Invited Speaker: MSD, GSK, BMS; Financial Interests, Institutional, Local PI: BMS, Tesaro, MSD, IO Biotech, Chimerix, Incyte; Financial Interests, Institutional, Coordinating PI: GSK, Celgene Aps. E. Ellebæk: Financial Interests, Personal, Invited Speaker: Pierre Fabre, BMS, Novartis, MSD, Pfizer; Other, Personal, Other, Travel and conference expenses: MSD, Pierre Fabre. I. Svane: Financial Interests, Personal, Advisory Board: BMS, Pierre Fabre, Novartis; Financial Interests, Personal, Invited Speaker: MSD, Pierre Fabre, Novartis, Roche, BMS; Financial Interests, Personal, Writing Engagement: MSD; Financial Interests, Personal, Stocks/Shares, Cofounder and Founder warrents: IO Biotech; Financial Interests, Institutional, Research Grant: Adaptimmune, Enara Bio, Lytix Biopharma, TILT Biotherapeutics; Financial Interests, Institutional, Funding: Evaxion; Non-Financial Interests, Personal, Principal Investigator: BMS, Novartis, Roche, TILT Biotherapeutics, Lytix Biopharma. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

110P - Outcomes of CUPem: A prospective Phase II multicentre clinical Trial of Pembrolizumab in patients with pre-treated Cancer of Unknown Primary

Presentation Number
110P
Lecture Time
20:30 - 20:30
Speakers
  • Harpreet S. Wasan (London, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Cancer of unknown primary (CUP) remains a common major challenge, diagnosed by serially excluding known sites of origin. Median survival, excluding rarer favourable subsets, with first-line combination cytotoxics (platinum based) remain poor under 10 months. Less than 10% are amenable to therapies linked to actionable mutations and less than 20% suitable for 2nd line therapies, with few studies beyond first line therapy and no standard of care. CUP may have unique biology in relation to immune system evasion leading to early metastatic spread. We investigated the feasibility of treatment with Pembrolizumab after failure of at least one line of therapy in a prospective phase 2 trial.

Methods

The study was initiated in 2019 to recruit a minimum of 57 CUP patients treated with and progressing after at least one line of chemotherapy with RECIST measurable disease, across 3 UK centres with Pembrolizumab 200 mg IV Q21d until disease progression or intolerance. Significant Covid trial recruitment disruption in this rare population, revised the study design down to a minimum of 31 patients, (after an interim pre-planned futility analysis at 24 patients) which gave a power 0.80 to detect a 2 months PFS improvement.

Results

35 patients consented with 30 patients assessable (Database lock Aug 2023). Median age 60 (range 33–77), 67% female, ECOG 0-1 vs 2: 83/17%. 63% had 1 line of prior chemotherapy, 27% 2 lines, 10% 3 or more lines & 10% prior radiotherapy. Median PFS (first RECIST PD) was 4.0 months (95% CI 3.2-7.5) and Median OS 11.5 months (95% CI 6.5-NR). At 6/12/18/24 & 36 months, the percentage of patients not progressing on trial treatment were 33/23/17/13/13% respectively. 2 patients who had not progressed, stopped treatment due to related AEs, one of whom has maintained stable disease for >36 mths including 9 months after discontinuation. Pembrolizumab was very well tolerated and there were no unexpected adverse events.

Conclusions

Pembrolizumab should be further investigated in CUP with promising activity and sustained disease control in a significant minority of patients beyond 2nd line with overall survival similar to first line CUP studies. Ongoing Clinico-translational research may identify predictive biomarkers.

Clinical trial identification

EudraCT: 2018-001327-39, NCT03752333.

Editorial acknowledgement

We acknowledge the contribution of Dr Michael Grayer from Floating Point Statistics Ltd who provided statistical consultancy services, supporting this conference paper by analysing the interim data as it stood on 30 August 2023, and producing production-quality tables and figures.

Legal entity responsible for the study

Imperial College London.

Funding

This research has been supported by 1) Grant Ref: MISP 55449 from Merck Sharp and Dohme Limited (MSD); 2) CUPFoundation UK; 3) SR was supported by NHS funding to the National Institute for Health and Care Research Biomedical Research Centre at Royal Marsden NHS Foundation Trust and the Institute of Cancer Research.

Disclosure

H.S. Wasan: Financial Interests, Personal, Advisory Board, Advisory Boards and Invited Speaker: Incyte, Pierre Fabre, Servier, Bayer, Roche/Genentech/FM AG, Sirtex Medical Erytech, Celgene, Array BioPharma, Merck KGaA: BMS; Financial Interests, Personal, Steering Committee Member, Trial steering committee and advisory: Zymeworks; Financial Interests, Personal and Institutional, Coordinating PI, Trial PI steering committee and advisory: Sirtex Medical; Financial Interests, Personal, Other, Consultancy for Submission for UK approval (BSI): ONCOSL; Non-Financial Interests, Personal, Advisory Role, Trial steering committee: Pfizer; Non-Financial Interests, Personal, Advisory Role, UK NICE submission: Bayer, Pierre Fabre. S. Rao: Financial Interests, Personal, Advisory Board: Merck Serono, Servier, Seagen, AstraZeneca, Hoopika, Bayer, BeiGene; Financial Interests, Personal, Expert Testimony: Boehringer; Financial Interests, Personal, Invited Speaker: Merck Serono, Servier, Bayer. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

111P - Characteristics and outcomes of immunotherapy-related liver injury in patients with hepatocellular carcinoma compared to patients with advanced solid tumours

Presentation Number
111P
Lecture Time
20:30 - 20:30
Speakers
  • Ciro Celsa (London, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immune-related liver injury (irLI) is commonly observed in patients with cancer treated with immune checkpoint inhibitors (ICIs). In this comparative study, we aimed to compare incidence, clinical characteristics and outcomes of irLI between patients receiving ICIs for HCC versus other solid tumour indications.

Methods

Two separate cohorts were included: 375 patients with advanced/unresectable HCC, Child-Pugh A class treated with first-line Atezolizumab+Bevacizumab from AB-real study and a non-HCC cohort, including 459 patients treated with first-line ICI therapy from INVIDIa-2 multicentre study. IrLI was defined as treatment-related increase of transaminases levels after exclusion of alternative aetiologies of liver injury. Incidence of irLI was adjusted for the duration of treatment exposure.

Results

In HCC patients, incidence of any-grade irLI was 11.4% over a median treatment exposure of 4.4 months (95%CI 3.7-5.2), compared to 2.6% in INVIDIa-2 cohort over a median treatment exposure of 12.4 months (95%CI 11.1-14.0). Exposure-adjusted incidence of any-grade irLI was 22.1 per 100-Patient-years (PY) in HCC patients and 2.1 per 100-PY in non-HCC patients (p<0.001), with median time to irLI of 1.4 in HCC and 4.7 months in non-HCC patients, respectively. Among patients who developed irLI, systemic corticosteroids were administered in 16.3% of HCC and in 75.0% of non-HCC patients (p<0.001) and irLI resolution was observed in 72.1% and 58.3%, respectively (p=0.362). In HCC patients, rates of hepatic decompensation and treatment discontinuation due to irLI were 7%. In both cohorts, no fatal irLI events occurred. Development of grade 1-2 irLI was associated with improved overall survival in HCC patients only (HR 0.53, 95%CI 0.29-0.96).

Conclusions

Despite higher incidence and earlier onset in patients with HCC, IrLI is characterised by high rates of remission, low requirement for corticosteroid therapy and low risk of decompensation compared to other solid tumours. Hepatotoxicity leads to discontinuation in 7% of patients with HCC and does not negatively affect oncological outcomes.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

C. Celsa: Financial Interests, Personal, Speaker, Consultant, Advisor: Eisai, AstraZeneca, MSD. G. Cabibbo: Financial Interests, Personal, Advisory Board: Bayer, Eisai, Ipsen, MSD, AstraZeneca, Roche. T.U. Marron: Financial Interests, Personal, Advisory Board: Rockfeller University, Regeneron, AbbVie, Merck, Bristol Myers Squibb, Boehringer Ingelheim, Atara, AstraZeneca, Genentech, Celldex, Chimeric, DrenBio, GlenMark, Simcere, Surface, G1 Therapeutics, NGMBio, DBV Technologies, Arcus, Astellas; Financial Interests, Personal, Research Grant: Regeneron, Bristo-Myers Squibb, Merck, Boehringer Ingelheim. A. Saeed: Financial Interests, Personal, Research Grant: AstraZeneca, Bristol Myers Squibb, Merck, Clovis, Exelixis, Actuate Therapeutics, Incyte Corporation, Daiichi Sankyo, Five Prime Therapeutics, Amgen, Innovent Biologics, Dragonfly Therapeutics, KAHR Medical, BioNtech; Financial Interests, Personal, Advisory Board: Merck, AstraZeneca, Bristol Myers Squibb, Exelixis, Taiho, Pfizer. M. Pinter: Financial Interests, Personal, Advisory Board: Bayer, Bristol Myers Squibb, Eisai, Ipsen, Lilly, MSD, Roche; Financial Interests, Personal, Non remunerated activity: Bayer, Bristol Myers Squibb. A. Pillai: Financial Interests, Personal, Speaker, Consultant, Advisor: Eisai, Exelixis, Genentech/Roche, AstraZeneca, Replimune. M. Schoenlein: Financial Interests, Personal, Other: Janssen, Ipsen, BMS, Astellas, Pfizer; Financial Interests, Personal, Speaker, Consultant, Advisor: Janssen. J. von Felden: Financial Interests, Personal, Advisory Board: Roche. P.R. Galle: Financial Interests, Personal, Advisory Board: Adaptimmune, AstraZeneca, Bayer, Bristol Myers Squibb, Eisai, Ipsen, Lilly, Merck Sharp and Dome, Roche, Sirtex; Financial Interests, Personal, Research Funding: Bayer, Roche; Financial Interests, Personal, Expert Testimony: Lilly; Financial Interests, Personal, Other: AstraZeneca, Bayer, Bristol Myers Squibb, Eisai, Ipsen, Lilly, Roche. M. Kudo: Financial Interests, Personal, Invited Speaker: Eisai, Chugai, Eli Lilly, Bayer, Takeda, AstraZeneca; Financial Interests, Institutional, Research Grant: Otsuka, EA Pharma, Taiho, Eisai, AbbVie, GE Healthcare, Chugai. L. Rimassa: Financial Interests, Personal, Advisory Board, Consulting and advisory role: AstraZeneca, Basilea, Bayer, Exelixis, Genenta, Hengrui, Incyte, Ipsen, IQVIA, Jazz Pharmaceuticals, Elevar Therapeutics, MSD, Nerviano Medical Sciences, Roche, Servier, Taiho Oncology, Zymeworks; Financial Interests, Personal, Invited Speaker, Lecture fees: AstraZeneca, Bayer, BMS, Incyte, Ipsen, Roche, Servier; Financial Interests, Personal, Other, Travel expenses: AstraZeneca; Financial Interests, Institutional, Steering Committee Member: Exelixis, Incyte, Ipsen, Nerviano Medical Sciences, Roche; Financial Interests, Institutional, Coordinating PI, National (Italian) coordinating PI: AstraZeneca, BeiGene, Zymeworks; Financial Interests, Institutional, Local PI: Agios, Eisai, Fibrogen, Lilly, MSD; Financial Interests, Institutional, Funding: Ipsen; Financial Interests, Institutional, Coordinating PI, European PI: AstraZeneca; Non-Financial Interests, Personal, Leadership Role, Treasurer: ILCA; Non-Financial Interests, Personal, Leadership Role, Co-chair: EORTC GITCG HB/NET Task Force; Non-Financial Interests, Personal, Other, Special Expert Clinical Trials Europe: NCI HB Task Force. A. Singal: Financial Interests, Personal, Advisory Board: Genentech, AstraZeneca, Eisai, Bayer, Exelixis, BMS, Roche, Glycotest, Exact Sciences, FujiFilm Medical Sciences, GRAIL. H. Chon: Financial Interests, Personal, Advisory Board: Eisai, Roche, Bayer, ONO, MSD, BMS, Celgene, Sanofi, Servier, AstraZeneca, Sillajen, Menarini, GreenCross Cell. M. Bersanelli: Financial Interests, Personal, Advisory Board: Pfizer, Novartis, Ipsen; Financial Interests, Personal, Other, Case report with copyright transfert: Pierre Fabre; Financial Interests, Institutional, Other, Research funding: Roche, Seqirus, Pfizer, Novartis; Financial Interests, Personal, Writing Engagement: Pierre Fabre, Sciclone Pharmaceuticals; Financial Interests, Personal, Invited Speaker: MSD, Novartis; Financial Interests, Institutional, Local PI: Pfizer; Non-Financial Interests, Personal, Product Samples, congresso grant and accomodation: Pierre Fabre. A. Cortellini: Financial Interests, Personal, Advisory Board: AstraZeneca, MSD, OncoC4, Ardelis Health, Access Infinity, AlphaSight; Financial Interests, Personal, Invited Speaker: AstraZeneca, Eisai, Pierre Fabre; Financial Interests, Personal, Writing Engagement: MSD, 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
Poster Display (ID 34) Poster Display

112P - Close cardiovascular monitoring during the early stages of treatment for patients receiving immune checkpoint inhibitors

Presentation Number
112P
Lecture Time
20:30 - 20:30
Speakers
  • Danielle L. Delombaerde (Gent, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immune checkpoint inhibitors (ICIs) can lead to a wide range of cardiovascular (CV) adverse events. As the population of cancer patients receiving ICIs is rapidly growing, there is a high need for both short- and long- term monitoring tools to promptly detect these side effects. Therefore, we developed a prospective, multicenter trial (NCT05699915) in which we assess different cardiac biomarkers as well as routine three dimensional (3D) echocardiography in cancer patients with a solid tumor treated with ICIs.

Methods

All patients underwent routine investigations of blood parameters (high-sensitivity troponin I and T (hs-TnI and hs-TnT), N-terminal pro-brain natriuretic peptide levels (NT-proBNP), in particular) and a thorough CV follow-up (electrocardiography and echocardiography) at fixed time points. The primary endpoint was defined as the incidence of an elevated hs-TnT above the upper limit of normal (ULN) if the baseline value was normal; or 1.5 ≥ times baseline if the baseline value was above the ULN, during the first three months of treatment. Here, we present the results of a preliminary analysis of the first cohort of 59 patients after three months of follow-up.

Results

The mean patient age was 68 ± 12 years; 76% were men. 56% of the patients received an anti-PD-1 agent, while 19% received anti-PD-L1. Overall, 25% of patients received combination therapy. Troponin elevation, as defined in the primary endpoint, occurred in 10.6% [95% CI: 5.0-22.5]. No patients, however, experienced a clinically significant CV event. Furthermore, there were no changes in 3D left ventricular (LV) ejection fraction or LV global longitudinal strain following three months of ICI treatment (55.95 ± 5.95% vs. 56.05 ± 6.46%, p = 0.903 and -17.8% [-18.5 ; -14.2] vs. -17.0% [-18.8 ; -15.1], p = 0.663). No significant changes in diastolic function or right ventricular function were found either. In addition, there was poor agreement between hs-TnT and hs-TnI.

Conclusions

Our preliminary findings found hs-TnT elevation in 10% of cancer patients during the first three months of ICI therapy. However, no significant changes were noted on 3D echocardiography at three months. Hence, longer follow-up and further research are needed.

Clinical trial identification

NCT05699915.

Legal entity responsible for the study

University of Antwerp.

Funding

Has not received any funding.

Disclosure

C. Vulsteke: Financial Interests, Personal, Advisory Board: MSD, Janssen-Cilag, GSK, Astellas Pharma, BMS, Leo Pharma, Bayer, AstraZeneca, Pfizer, Merck; Financial Interests, Institutional, Research Grant, Funding for research project on immune related toxicities: MSD. N. Van de Veire: Financial Interests, Personal, Advisory Role: Pfizer; Financial Interests, Personal, Speaker’s Bureau: Pfizer, Menarini, Daiichi Sankyo Pharmaceutical; Financial Interests, Personal, Other, Travel, Accomodations, Expenses: Boehringher Ingelheim. D. Vervloet: Financial Interests, Personal, Advisory Role: Novartis, Pfizer, AstraZeneca Co.; Financial Interests, Personal, Speaker’s Bureau: Novartis, Pfizer, Izidok, AstraZeneca Co.; Financial Interests, Personal, Other, Travel, Accomodations, Expenses: Pfizer, Novartis. V. Moerman: Financial Interests, Personal, Other, Honoraria: Menarini; Financial Interests, Personal, Other, Travel, Accomodations, Expenses: Menarini, Daiichi Sankyo Pharmaceutical, Bayer US, LLC. L. van Calster: Financial Interests, Personal, Other, Employment: Maatschap Cardiologie. M. Peeters: Financial Interests, Personal, Advisory Role: Amgen, Bayer, Ipsen, Remedus, Sanofi, Sirtex Medical, Terumo, Merck, MSD, Qurin; Financial Interests, Personal, Leadership Role: Qurin; Financial Interests, Personal, Speaker’s Bureau: Amgen, Bayer US, Llc, Merck Serono, Roche, Sanofi, Servier, Sirtex Medical; Financial Interests, Personal, Stocks or ownership: BIMINI BioTech; Financial Interests, Personal, Other, Honoraria: Amgen, Bayer US, Llc, Sanofi, Servier, Merck, Bristol Myers Squibb, MSD, Roche, Sirtex Medical; Financial Interests, Personal, Funding: Amgen, Bayer US, Llc, Ipsen, Novartis, Roche. H. Prenen: Financial Interests, Institutional, Advisory Board: Amgen, Roche, AstraZeneca; Financial Interests, Institutional, Invited Speaker: Bayer, Ipsen, Sanofi. J. De Sutter: Financial Interests, Personal, Advisory Role: Novartis, Bayer US, Llc. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

113P - A multidisciplinary management of immune-checkpoint inhibitor (ICI)-related pneumonitis to improve its clinical management

Presentation Number
113P
Lecture Time
20:30 - 20:30
Speakers
  • Monica Valente (Siena, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Treatment with ICI can associate with a wide spectrum of immune-related adverse events (irAEs). Among irAEs is immune-mediated pneumonitis (im-PN), a rare but potentially life-threatening side effect; thus, prompt diagnosis and effective management of im-PN is essential to avoid severe complications.

Methods

We collected a case series of skin cancer (melanoma-MM, squamous cell carcinoma-SCC), lung cancer (LC) and mesothelioma (MESO) patients (pts), treated with ICI at the Center for Immuno-Oncology of the University Hospital of Siena, Italy, diagnosed with im-PN. Clinical and radiologic data were thoroughly collected, as well as bronchoalveolar lavage (BAL) samples; im-PN were graded using CTCAE v. 5.0.

Results

From Jan 2014 to Feb 2023, 1004 pts with MM (n=522), SCC (n=42), LC (n=342) or MESO (n=98) were treated with ICI (619 anti-PD-1, 385 combinations). Among treated pts 24 (2%) developed an im-PN, and 14 (58%) were symptomatic. Im-PN was classified grade (G)1 (10 pts), G2 (13 pts), and G5 (1 patient). Steroid treatment was promptly activated leading to complete resolution of im-PN in 22 pts. Thirteen pts resumed ICI therapy once fully-recovered from im-PN, and 2 of them experienced im-PN recurrence that completely resolved with steroids re-treatment. According to the Fleischner Society classification of drug-related pneumonitis, 3 main radiologic patterns were identified: organizational pneumonia (OP)-like (16,6%), pulmonary eosinophilia (PEo) (7,3%), and hypersensitivity pneumonitis (HP) (1,4%). Furthermore, BAL samples analysis performed in 8 (33%) symptomatic pts showed an inflammatory lymphocytic infiltrate, predominantly consisting in a foam cell-like macrophages infiltrate in 6 cases. Notably, Transmission Electron Microscopy evaluation performed in 2 of these pts, revealed multilamellar bodies, lysosomes, and lipid vacuoles into the alveolar macrophages, a scenario suggestives for a drug-mediated toxicity.

Conclusions

Im-PN associated with ICI therapy was found to be a rare and challenging side effect, with variable time onset, and heterogenous clinical presentation. A multidisciplinary characterization of im-PN helps optimizing its clinical management to resume ICI therapy.

Legal entity responsible for the study

Anna Maria Di Giacomo.

Funding

Has not received any funding.

Disclosure

M. Valente: Financial Interests, Personal, Advisory Board: Novartis. M. Maio: Financial Interests, Personal, Advisory Board: Roche, Bristol Myers Squibb, Merck Sharp Dohme, Incyte, AstraZeneca, Amgen, Pierre Fabre, Eli Lilly, GSK, Sciclone, Sanofi, Alfasigma, Merck Serono; Financial Interests, Personal, Ownership Interest: Theravance, Epigen Therapeutics Srl. L. Calabro: Financial Interests, Personal, Advisory Board: Roche, Bristol Myers Squibb, Merck Sharp Dohme; Financial Interests, Personal, Other, Educational Activities: Bristol Myers Squibb, Sanofi, AstraZeneca. A.M. Di Giacomo: Financial Interests, Personal, Advisory Board: Incyte, Pierre Fabre, GSK, Bristol Myers Squibb, Merck Sharp Dohme, Sanofi; Financial Interests, Personal, Other, Educational activities: Bristol Myers Squibb, Merck Sharp Dohme, Pierre Fabre, Sanofi. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

114P - Real-World Insights on Pan-Cancer Immune Checkpoint Inhibitor Treatment: Initial Findings of a Belgian Multicenter Study

Presentation Number
114P
Lecture Time
20:30 - 20:30
Speakers
  • Annelies T. Verbiest (Edegem, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

To bridge the gap between clinical trial patients and real-world populations, we conducted a comprehensive study in Belgium to characterize cancer patients treated with immune checkpoint inhibitors (ICIs), which have demonstrated survival advantages in various cancer types.

Methods

Retrospective multicenter study in three Belgian hospitals, processing anonymized electronic health records including 10 data sources, using natural language processing (NLP) and machine learning. NLP model validation compared algorithm outputs to a physician-generated standard. The algorithm mapped 597 variables to SNOMED-CT, generating OMOP CDM databases, validated per hospital (federated), ensuring patient privacy. Cancer patients (≥18 years old, regardless of stage) receiving ICIs between March 2017 and August 2022 were included.

Results

Our study included 1,659 patients (median age [IQR]: 67 [60-74]); age categories: 2.4% (18-40 years), 6.1% (41-50), 17.8% (51-60), 35.1% (61-70), and 38.6% (>71). Most patients were male (65.9%). Current/former smokers totaled 43.2%, 18.6% were never smokers, and 38.2% had unknown smoking status. Alcohol abuse was observed in 10.8% of patients and negative/unknown in 89.2%. The most common ICI treatment (monotherapy or with other ICIs or antineoplastic drugs) was pembrolizumab (43.6%), followed by nivolumab (29.7%), atezolizumab (10.7%), ipilimumab (8.7%), durvalumab (4.2%), avelumab (2.1%), cemiplimab (0.8%) and dostarlimab (0.1%). Lung cancer was the most frequent cancer type (54.94%), followed by renal cancer (9.23%), bladder cancer (7.27%), melanoma (6.08%), and head and neck cancer (5.97%). Median overall survival by ICI ranged from 8 to 38 months.

Conclusions

These initial findings highlight the feasibility of automatically extracting and locally validating federated hospital databases- an invaluable tool for real-world data on ICI-treated patients. Comparable datasets would require linking government databases, which cannot be done in a federated manner nor enriched with hospital-level data. Ongoing analyses will explore immune-related adverse events, comorbidities, tumor stage, anatomical pathology, and outcomes in various cancer types and treatment lines.

Legal entity responsible for the study

LynxCare Clinical Informatics NV.

Funding

LynxCare Clinical Informatics NV.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

115TiP - MDT-BRIDGE: A phase 2 study of neoadjuvant durvalumab (D) + chemotherapy (CT) followed by either surgery and adjuvant D or chemoradiotherapy (CRT) and consolidation D in patients (pts) with resectable or borderline resectable stage IIB-IIIB NSCLC

Presentation Number
115TiP
Lecture Time
20:30 - 20:30
Speakers
  • Martin Reck (Grosshansdorf, Germany)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

In the phase 3 AEGEAN study, neoadjuvant D + platinum-based CT followed by adjuvant D, versus neoadjuvant CT alone, significantly improved pathological complete response (pCR) and event-free survival (EFS) in pts with resectable NSCLC. In the placebo-controlled phase 3 PACIFIC study, consolidation D significantly improved outcomes for pts with unresectable stage III NSCLC after CRT. The strong pathological and clinical outcomes seen with CT + immunotherapy (IO) have generated interest in its use to enable pts with borderline resectable NSCLC to undergo surgery (Sx). In parallel, for pts who are initially deemed resectable but later become unresectable/inoperable during neoadjuvant treatment (Tx), the safety and efficacy of initial neoadjuvant IO + CT, followed by CRT and consolidation IO should be explored. To inform clinical practice, MDT-BRIDGE (NCT05925530) assesses the efficacy and safety of neoadjuvant D + CT followed by either Sx and adjuvant D or CRT and consolidation D in pts with resectable/borderline resectable Stage IIB–IIIB NSCLC.

Trial design

This is a global, phase 2, non-randomized study of pts aged ≥18 yr with ECOG PS 0/1 and histologically/cytologically confirmed, Tx-naïve, EGFR/ALK wild-type, Stage IIB to select IIIB [N2] NSCLC (AJCC 8th ed). Initially deemed resectable/borderline resectable by a multidisciplinary team (MDT), ∼140 pts will receive 2 cycles of neoadjuvant D + investigator’s choice platinum-based CT Q3W IV, followed by MDT assessment of resectability. Pts deemed resectable will receive a further 1–2 cycles of neoadjuvant D + CT followed by Sx (Cohort 1); pts deemed unresectable will receive CRT for ∼6 wk (Cohort 2). Pts in Cohort 1 who become ineligible for Sx can enter Cohort 2. After Sx/CRT, all pts will receive D Q4W IV for up to 1 yr. The primary endpoint is the resection rate in all pts. Secondary endpoints include resection rates in the subsets of pts deemed resectable or borderline resectable at baseline; R0, R1, and R2 resection outcomes; pCR; EFS; progression-free survival; overall survival; objective response rate; ctDNA clearance; and safety.

Clinical trial identification

NCT05925530 (Release date June 29, 2023).

Editorial acknowledgement

Medical writing support for this abstract, under the direction of the authors, was provided by Werner Gerber, PhD, (Fourways, South Africa) and Aaron Korpal, PhD, (Macclesfield, UK) of Ashfield MedComms, an Inizio Company, and was funded by AstraZeneca.

Legal entity responsible for the study

AstraZeneca, Sweden.

Funding

AstraZeneca.

Disclosure

M. Reck: Financial Interests, Personal, Invited Speaker: Amgen, AstraZeneca, BMS, Boehringer Ingelheim, Daiichi Sankyo, GSK, Lilly, MSD, Merck, Mirati, Novartis, Pfizer, Roche, Regeneron, Sanofi; Financial Interests, Personal, Speaker’s Bureau: Amgen, AstraZeneca, BMS, Boehringer Ingelheim, Daiichi Sankyo, GSK, Lilly, MSD, Merck, Mirati, Novartis, Pfizer, Roche, Regeneron, Sanofi; Financial Interests, Personal, Advisory Board: Amgen, AstraZeneca, BMS, Boehringer Ingelheim, Daiichi Sankyo, GSK, Lilly, MSD, Merck, Mirati, Novartis, Pfizer, Roche, Regeneron, Sanofi; Non-Financial Interests, Personal and Institutional, Officer: ESMO; Non-Financial Interests, Institutional, Research Grant: BMS, Boehringer Ingelheim; Non-Financial Interests, Institutional, Principal Investigator: Amgen, AstraZeneca, BMS, Boehringer Ingelheim, Daiichi Sankyo, BeiGene, GSK, Lilly, MSD, Merck, Mirati, Novartis, Roche. E. Nadal: Financial Interests, Personal, Invited Speaker: Roche, Bristol Myers Squibb, Merck Sharp & Dohme, Merck Serono, Sanofi, Pfizer, Lilly, Amgen, Janssen, Daiichi Sankyo, Boehringer Ingelheim, AstraZeneca, Qiagen, Pierre Fabre, Takeda; Financial Interests, Personal, Advisory Board: Roche, Bristol Myers Squibb, Merck Sharp & Dohme, Sanofi, Pfizer, Lilly, Amgen, Janssen, Daiichi Sankyo, Boehringer Ingelheim, AstraZeneca, Qiagen, Pierre Fabre, Takeda, Bayer; Non-Financial Interests, Personal, Funding: Roche, Pfizer, Merck Serono, Bristol Myers Squibb. N. Girard: Financial Interests, Personal, Invited Speaker: AstraZeneca, BMS, MSD, Roche, Pfizer, Mirati, Amgen, Novartis, Sanofi, Gilead; Financial Interests, Personal, Advisory Board: AstraZeneca, BMS, MSD, Roche, Pfizer, Janssen, Boehringer, Novartis, Sanofi, AbbVie, Amgen, Lilly, Grunenthal, Takeda, Owkin, Leo Pharma, Daiichi Sankyo, Ipsen; Financial Interests, Institutional, Research Grant, Local: Roche, Sivan, Janssen; Financial Interests, Institutional, Funding: BMS, Leo Pharma; Financial Interests, Institutional, Research Grant: MSD; Non-Financial Interests, Personal, Officer, International Thymic malignancy interest group, president: ITMIG; Other, Personal, Other, Family member is an employee: AstraZeneca. A.R.R. Filippi: Financial Interests, Personal, Invited Speaker: AstraZeneca, Roche; Financial Interests, Personal, Speaker’s Bureau: AstraZeneca; Financial Interests, Personal, Advisory Board: AstraZeneca; Financial Interests, Personal, Research Grant: AstraZeneca; Financial Interests, Personal, Principal Investigator: AstraZeneca; Financial Interests, Personal, Advisory Role: Radiomics; Financial Interests, Personal, Funding: AstraZeneca, MSD, Roche. L.W. Martin: Financial Interests, Personal, Speaker’s Bureau: Genentech, Ethicon; Financial Interests, Personal, Advisory Board: AstraZeneca, Genentech; Non-Financial Interests, Personal, Officer: General Thoracic Surgical Club; Financial Interests, Personal, Full or part-time Employment: University of Virginia School of Medicine; Non-Financial Interests, Personal, Principal Investigator: Alliance Foundation Trial 46; Financial Interests, Personal, Advisory Role: OnTarget Laboratories; Non-Financial Interests, Personal, Leadership Role: Alliance for Clinical Trials. C.M. Gay: Financial Interests, Personal, Invited Speaker: Aptitude Health, Dava Oncology, MJH Healthcare, OncLive, Peer View Institute; Financial Interests, Personal, Speaker’s Bureau: AstraZeneca, BeiGene; Financial Interests, Personal, Advisory Board: AstraZeneca, BMS, Daiichi Sankyo, Jazz, MonteRosa; Financial Interests, Personal, Research Grant: AstraZeneca; Financial Interests, Personal, Advisory Role: AstraZeneca, Catalyst, Roche/Genentech, STCube. C. Petersen: Non-Financial Interests, Personal, Invited Speaker: Brainlab, Siemens/Varian, AstraZeneca, Novocure; Non-Financial Interests, Personal, Research Grant: Siemens; Financial Interests, Personal, Principal Investigator: MSD, Merck ; Non-Financial Interests, Personal, Training: Brainlab. D. Gale: Financial Interests, Personal, Full or part-time Employment: AstraZeneca; Financial Interests, Personal, Stocks/Shares: AstraZeneca, GSK; Financial Interests, Personal, Other: Co-founder of Inivata (acquired by NeoGenomics in 2021). H.R. Bridge, N. Donner, N.E. Georgoulia : Financial Interests, Personal, Full or part-time Employment: AstraZeneca; Financial Interests, Personal, Stocks/Shares: AstraZeneca. J. Spicer: Financial Interests, Personal, Other, Honoraria: Amgen, AstraZeneca, Bristol Myers Squibb/Medarex, Chemocentryx, Merck, Novartis; Financial Interests, Personal, Advisory Role: AstraZeneca, Bristol Myers Squibb/Medarex, Merck, Protalix Biotherapeutics, Regeneron, Xenetic Biosciences; Financial Interests, Institutional, Research Funding: AstraZeneca, Bristol Myers Squibb/Medarex, CLS-Therapeutics, CLS-Therapeutics, Merck, Protalix Biotherapeutics, Roche; Financial Interests, Personal, Other, Travel and Accommodations: AstraZeneca, Bristol Myers Squibb/Medarex, Merck.

Collapse
Poster Display (ID 34) Poster Display

117TiP - BGB-HNSCC-201 (NCT05909904): Phase 2, Open-Label, Multi-Arm, Global Study of Tislelizumab (TIS) + Investigational Agents as First-Line (1L) Treatment in Patients (Pts) With Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma (R/M HNSCC)

Presentation Number
117TiP
Lecture Time
20:30 - 00:45
Speakers
  • Kevin J. Harrington (London, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

HNSCC is the 7th most common cancer worldwide (Sung et al, CA Cancer J Clin 2021;71:209–249). Anti-programmed cell death-protein 1 (PD-1) therapy, either alone or in combination with chemotherapy (CT), extended overall survival (OS) vs cetuximab + CT in pts with programmed cell death-ligand 1 (PD-L1)-positive R/M HNSCC in the 1L setting; however, not all pts respond to single-agent anti-PD-1 therapy. Identifying novel agents that synergize with anti-PD-1 therapies by targeting distinct biological pathways may improve efficacy. TIS is an anti-PD-1 monoclonal antibody (mAb) currently approved in China for multiple indications. This Phase 2 study will assess efficacy and safety of TIS in combination with investigational agents targeting the immune-checkpoint inhibitors TIM-3 (BGB-A425) and/or LAG-3 (LBL-007) as 1L treatment in pts with R/M HNSCC.

Trial design

This multicenter (77 sites; 14 countries) study will enroll approximately 160 pts (40 per arm) aged ≥18 years with immunotherapy-naïve, PD-L1 positive (combined positive score [CPS] ≥1) R/M HNSCC of the oropharynx, oral cavity, hypopharynx, or larynx, who are not candidates for local/curative therapy and have ≥1 measurable lesion (per RECIST v 1.1). Pts will be randomized 1:1:1:1 (stratified by PD-L1 CPS: 1–19 vs ≥20) to TIS monotherapy, TIS + BGB-A425, TIS + LBL-007, or TIS + BGB-A425 + LBL-007. TIS 200 mg, BGB-A425, and LBL-007will be administered by separate intravenous infusions once every 3 weeks for up to 2 years, until disease progression, intolerable toxicity, withdrawal of informed consent, or other discontinuation event, whichever occurs first. The primary endpoint is confirmed objective response rate (per investigator; RECIST v1.1). Secondary endpoints include progression-free survival, duration of response, clinical benefit rate, and disease control rate (all per investigator; RECIST v1.1) as well as safety, OS, and immunogenicity to study drugs. Enrollment is ongoing and additional experimental arms may be added in the future.

Editorial acknowledgement

Medical writing support, under the direction of the authors, was provided by Smitha Reddy, PhD, of Envision Pharma Group, and was funded by BeiGene.

Legal entity responsible for the study

BeiGene, Ltd.

Funding

BeiGene, Ltd.

Disclosure

K.J. Harrington: Financial Interests, Institutional, Advisory Board: Amgen, AstraZeneca, BMS, Boehringer Ingelheim, Merck, MSD, Pfizer, Replimune, Oncolys, Vyriad, Idera; Financial Interests, Institutional, Other, Honoraria for lectures: Amgen, AstraZeneca, BMS, Boehringer Ingelheim; Financial Interests, Institutional, Advisory Board, Advisory board for CD47 assets: Arch Oncology; Financial Interests, Institutional, Advisory Board, Development of DDR assets: ARTIOS; Financial Interests, Institutional, Advisory Board, Development of exosomal STING agonist: Codiak; Financial Interests, Institutional, Advisory Board, Development of oncolytic adenovirus: PsiVac; Financial Interests, Institutional, Funding, Research: AstraZeneca, Boehringer Ingelheim, MSD; Financial Interests, Institutional, Funding, Development of oncolytic HSV platform: Replimune; Non-Financial Interests, Personal, Leadership Role, Chair of Steering Committee: ART NET; Non-Financial Interests, Personal, Other, Member of Global Steering Committee: MR - Linac. Y. Guo: Financial Interests, Personal, Invited Speaker: Merck Serono, Roche, MSD, BMS, BeiGene. R. Haddad: Financial Interests, Personal, Advisory Board: BMS, Merck, EMD Serono, Boehringer Ingelheim, Eisai, Bayer, Merus, AstraZeneca, Genentech, Pfizer, Genmab, Exelexis, Coherus; Financial Interests, Personal, Other, Data Safety Monitoring Board ( DSMB): Nanobiotix, PSI, Hookipa; Financial Interests, Personal, Royalties: up to date; Financial Interests, Institutional, Local PI, Grant to Hospital For Clinical Trials: Merck BMS, EMD , AstraZeneca, Incyte, Kura; Non-Financial Interests, Personal, Other, panel chair guidelines: Nccn. H.R. Kim: Financial Interests, Personal, Speaker, Consultant, Advisor, Payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events: AstraZeneca, Bristol Myers Squibb, Genentech/Roche, MSD. C.A. Perez: Financial Interests, Personal, Advisory Board: EMD Serono; Financial Interests, Personal, Other, Steering Committee: Kinnate Biopharma Inc.; Financial Interests, Institutional, Local PI: Accutar Biotech, Kinnate Biopharma, Relay Therapeutics, Seagen Inc, Kura Oncology, Hyamab Inc, Xilio Therapeutics, Elucida Oncology, Tallac Therapeutics, Ribbon Therapeutics, Mirati Therpeutics, Elpiscience Biopharmaceuticals, Dracen Pharmaceuticals, Zhuhai Yufan Biotechnologies Co., Genentech, Inc., Jazz Pharmaceuticals, Artios Pharma, Ayala Pharmaceuticals, Elevation Oncology. I. Xiang, H. Li, G. Dong: Financial Interests, Personal, Full or part-time Employment: BeiGene. C. Even: Financial Interests, Personal, Advisory Board: BMS, MSD, Innate Pharma, Merck Serono; Financial Interests, Institutional, Advisory Board: F Star Therapeutics, Novartis, Elevar; Financial Interests, Institutional, Local PI: BMS, AstraZeneca, ISA Pharmaceutics, MSD, Debiopharma, Ayala, Gilead; Financial Interests, Institutional, Coordinating PI: BMS, Novartis, Sanofi. L.D. Locati: Financial Interests, Personal, Funding, Consulting Fees: Merck, MSD, Eisai, Bayer, Roche, Ipsen, Sanofi; Sunpharma; New Bridge; Gentili SrL; Financial Interests, Personal, Funding, Payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events: Lilly; MSD; Esiai; Financial Interests, Personal, Funding, Support for attending meetings and/or travel: Lilly; MSD; Financial Interests, Personal, Funding, Grants or contracts: MSD; Financial Interests, Personal, Advisory Board: Lilly, Bayer. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

121P - MK-7684A (Vibostolimab [Vibo] Plus Pembrolizumab [Pembro] Coformulation) With/Without Docetaxel in Metastatic NSCLC After Platinum-Chemotherapy (Chemo) and Immunotherapy

Presentation Number
121P
Lecture Time
00:45 - 00:45
Speakers
  • Nir Peled (Jerusalem, Israel)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Patients (pts) with metastatic non–small-cell lung cancer (mNSCLC) who progress on anti–PD-(L)1 therapy in combination with or after chemo often have poor prognosis resulting in high unmet need. The anti–T-cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT) antibody vibo has shown antitumor activity alone and in combination with pembro (anti–PD-1). The phase 2 KEYVIBE-002 study (NCT04725188) evaluated MK-7684A, a coformulation of vibo + pembro, with/without docetaxel vs docetaxel alone for previously treated mNSCLC.

Methods

Eligible pts aged ≥18 y had pathologically confirmed mNSCLC without EGFR/ALK/ROS1 alterations, PD after 1 prior anti–PD-(L)1 therapy (PD ≤12 wks from last dose) and platinum-doublet chemo, measurable disease per RECIST v1.1, ECOG PS 0 or 1, and tumor sample for PD-L1 assessment. Pts were randomized 1:1:1 to MK-7684A (vibo 200 mg + pembro 200 mg) + docetaxel 75 mg/m2 Q3W (Arm 1; blinded), MK-7684A (Arm 2; open label), or placebo + docetaxel (Arm 3; blinded). Treatment continued for ≤35 cycles (∼2 y) for MK-7684A/placebo, per local guidelines for docetaxel, or until PD, unacceptable AEs, or pt/physician decision. Primary endpoint was PFS per RECIST v1.1 by BICR. Safety was a secondary endpoint.

Results

87 pts were randomized to Arm 1, 83 to Arm 2, and 85 to Arm 3. Median follow-up at data cutoff (Jan 26, 2023) was 12.3 (range, 6.2–19.8) mo. Median PFS (95% CI) was 5.6 (3.9–6.8) mo in Arm 1, 2.7 (1.8–4.0) mo in Arm 2, and 3.2 (2.8–5.7) mo in Arm 3. HR (95% CI) for PFS for Arm 1 vs Arm 3 was 0.77 (0.53–1.13), P = 0.0910; and for Arm 2 vs Arm 3 was 1.40 (0.96–2.02), P = 0.9622. Treatment-related AEs occurred in 82 pts (96.5%) in Arm 1, 50 (60.2%) in Arm 2, and 74 (89.2%) in Arm 3; these were fatal in 4 (4.7%), 1 (1.2%), and 1 (1.2%), respectively. Immune-mediated AEs and infusion reactions occurred in 25 pts (29.4%), 17 (20.5%), and 10 (12.0%), respectively. Additional data will be presented.

Conclusions

In pts with mNSCLC previously treated with anti-PD-(L)1 therapy and platinum-doublet chemo, MK-7684A and MK-7684A + docetaxel vs docetaxel alone did not show statistically significant differences in PFS, and no new safety signals. Follow-up is ongoing.

Clinical trial identification

NCT04725188.

Editorial acknowledgement

Medical writing assistance was provided by Kathleen Estes, PhD, of ICON plc (Blue Bell, PA, USA).

Legal entity responsible for the study

Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA.

Funding

Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA.

Disclosure

N. Peled: Financial Interests, Personal, Other, Advisor, honorarium, Research: AstraZeneca, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly, Foundation Medicine, Guardant360, Imagene, Merck KGaA, MSD, Novartis, Novocure, Pfizer, Roche, Renium, Takeda. J. Mazieres: Financial Interests, Personal, Other, Personal fees: Roche, AstraZeneca, Pierre Fabre, Takeda, BMS, MSD, Pfizer, Jiangsu, Hengruii, Blueprint, Daiichi, Novartis, Amgen; Financial Interests, Personal, Research Grant: Roche, AstraZeneca, Pierre Fabre, BMS. D.M. Kowalski: Financial Interests, Personal, Other, Advisory Board & Honorarium: Roche, Takeda, AstraZeneca, MSD, BMS, Merck KGaA, Pfizer, Amgen, Johnson & Johnson, Sanofi-Aventis, Boehringer Ingelheim, Novartis. W. Lam: Financial Interests, Personal, Other, Honorarium & Education Support: BMS, MSD, Roche, Prifer, Novartis, AstraZeneca, Sanofi, Merck Healthcare Pty. Ltd. M.J. Hochmair: Financial Interests, Personal, Other, Lectures and advisory boards: MSD, Roche, Lilly, AstraZeneca, Takeda. M. Majem: Financial Interests, Personal, Advisory Board: Amgen, Roche, AstraZeneca, Takeda, Janssen, Cassen Recordati, BMS; Financial Interests, Personal, Invited Speaker: Amgen, Roche, AstraZeneca, Pfizer, Takeda, Helsinn; Financial Interests, Institutional, Funding: BMS, AstraZeneca, Roche. A. Calles Blanco: Financial Interests, Personal, Advisory Board: AstraZeneca, Boehringer Ingelheim, Pfizer, Roche, Lilly, Merck Sharp & Dohme, Novartis, Bristol Myers Squibb, Takeda, Sanofi, Janssen; Financial Interests, Personal, Other, Speaker honoraria: Bayer; Financial Interests, Institutional, Research Grant, Drug-only for Investigator-initiated trial: Merck Sharp & Dohme. K. Cuppens: Financial Interests, Personal, Advisory Board: F. Hoffmann-La Roche, AstraZeneca, Merck Sharp Dohme, Merck Sharp Dohme, AstraZeneca, Bristol Myers Squibb, Boehringer Ingelheim, Bayer; Financial Interests, Personal, Invited Speaker: Pfizer, Bristol Myers Squibb, Merck Sharp & Dohme, F. Hoffmann-La Roche. I.A. Casarini: Financial Interests, Institutional, Research Funding: Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA. O. Bylicki: Financial Interests, Personal, Advisory Board, Expert Board: BMS, Roche, Takeda; Financial Interests, Personal, Advisory Board, Annuel contrat: MSD; Financial Interests, Personal, Advisory Board, expert board: AstraZeneca, Janssen. B. Shim: Financial Interests, Personal, Advisory Role: Gadent, Takeda, Roche, Bion, Chogendang, J INST Bio; Financial Interests, Personal, Research Funding: Yohan. J.B. Sørensen: Financial Interests, Personal, Other, Advisor and Honorarium: MSD, BMS, AstraZeneca, Roche, Sanofi. M. Gilli: Financial Interests, Institutional, Research Funding: Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA. S.H. How: Financial Interests, Personal, Other, Advisor & Honorarium & Research grant: AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, MSD, Novartis, Roche, Takeda. C. Schumann: Financial Interests, Personal, Other, Honoraria for Advisory and Research: AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly, MSD, Novartis, Pfizer, Roche, Takeda. C. Gonzalez Arenas: Financial Interests, Personal, Full or part-time Employment: MSD, Spain; Financial Interests, Personal, Stocks/Shares: Merck & Co., Inc., Rahway, NJ, USA. T. He: Financial Interests, Personal, Full or part-time Employment: Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA. A. Samkari: Financial Interests, Personal, Full or part-time Employment: Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA; Financial Interests, Personal, Stocks/Shares: Merck & Co., Inc., Rahway, NJ, USA. S. Novello: Financial Interests, Personal, Other, Advisor/Speaker Bureau: AZ, AMG, MSD, Eli Lilly, Sanofi, Roche, Takeda, Pfizer, Thermofisher, Janssen. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

122P - Casdozokitug (casdozo, SRF388), a first-in-class IL-27 targeting antibody, as monotherapy (monotx) or in combination with pembrolizumab (pembro) in treatment-refractory non-small cell lung cancer (NSCLC)

Presentation Number
122P
Lecture Time
00:45 - 00:45
Speakers
  • Thomas U. Marron (New York, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Casdozo neutralizes immunoregulatory IL-27 to stimulate antitumor immunity. In preclinical studies, casdozo + anti-PD-1 enhanced immune activation and inflammatory cytokine production. A Ph 1 dose escalation study of casdozo in advanced solid tumors demonstrated favorable safety and reversal of IL-27-mediated immune suppression as evidenced by increases in serum IFN-g and NK cell gene activation. We present antitumor activity and safety of casdozo as monotx and with pembro in NSCLC patients (pts).

Methods

A Ph 1 dose escalation study triggered Ph 2 expansion cohorts exploring casdozo 10 mg/kg IV q4 wks as monotx and q3 wks in combination with pembro in treatment-refractory NSCLC. Tumor response was assessed by RECIST1.1.

Results

As of July 12, 2023, 51 pts received casdozo as monotx (5 dose escalation; 40 expansion) or with pembro (n=6). Most had adenocarcinoma (73%) or squamous (25%) histology. The majority received casdozo as 3rd line or greater (67%). Treatment-related adverse events (TRAEs) occurred in 44% on monotx. TRAEs were primarily grade 1/2, with fatigue being most common (9%, n=4). Of the 41 response-evaluable monotx pts, 2 pts with primary resistance to aPD-(L)1 experienced confirmed, durable partial responses and remained on therapy ≥6 mo. Responders had squamous disease and no or low (10%) archival tumor PD-L1 expression. The monotx ORR in the 7 RECIST-evaluable pts with squamous disease was 29%. IHC of a responder’s archival squamous tumors showed an immune excluded phenotype by CD8 staining and a high density of peritumoral IL-27+ macrophages. In the PD-1 relapsed/refractory combination cohort (n=6), no responses in the 4 response-evaluable pts, high-grade TRAEs or study drug discontinuations have been reported.

Conclusions

The only clinical stage anti-IL-27 targeting antibody, casdozo, is well tolerated with monotx antitumor activity in heavily pretreated, PD-(L)1 experienced NSCLC. Given the antitumor activity, safety, and tolerability of the pembro combination, as well as casdozo’s distinct mechanism, a Ph 2 study will investigate casdozo + toripalimab (aPD-1).

Clinical trial identification

NCT04374877.

Editorial acknowledgement

Editorial assistance was provided by Mark Phillips, PharmD, MBA, with The Phillips Group Oncology Communications, Inc.

Legal entity responsible for the study

Surface Oncology.

Funding

Surface Oncology.

Disclosure

T.U. Marron: Non-Financial Interests, Personal, Advisory Role: Regeneron, BI, AZ, DBV technologies, Celldex, Surface Oncology, NGM Biopharmaceuticals, Glenmark, AbbVie; Non-Financial Interests, Personal and Institutional, Proprietary Information: Surface Oncology; Financial Interests, Personal, Research Funding: Regeneron, BMS, Merck, BI. A. Naing: Non-Financial Interests, Personal, Full or part-time Employment: MDACC; Non-Financial Interests, Personal and Institutional, Advisory Role: CytomX Therapeutics, OncoSec, STCube Pharmaceuticals, Genome and Co, Deka Biosciences, NGM Biopharmaceuticals, PsiOxus Therapeutics, Nouscom, Merck Sharp & Dohme, OncoNano, Servier, Lynx Health, AbbVie, PsiOxus Therapeutics, Takeda, Pharming NV, Horizon T; Other, Personal, Financially compensated role: ARMO Biosciences, NeoImmuneTech, NGM Biopharmaceuticals; Other, Personal, Other: AKH Inc, Lynx Group, Society for Immunotherapy of Cancer, KSMO, Scripps Hospital, ASCO, ESMO, CME outfitters; Financial Interests, Personal, Research Funding: NCI, EMD Serono, MedImmune, Atterocor, Amplimmune, ARMO biosciences, Karyopharm, Incyte, Novartis, Regeneron, Merck, BMS, Pfizer, CytomX Therapeutics. C. Mantia: Financial Interests, Personal, Advisory Board, I participated in a one-time advisory board: Aadi Bioscience; Financial Interests, Institutional, Coordinating PI, Institutional funding for research: Bristol Myers Squibb. N. Pennell: Non-Financial Interests, Personal, Advisory Role: Lilly, Merck, Genentech, Pfizer, Mirati Therapeutics, Janssen Oncology, Sanofi, ResistanceBio, Takeda, Novartis, Vial, Bayer, Anheart Therapeutics, Summit Therapeutics, Lovance Biotherapeutics; Financial Interests, Institutional, Research Funding: AZ, Merck, Loxo, Spectrum Pharmaceuticals, BMS, Mirati Therapeutics, Sanofi, Anheart Therapeutics, Navire. H.R. Kim: Non-Financial Interests, Personal, Speaker’s Bureau: Ono Pharmaceutical, Roche/Genentech. A.B. El-Khoueiry: Non-Financial Interests, Personal, Advisory Role: BMS, Bayer, Eisai, Roche, Merck, Exelixis, Pieris Pharmaceuticals, Agenus, Gilead, AZ/MedImmune, ABL bio, QED Therapeutics, SERVIER, Tallac Therapeutics, Senti Biosciences, Qurient; Non-Financial Interests, Personal, Royalties: Bayer, BMS, Roche/Genentech, EMD Serono, Eisai, Merck, Agenus, Exelixis, Gilead, AZ/MedImmune, ABL bio, QED Therapeutics, Servier, Tallac Therapeutics, Senti Biosciences, Qurient; Non-Financial Interests, Personal, Research Funding: AZ, Astex, Fulgent. D.E. Gerber: Non-Financial Interests, Personal, Advisory Role: Catalyst Pharmaceuticals, Janssen Oncology, Sanofi, Regeneron, DSI, Elevation Oncology; Non-Financial Interests, Personal, Proprietary Information: UTSD; Other, Personal, Other: OncoSeer Diagnostics; Financial Interests, Personal, Stocks or ownership: Gilead, Walgreens; Financial Interests, Institutional, Research Funding: BerGenBio, Karyopharm, AZ, Novocure. A. Qin: Non-Financial Interests, Personal, Advisory Role: Amgen; Financial Interests, Institutional, Research Funding: Takeda, Clovis Oncology, Merck, Xencor, AZ, Roche. M. Altan: Non-Financial Interests, Personal, Advisory Role: BMS, GSK, AZ; Non-Financial Interests, Personal, Speaker’s Bureau: Nektar; Financial Interests, Institutional, Research Funding: Lilly, BMS, Novartis, GSK, Jounce, Adaptimmune, Merck, Genentech, Nektar, Shattuck Labs. L.J. Appleman: Non-Financial Interests, Personal, Advisory Role: AADi; Financial Interests, Institutional, Research Funding: Pfizer, Exelixis, BMS, Astellas Pharma, Novartis, Bayer, Merck, Genentech, AVEO, Peloton Therapeutics, Calithera Biosciences, Seagen, Inovio Pharmaceuticals, Eisai, Lilly, Amgen, Surface Oncology, BioNTech, Epizyme, Janssen Oncology, Ipsen, Arvinas. J. Hill: Non-Financial Interests, Personal, Full or part-time Employment: Surface Oncology; Non-Financial Interests, Personal, Proprietary Information: Surface Oncology; Financial Interests, Personal, Stocks or ownership: Surface Oncology; Financial Interests, Personal, Research Funding: Surface Oncology. V. Reichert: Non-Financial Interests, Personal, Full or part-time Employment: Coherus Biosciences, Surface Oncology, Deciphera; Financial Interests, Personal, Stocks or ownership: Coherus Biosciences, Surface Oncology, Deciphera; Non-Financial Interests, Personal, Advisory Role: Kineta, Checkpoint Therapeutics. R. Masia: Non-Financial Interests, Personal, Full or part-time Employment: Surface Oncology. L. Harshman: Financial Interests, Personal, Full or part-time Employment: Surface Oncology; Financial Interests, Personal, Stocks or ownership: Surface Oncology. A. Patnaik: Financial Interests, Institutional, Other, Institutional research funding: Compugen. D. Morgensztern: Non-Financial Interests, Personal, Advisory Role: AbbVie, G1 Therapeutics, Lilly Medical, Mirati Therapeutics, Arcus Biosciences; Financial Interests, Personal, Stocks or ownership: BMS, Abbott Labratories; Financial Interests, Institutional, Research Funding: Heat Biologics, Merck, Celgene, AZ, Baxter, Incyte, AbbVie, BMS, EpicentRx, Pfizer, Roche, Lilly, Altum Pharmaceuticals, Array Biopharma, Surface Oncology, Arcus Biosciences, BI, Y-mAbs Therapeutics. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

123P - A phase II study of nivolumab (N) plus ipilimumab (I) and ASTX727 or N plus I in PD-1/PD-L1 resistant melanoma or NSCLC patients: the run-in phase of the NIBIT Foundation ML1 Study

Presentation Number
123P
Lecture Time
00:45 - 00:45
Speakers
  • Anna M. Di Giacomo (Siena, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Our phase Ib NIBIT Foundation M4 study firstly reported that administration of the hypomethylating agent (DHA) guadecitabine (guade), a prodrug of decitabine (D), followed by I in metastatic melanoma (MM) patients (pts) is safe and has promising clinical and tumor immune-modulating activity (Di Giacomo, CCR 2019). To further explore the activity of DHA combined with immune-checkpoints blockade, the NIBIT-ML1 trial is investigating the efficacy of guade plus I and N in PD-1/PD-L1 resistant MM or NSCLC pts. The run-in phase of the study is reported.

Methods

The NIBIT-ML1 is a randomized, phase II study (Simon two stages optimal design), in unresectable Stage III/IV MM (Cohort A) or NSCLC (Cohort B) pts progressing to anti-PD-1/PD-L1. A trial amendment substituted guade with ASTX727, an oral formulation of D with cedazuridine. Following the safety run-in of 6 subjects per Cohort, eligible pts are randomized to ASTX727 plus I and N or I and N. Primary objective is immune(i)-ORR; secondary are safety, DCR, PFS, median OS, survival rate at 1- and 2-year. For PK analyses D was measured in plasma as active drug after oral ASTX727 or as active metabolite after s.c. guade. Extensive cellular and molecular immunocorrelates are also explored.

Results

Cohort A. Six pts [4 male; median age 71 years, ECOG 0-1] with Stage III/IVMM, received guade (2 pts) or ASTX727 (4 pts) plus I and N. No DLT or overlapping toxicities occurred. Any grade (G) treatment-related AEs occurred in 6 (100%) pts, 50% were G3/4. Three PR, 2 SD, and 1 PD were observed. Cohort B. Six pts [2 male; median age 70 ECOG 0-1] with Stage III/IV NSCLC received guade (2 pts) or ASTX727 (4 pts) plus I and N. Two DLTs occurred. Any G treatment-related AEs occurred in 5 (83.3%) pts, 67% were G3/4. One CR, 2 SD and 3 PD were observed. PK analyses showed that D systemic exposures (mean AUC) dosed as 20mg ASTX727 and dosed at 80mg guade (∼45 mg/m2) were similar. Mean (% CV) D AUC 0-24 for combined Cohorts A and B after ASTX727 was 114 (90%) ng*hr/mL (n=8), and after guade was 110 (21%) ng*hr/mL (n=4).

Conclusions

Treatment with ASTX727 plus I and N is feasible in PD-1/PD-L1 refractory MM and NSCLC pts. The Stage 1 of the NIBIT-ML1 study is recruiting.

Clinical trial identification

NCT04250246, EudraCT 2019-002986-36.

Legal entity responsible for the study

NIBIT Foundation Onlus.

Funding

NIBITFoundation Onlus with unresctricted grant from Astex Pharmaceuticals and Bristol Myers Squibb.

Disclosure

A.M. Di Giacomo: Financial Interests, Personal, Advisory Board: Incyte, Pierre Fabre, GSK, Bristol Myers Squibb, Merck Sharp Dohme, Sanofi; Financial Interests, Personal, Other, Educational activities: Bristol Myers Squibb, Merck Sharp Dohme, Pierre Fabre, Sanofi. L. Calabro: Financial Interests, Personal, Advisory Board: Bristol Myers Squibb, Merck Sharp and Dohme, Roche; Financial Interests, Personal, Other, Educational activities: Bristol Myers Squibb, AstraZeneca, Sanofi. M. Valente: Financial Interests, Personal, Advisory Board: Novartis. H.N. Keer, A. Oganesian, D. Chan: Financial Interests, Personal, Full or part-time Employment: Astex Pharmaceuticals. M. Ceccarelli: Financial Interests, Personal, Advisory Board: Moderna Therapeutics; Financial Interests, Personal, Funding: Immunomica srl. M. Maio: Financial Interests, Personal, Advisory Board: Roche, Bristol Myers Squibb, Merck Sharp Dohme, Incyte, AstraZeneca, Amgen, Pierre Fabre, Eli Lilly, GSK, Sciclone, Sanofi, Alfasigma, Merck Serono; Financial Interests, Personal, Ownership Interest: Theravance, Epigen Therapeutics, Srl. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

107P - Final analysis of a phase II trial of neoadjuvant chemoimmunotherapy for locally advanced squamous cell carcinoma of the head and neck

Presentation Number
107P
Lecture Time
00:45 - 00:45
Speakers
  • Kunyu Yang (Wuhan, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

124P - Surufatinib plus toripalimab combined with etoposide (E) and cisplatin (P) in patients (pts) with advanced naive small cell lung cancer (SCLC) -Updated results of a phase ?b/? trial

Presentation Number
124P
Lecture Time
00:45 - 00:45
Speakers
  • Wen Feng Fang (Guangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Surufatinib (S) is a novel, small-molecule inhibitor that simultaneously targets VEGFR1-3, FGFR1 and CSF-1R. In a phase 2 study, S plus toripalimab (T, an anti-PD-1 antibody) showed promising efficacy in advanced SCLC pts failing with first-line(1L) chemotherapy. We previously reported preliminary results of NCT04996771, which demonstrated that S plus T and EP had encouraging efficacy as 1L regimen for SCLC. Here, we present an updated results.

Methods

This single-arm, open-label study consists of 3+3 dose-escalation (phase Ⅰb) and dose expansion (phase II). Eligible pts were≥18 years old with histologically confirmed advanced SCLC, ECOG PS 0-1, with at least one measurable lesion. Pts with treated, stable, and asymptomatic brain metastases are allowed. In phase Ib, S was dosed at 150mg - 250mg qd, po, Q3W, in combination with a fixed dose of T (200mg, iv, d1, Q3W) and EP (Q3W). After 4 cycles followed by maintenance therapy with S plus T, Q3W. The primary endpoints are RP2D of S and PFS. The secondary endpoints include ORR, DCR, OS, and safety.

Results

At cutoff date (Aug 31, 2023), 39 pts were enrolled. The median age was 64 years with male 82.1%, ECOG PS 1 59.0%, TNM stage IV 89.7%. Pts with liver, bone, and brain metastases were 33.3%, 30.8%, and 12.8%, respectively. The RP2D of S was identified as 200mg, po, qd, Q3W. Among pts with at least one post-baseline tumor assessment (n=35), median PFS was 6.0 months (95%CI 4.7, 7.3). Compared with pts with liver metastases, pts without metastases showed significantly longer PFS (5.7m vs 8.4m, p=0.0169). So as pts with or without bone metastases (5.7m vs 8.4m, p=0.0198). The ORR was 97.1% and DCR was 100%. The most common (≥10%) grade ≥3 treatment-emergent adverse events (TEAEs) were neutrophil count decreased (31.6%), white blood cell count decreased (23.7%), and platelet count decreased (10.5%).

Conclusions

Surufatinib plus toripalimab combined with etoposide and cisplatin showed encouraging anti-tumor activity and acceptable toxicity for the 1L treatment of advanced SCLC, especially in patients without liver metastases or bone metastases. The combination of the 4 agents might be a novel first-line therapeutic option for SCLC.

Clinical trial identification

NCT04996771; Release date: November 9, 2021.

Legal entity responsible for the study

Sun Yat-sen University Cancer Center.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

126P - Evaluation of Myeloid Targeting Agents, PY159 and PY314, in Two Dose Expansion Phase 1b Trials in Platinum-Resistant Ovarian Cancer

Presentation Number
126P
Lecture Time
00:45 - 00:45
Speakers
  • Oladapo O. Yeku (Boston, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Novel treatment options are required in patients with platinum-resistant ovarian cancer (PROC). Myeloid-derived suppressor cells facilitate a hostile tumor microenvironment and are associated with worse clinical outcomes in PROC. We evaluated the safety and preliminary efficacy of PY159, an agonist antibody to TREM1 that reprograms immunosuppressive intratumoral myeloid cells and PY314, an antagonist antibody to TREM2 that depletes tumor associated macrophages, as a single agent and in combination with pembrolizumab in subjects with PROC.

Methods

PY159 and PY314 were individually evaluated in patients with PROC. Subjects were treated with monotherapy (PY159 3 mg/kg or PY314 10 mg/kg), based on the recommended dose for expansion derived from the Phase 1a studies. At the time of first progression, subjects could continue study drug and crossover to combination therapy with pembrolizumab (200 mg) every 3 weeks at the discretion of the investigator. Disease assessment by RECIST 1.1 was performed every 6 weeks.

Results

17 subjects were enrolled in the PY159 study (median age 67, range 22-77; median prior therapies 6, range 2-18) and 16 subjects in PY314 (median age 65.5, range 49-81; median prior therapies 4, range 2-18). 7 subjects in PY159 and 8 subjects in PY314 crossed over to combination therapy. Safety events included the following: TRAEs occurred in 9 (56.3%) subjects in PY314 and 15 (88.2%) in PY159. IRRs occurred in 3 (18.8%) subjects in PY314 and 6 (35.3%) in PY159. irAEs occurred in 1 (6.3%) subject in PY314 (diarrhea) and 13 (76.5%) in PY159 (arthralgias). SAEs occurred in 12 (75%) subjects in PY314 (all unrelated) and 6 (36.3%) in PY159 (1 related). The best radiographic response in PY159 was stable disease in 8/16 subjects (50%; median 16 weeks, range 9-33), and in PY314, it was stable disease in 8/16 subjects (50%; median 12 weeks, range 6-36). Median PFS was 2.76 months and 2.69 months in PY159 and PY314, respectively. There were no responses in the crossover arm.

Conclusions

Both PY159 and PY314 were well tolerated, with an acceptable safety profile, as a single agent and in combination with pembrolizumab. Both agents warrant further investigation in heavily pretreated PROC.

Clinical trial identification

PY159: NCT04682431; PY314: NCT04691375.

Legal entity responsible for the study

The authors.

Funding

Pionyr Immunotherapeutics Inc.

Disclosure

O.O. Yeku: Financial Interests, Personal, Other, Consultant: GIMV NV, TigaTx Inc; Financial Interests, Personal, Advisory Board: hC Bioscience; Financial Interests, Personal, Full or part-time Employment, Associate Editor: NEJM Evidence; Financial Interests, Personal, Other, Patent Pending: MUC16 Directed Antibodies for therapeutic applications; Financial Interests, Personal, Other, Patent pending: Human Artificial Chromosomes for therapeutic applications; Financial Interests, Institutional, Local PI: Ascendis Pharma A/S, Avenge Bio, Inc, Duality Biologics, Immunocore Limited, Merck Sharp & Dohme Corporation, Pionyr Immunotherapeutics Inc, ProfoundBio. J.S. Wang: Financial Interests, Personal, Invited Speaker, Previous Member of Speaker's Bureau for Tagrisso and Imfinzi, stopped in 2021: AstraZeneca; Financial Interests, Personal, Invited Speaker, Previous Member of Speaker's Bureau for Lenvima, stopped 2021: Eisai; Financial Interests, Institutional, Local PI: 7,8 Pharma, Accutar, Adagene, Artios Pharma, AstraZeneca, Blueprint, Boehringer Ingelheim, Celgene/BMS, Clovis Pharma, Cyteir, Daiichi Sankyo, Forty Seven, Genentech/Roche, Hotspot, ImmunoOnc, Janssen, Kymab, LSK BioPartners, MabSpace, Macrogenics, Moderna, Novartis, Nurix, ORIC, Olema Therapeutics, Prelude Therapeutics, Ribon Therapeutics, Syndax, Teneobio, Zymeworks; Financial Interests, Institutional, Coordinating PI: Astellas, Bayer Healthcare, BeiGene, Bicycle Therapeutics, BioNTech, BioTheryX, Biosplice, Cullinan, Erasca, GSK, H3 Biomedicine, Hutchinson MediPharma, IGM Biosciences, Immuno-Gen, Jazz Pharma, Klus Pharma, Medikine, NGM Bio, Phoenix Molecular Designs, Pionyr, PureTech Health, Revolution Medicines, Pyxis, Qilu Pugent Sound, Relay Therapeutics, Sanofi, StingThera, TopAlliance, Treadwell Therapeutics, Xencor. P. Lorusso: Financial Interests, Personal, Advisory Board: AbbVie, Genmab, Genentech, CytomX, Takeda, Cybrexa, Agenus, IQVIA, TRIGR, Pfizer, ImmunoMet, Black Diamond, GSK, QED Therapeutics, AstraZeneca, EMD Serono, Shattuck, Astellas, Salarius, Silverback, MacroGenics, Kyowa Kirin, Kineta, Zentalis, Molecular Templates, Bayer, ABL Bio, STCube, Relay Therapeutics, Stemline, Compass BADX, BAKX Therapeutics, Scenic Biotech, Qualigen, Roivant Sciences, NeuroTrials, Seagen, imCheck, Mekanist, Mersana; Financial Interests, Personal, Other, Data Safety Monitoring Board: Agios, Five Prime, Halozyme; Financial Interests, Personal, Other, imCORE Alliance: Roche-Genentech; Financial Interests, Personal, Other, Consultant: Sotio, SK Life; Financial Interests, Personal, Other, Data Safety Monitoring Committee: Tyme; Financial Interests, Personal, Stocks/Shares: BAXK; Financial Interests, Institutional, Local PI: AbbVie, ADC Therapeutics, Boehringer Ingelheim, ALX Oncology, Astellas Pharma, Astex Pharmaceuticals, AstraZeneca, Bayer, Black Diamond, Calico Life Sciences, Corvus Pharmaceuticals, CytomX Therapeutics, Eisai Pharmaceuticals, Eli Lilly, EMD Sernono, Five Prime, FLX Bio, F-Star Delta Limited, Genentech, MedImmune, Genmab, Incyte, Linnaeus Therapeutics, Merck Sharp & Dohme, Moderna Therapeutics, NextCure, Pfizer, Takeda, Ribon Therapeutics, Sotio, Stemline Therapeutics, Tesaro, Jounce; Financial Interests, Personal, Advisory Board, Advisory Board & Consultant: I-Mab; Non-Financial Interests, Personal, Other, AACR Methods in Clinical Cancer Research Workshop - Co-Director: American Association for Cancer Research; Non-Financial Interests, Personal, Other, AACR Annual Report Committee - Member: American Association for Cancer Research; Non-Financial Interests, Personal, Other, Continuing Medical Education Committee - Member: American Association for Cancer Research; Non-Financial Interests, Personal, Other, Molecular Cancer Therapeutics Editorial Board - Member: American Association for Cancer Research; Non-Financial Interests, Personal, Other, ASCO Conquer Cancer Young Investigator Award Grand Selection Committee - Member: American Society of Clinical Oncology; Non-Financial Interests, Personal, Other, Chair - Phase 0 Task Force: American Association of Cancer Research; Non-Financial Interests, Personal, Other, AACI Clinical Research Innovation Steering Committee - Member: Association of American Cancer Institutes; Non-Financial Interests, Personal, Other, New Drugs in Oncology Seminar Planning Committee - Member: American Society of Clinical Oncology; Non-Financial Interests, Personal, Other, New Agents Committee: Translational Research Panel - Chair: Cancer Research Unite Kingdom; Non-Financial Interests, Personal, Other, Scientific Advisory Board - Member: Targeted Anti-Cancer Therapies; Non-Financial Interests, Personal, Member: American Association for Cancer Research, ASCO; Other, Personal, Other, Investigational Drug Steering Committee - Committee Member: National Cancer Institute; Other, Personal, Other, Phase I Special Emphasis Panel - Grant Reviewer/Discussion Leader: National Cancer Institute; Other, Personal, Other, NeXT Special Emphasis Panel - Grant Reviewer: National Cancer Institute/National Institute of Health; Other, Personal, Other, Board of Scientific Counselors, Clinical Sciences & Epidemiology: National Cancer Institute; Other, Personal, Other, Young Investigator Meeting, Cancer Therapy Evaluation Program - Professor: National Cancer Institute; Other, Personal, Other, Academic Advisory Board SPORE GI Malignancies - Case Western Reserve University: Case Western Reserve University; Other, Personal, Other, Scientific External Advisory Board - Member: University of California at San Diego; Other, Personal, Other, External Advisory Board - Member: University of Arizona; Other, Personal, Other, External Scientific Advisory Board - Member: University of New Mexico. A. Naqash: Financial Interests, Personal, Other, Social Media Editor and Consultant: JCO Precision Oncology; Financial Interests, Institutional, Local PI: Loxo, Surface Oncology, ADC Therapeutics, IGM Biosciences, Nikang Therapeutics, Inspirna, Revolution Medicine, Jacobio, Pionyr, Jazz, NGM; Financial Interests, Institutional, Coordinating PI: EMD Serono, Aravive; Non-Financial Interests, Personal, Other, Social Media Consultant and Scientific Committee Member: ASCO; Non-Financial Interests, Personal, Other, Educational Activity: OncLive; Non-Financial Interests, Personal, Leadership Role, Scientific Committee Co-Chair: ORIEN; Non-Financial Interests, Personal, Leadership Role, Early Career Scientist Committee: SITC. J. Hubbard: Financial Interests, Personal, Advisory Board: Merck; Financial Interests, Institutional, Advisory Board: Incyte, Bayer, BeiGene; Financial Interests, Institutional, Local PI: Boston Biomedical, Senhwa Biosciences, Bayer, Merck, Hutchison MediPharma, Seattle Genetics, Tovogene, TriOncology, Incyte, Pionyr, G1 Therapeutics, eFFECTOR Therapeutics, Roche; Financial Interests, Institutional, Coordinating PI: Taiho Pharmaceutical, TreoBio. S. Kummar: Financial Interests, Personal, Advisory Board: Bayer, Gilead, Mundibiopharma, Boehringer Ingelheim, Springworks Theraepeutics, HarbourBiomed, Boehringer Ingelheim, Oxford BioTherapeutics; Financial Interests, Personal, Advisory Board, Spouse: Cadila Pharmaceuticals; Financial Interests, Personal, Other, Chair, DSMC: Mirati; Financial Interests, Personal, Advisory Board, Consultant: Genome Insight; Financial Interests, Personal, Ownership Interest: Pahtomiq; Financial Interests, Personal, Ownership Interest, Spouse (co-founder): Arexeon; Financial Interests, Personal, Other, co-founder: Pathomiq; Financial Interests, Institutional, Local PI: ADC Therapeutics, Pionyr Therapeutics, Eisai, Bristol Myers Squibb, Syndax, SeaGen, ORIC, EMD Serono, Genome & Company, Moderna, Amgen, Elevation Oncology, VelosBio Inc, Gilead, Day One Biopharmaceuticals, ASTX Therapeutics, PMV Pharmaceuticals, Vincerx Pharma, Inc; Financial Interests, Institutional, Local PI, trial funding: 23&Me. C.E. Kyriakopoulos: Financial Interests, Personal, Advisory Board: Exelixis, AVEO, Sanofi-Aventis, EMD Serono, Janssen Pharmaceuticals; Financial Interests, Personal, Writing Engagement: Pfizer; Financial Interests, Personal, Stocks/Shares: Biogen, Epic Systems; Financial Interests, Institutional, Local PI: Gilead, Incyte Corporation, Sanofi-Aventis, AstraZeneca. E. Schenk: Financial Interests, Personal, Invited Speaker: OncLive, Ideology Health, MJH Life Sciences, Sanofi, MedPro, Janssen; Financial Interests, Personal, Advisory Board: Prescient Advisory, G1 Therapeutics, Regeneron, BioAtla; Non-Financial Interests, Personal, Other, Expert member, NSCLC guidelines panel: ASCO. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

116TiP - Randomized, open-label, phase II study of botensilimab (BOT) alone and in combination with balstilimab (BAL) versus standard-of-care in patients with refractory metastatic colorectal cancer

Presentation Number
116TiP
Lecture Time
00:45 - 05:00
Speakers
  • Eric Van Cutsem (Leuven, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Over 95% of patients with metastatic colorectal cancer (mCRC) are ineligible for immune checkpoint inhibitors as standard of care due to a lack of observed response in microsatellite stable/mismatch repair proficient (MSS/pMMR) tumors. Current standard of care includes chemotherapy and targeted therapy regimens. Botensilimab (BOT) – a multifunctional Fc-enhanced anti-CTLA-4 antibody designed to enhance T cell priming, activation, and memory formation; deplete intratumoral Treg cells; and minimize complement fixation – has demonstrated preclinical and promising early phase clinical data. In an ongoing Phase 1 study (NCT03860272), BOT ± balstilimab (BAL; anti-PD-1) has shown responses across a wide variety of cold/I-O refractory solid tumors, particularly in CRC patients without active liver metastases, with a managable safety profile. The aim of this randomized Phase 2 study is to evaluate the clinical efficacy and safety of BOT alone and in combination with BAL in patients with refractory non-microsatellite instability-high (non-MSI-H)/non-deficient MMR mCRC.

Trial design

This study (NCT05608044) includes adults ≥18 years old with confirmed non-MSI-H/non-deficient MMR mCRC who have received ≥1 prior chemotherapy regimen for metastatic disease with fluoropyrimidine, oxaliplatin, and irinotecan plus a monoclonal antibody as appropriate. Patients are excluded if they have active liver metastases or have previously received an immune checkpoint inhibitor, regorafenib, or trifluridine/tipiracil. Patients are randomized 1:1:1:1:1 to the following treatments: Arm A: BOT dose 1 every 6 weeks (Q6W) and BAL dose Q2W; Arm B: BOT dose 2 Q6W and BAL dose Q2W; Arm C: BOT dose 1 Q6W; Arm D: BOT dose 2 Q6W; Arm E: Investigator choice of regorafenib or trifluridine/tipiracil until disease progression/unacceptable toxicity. The primary endpoint is objective response rate (ORR) by RECISTv1.1. Secondary endpoints include duration of response, progression-free survival, overall survival, and safety. Global enrollment is ongoing with 111 patients currently enrolled across 8 countries.

Clinical trial identification

NCT05608044.

Editorial acknowledgement

Editorial assistance was provided by Dan Rigotti and Frankie Sorrell, Excel Medical Affairs, Glasgow, UK.

Legal entity responsible for the study

Agenus Inc.

Funding

Agenus Inc.

Disclosure

E. Van Cutsem: Financial Interests, Personal, Advisory Board: AbbVie, ALX, Amgen, Array, Astellas, AstraZeneca, Bayer, BeiGene, Boehringer Ingelheim, Bristol Myers Squibb, Daiichi, GSK, Incyte, Ipsen, Lilly, Merck Sharp & Dohme, Merck KGaA, Mirati, Novartis, Nordic, Pierre Fabre, Pfizer, Roche, Seattle Genetics, Servier, Takeda, Terumo, Taiho, Zymeworks; Financial Interests, Institutional, Research Grant: Amgen, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Ipsen, Lilly, Merck Sharp & Dohme, Merck KGaA, Novartis, Roche, Servier. M. Fakih: Financial Interests, Personal, Advisory Board, Consultant: AstraZeneca, Bayer Corporation, Bristol Myers Squibb; Financial Interests, Personal, Advisory Board, One meeting: Eisai Oncology; Financial Interests, Personal, Advisory Board, One meeting: Entos, Merck, Seattle Genetics, Xenthera; Financial Interests, Personal, Advisory Board, Also Editorial Boards & Consulting: Mirati Therapeutics; Financial Interests, Personal, Advisory Board: Nouscom, Roche/Genentech; Financial Interests, Personal, Advisory Board, Consulting: Pfizer, Taiho Oncology; Financial Interests, Institutional, Research Grant: AgenusBio, Genentech / imCORE, Verastem. N.H. Segal: Financial Interests, Personal, Advisory Role: Agenus, Puretech, Novartis, Numab, AstraZeneca, GSK, ABL Bio, Revitope, Roche/Genentech, Boehringer Ingelheim; Financial Interests, Personal, Funding: AstraZeneca, Regeneron; Financial Interests, Institutional, Funding: Roche/Genentech, Pfizer, Merck, BMS, AstraZeneca, Puretech, Immunocore, Regeneron, Agenus. B. Johnson: Financial Interests, Personal, Research Grant: Gateway for Cancer Research, Syntrix , BMS; Financial Interests, Personal, Speaker, Consultant, Advisor: Incyte, Taiho Oncology, Insmed Oncology, Iota, Gritstone Bio. A. Kardosh: Financial Interests, Personal, Funding: Nastera Inc.; Financial Interests, Personal, Speaker, Consultant, Advisor: Seagen, AstraZeneca, Genentech, Exelixis, Seagen. J. Dekervel: Financial Interests, Personal, Invited Speaker: Amgen, Bayer, Eisai, Ipsen, Lilly, Merck, MSD, Roche; Financial Interests, Personal, Advisory Board: BMS, Eisai, Novartis, Roche; Financial Interests, Institutional, Research Grant: Bayer. B.L. Schlechter: Financial Interests, Personal, Advisory Board: Janssen Pharmaceuticals, Quercegen Pharmaceuticals. J. Grossman: Financial Interests, Personal, Full or part-time Employment: Agenus; Financial Interests, Personal, Other, Patent: Agenus; Financial Interests, Personal, Stocks/Shares: Agenus. M.E. Elez Fernandez: Financial Interests, Personal, Advisory Board: Hoffman-La Roche, Servier, Amgen, Merck Serono, Sanofi, Bayer, Pierre Fabre, MSD, Takeda; Financial Interests, Personal, Invited Speaker: Organon, Novartis, Pfizer; Financial Interests, Personal, Other, Educational training: Seagen International GmbH; Financial Interests, Institutional, Funding: Hoffmann-La Roche Ltd, Sanofi Aventis Recherche & Développement, Amgen Inc., Boehringer Ingelheim, Novartis Farmacéutica SA, Bristol Myers Squibb International Corporation, BeiGene, HalioDX SAS, Janssen-Cilag SA, Merck Health KGAA, Merck Sharp & Dohme de España SA, PharmaMar SA, Servier, Taiho Pharma USA Inc, Hutchison MediPharma International, Menarini, Merus NV, Pfizer, Mirati, Array Biopharma Inc, AstraZeneca Pharmaceuticals LP, Celgene International SARL, Debiopharm International SA, Genentech Inc, MedImmune; Non-Financial Interests, Personal, Other, Coordinator of the SEOM +MIR Section of Residents and Young Assistants: Sociedad Española de Oncología Médica (SEOM); Non-Financial Interests, Personal, Other, Speaker of the ESMO Academy: European Society for Medical Oncology (ESMO); Non-Financial Interests, Personal, Other, Volunteer member of the ASCO Annual Meeting Scientific Program Committee: Developmental Therapeutics – Immunotherapy: American Society of Clinical Oncology (ASCO); Other, Personal, Other, Travel, accommodations, expenses: Roche, Merck Serono, Sanofi, Amgen, Array BioPharma, Servier, Bristol Myers Squibb. C. Eng: Financial Interests, Personal, Advisory Board: AbbVie, Amgen, California Institute of Regenerative Medicine, Hoopika, Janssen, Merck, Taiho, Takeda, Seagen; Financial Interests, Institutional, Local PI: Janssen, Gritstone, Pfizer, Merck, Sumitomo, Agenus; Financial Interests, Institutional, Research Grant: Haystack, Natera; Financial Interests, Institutional, Coordinating PI: Hutchinson. M.P. Ducreux: Financial Interests, Personal, Invited Speaker: Roche, Amgen, Pierre Fabre, Merck Kga, Pfizer, Bayer, Lilly, Servier, MSD; Financial Interests, Personal, Advisory Board: Roche, Basilea, Sotio, Pierre Fabre, Boehringer, Rafael, Servier, Zymeworks, Ipsen, Bayer, HalioDX, Lilly, GSK, Daiichi Sankyo, MSD, Servier; Financial Interests, Institutional, Advisory Board: AstraZeneca; Financial Interests, Institutional, Funding, Partial funding of a trial evaluating the role of bevacizumab in NET: Roche; Financial Interests, Institutional, Funding, Partial funding of a trial evaluating the role of steptozotocin in NET: Keocyt; Financial Interests, Institutional, Local PI: Rafael, Amgen; Financial Interests, Institutional, Funding: Bayer; Other, Personal, Other, My wife is head of the oncology business unit in the French Affiliate of Sandoz France. T. André: Financial Interests, Personal, Advisory Board, Advosiry Board on February 12, 2021: Astellas pharma; Financial Interests, Personal, Advisory Board, Advisory Board on February 2021: Kaleido Biosciences; Financial Interests, Personal, Invited Speaker, and advisory board 2021: Amgen; Financial Interests, Personal, Invited Speaker, Invited speaker in a symposuim december 2020: AstraZeneca; Financial Interests, Personal, Advisory Board, and consultant fees and consultant contract 2021 and 2022: Bristol Myers Squibb; Financial Interests, Personal, Advisory Board, Advisory board in Janaury 2020: Clovis; Financial Interests, Personal, Advisory Board, Advisory board in January 2020: Gritstone Oncology; Financial Interests, Personal, Advisory Board, Advisory board 2020: Haliodx; Financial Interests, Personal, Advisory Board, and consultant fees/consultant contract and invited speaker: MSD Oncology; Financial Interests, Personal, Invited Speaker, and other: Pierre Fabre; Financial Interests, Personal, Invited Speaker, in an symposuim in 2020: Roche; Financial Interests, Personal, Invited Speaker, in a meeting in 2019: Ventana; Financial Interests, Personal, Invited Speaker in a educational meeting in 2019: Sanofi; Financial Interests, Personal, Advisory Board, in a symposuim in 2020: Servier; Financial Interests, Personal, Advisory Board, Consultant with personal fees and invited speaker: Servier; Financial Interests, Personal, Advisory Board, in 2019: GSK; Financial Interests, Personal, Invited Speaker, in 2020 and 2021: GSK; Financial Interests, Personal, Invited Speaker, Virtual symposium Lecture: 1 MSI-H CRC: Implementation of Immunotherapy in clinical practice (30 minutes) – (this will be pre-recorded)Q&A – Live Q&A – (10 minutes) (on July 2, 2021): MSD Oncology; Financial Interests, Personal, Invited Speaker, June 2022: Sanofi; Financial Interests, Personal, Advisory Board, Contract 2021, 2022, 2023: Merck & Co., Inc; Financial Interests, Personal, Advisory Board, Contrat 2019, 2020, 2021, 2022: BMS; Financial Interests, Personal, Advisory Board, Contract 2021: Gritstone Oncologie; Financial Interests, Personal, Invited Speaker, June 2022 and June 2023 during ESMO GI meeting: Seagen; Financial Interests, Personal, Writing Engagement, Contract of consulting 2021 and 2022 and 2023: MSD Oncology; Financial Interests, Personal, Advisory Board, September 2022: GSK, Seagen; Financial Interests, Personal, Writing Engagement, Contract of consulting 2022 and 2023: GSK; Financial Interests, Personal, Writing Engagement, Contract of consulting 2022: Nordic Pharma; Financial Interests, Personal, Invited Speaker, October 2022: Merk Serono; Financial Interests, Personal, Other, Educationnal in 2022: Roche; Financial Interests, Personal, Other, Contract of consulting 2020, 2021, 2022,2023: Servier; Financial Interests, Personal, Advisory Board, Gilead Sciences Global GI Advisory Board on January 18, 2023: Aptitude Health, Glilead; Financial Interests, Personal, Other, Consultant like member of the SGNTUC-029 Steering Committee (the “Steering Committee”) Services related to the Company’s Study titled “An Open-label Randomized Phase 3 Study of Tucatinib in Combination with Trastuzumab and mFOLFOX6 versus mFOLFOX6 given with or without either Cetuximab or Bevacizumab as First-line Treatment for Subjects with HER2+ Metastatic Colorectal Cancer” (collectively, the “Services”).Including teleconferences and board preparation before the board (6h)Participation of the Board on September 2022 in Paris, September 10, 2022 MOUNTAINEER-03 Steering Committee Meeting during ESMO: Seagen; Financial Interests, Personal, Other, Consultant like member of the to be a member of the Colorectal Program Scientific Advisory Committee (SAC). In charges for reviewing V940 Pembro on Engagezone (as per your contract in ref).and also an interview about Pembrolizumab for MSI/dMMR Metastaic colo-rectal 2023: MSD Oncology; Financial Interests, Personal, Advisory Board, but also formation for the Takeda team, and consultant for Takeda Contract beetween 1 sept 2023 and 15 october 2024: Takeda; Financial Interests, Personal, Advisory Board, Board Meeting (October 19, 2023): AbbVie; Financial Interests, Institutional, Coordinating PI, PI Garnet study: GSK; Financial Interests, Institutional, Coordinating PI, Keynote 164 and 171 and 811 and C08: MSD; Financial Interests, Institutional, Coordinating PI, BMS CA209-8HW, BMS CA209-142, BMS CA209-577: BMS; Financial Interests, Institutional, Coordinating PI, SPOTLIGHT study: Astellas; Financial Interests, Personal, Steering Committee Member, and international PI (trial chair Solstice study): Servier; Financial Interests, Personal, Steering Committee Member, MOUNTAINEER and MOUNTAINEER-03 study (PI for France) 2022, 2023: Seagen; Financial Interests, Institutional, Local PI, A randomized, open-label, phase 2 study of botensilimab (agen1181) as monotherapy and in combination with balstilimab (agen2034) or Investigator’s choice standard of care (regorafenib or trifluridine and tipiracil) for the treatment of refractory metastatic colorectal cancer: Agenus; Non-Financial Interests, Personal, Member of Board of Directors, Investigator: Gercor group; Non-Financial Interests, Personal, Member of Board of Directors, President since October 2022: ARCAD Foundation. J. Tabernero: Financial Interests, Personal, Advisory Board, scientific consultancy role: Orion Biotechnology, Array Biopharma, AstraZeneca, Bayer, Boehringer Ingelheim, Chugai, Daiichi Sankyo, F. Hoffmann-La Roche Ltd, Genentech Inc., HalioDX SAS, Ikena Oncology, IQVIA, Lilly, Menarini, Merck Serono, Merus, MSD, Mirati, Neophore, Novartis, Peptomyc, Pfizer, Pierre Fabre, Samsung Bioepis, Sanofi, Seattle Genetics, Servier, Taiho, Tessa Therapeutics, TheraMyc, Hutchinson MediPharma International, Scandion Oncology, Ona Therapeutics, Sotio Biotech, Inspirna Inc, Scorpion Therapeutics, Tolremo Therapeutics; Financial Interests, Personal, Invited Speaker, educational collaboration: Medscape Education, Physicians Education Resource (PER), PeerView Institute for Medical Education, Imedex / HMP; Financial Interests, Personal, Invited Speaker, educacional collaboration: MJH Life Sciences; Financial Interests, Personal, Advisory Board: Cardiff Oncology; Financial Interests, Personal, Stocks/Shares: Oniria Therapeuics; Financial Interests, Institutional, Research Grant, ACRCelerate: Colorectal Cancer Stratified: Fundación Científica de la Asociación Española Contra el Cáncer; Financial Interests, Institutional, Research Grant, OPTIMISTICC: Opportunity to Investigate the Microbiome’s Impact on Science and Treatment In Colorectal Cancer: Cancer Research UK; Financial Interests, Institutional, Funding, Clinical Trials & Research: Amgen Inc, Array Biopharma Inc, Debiopharm International SA, AstraZeneca Pharmaceuticals LP, Bristol Myers Squibb International Corporation, Celgene International SARL, F. Hoffmann-La Roche Ltd, Merck, Sharp & Dohme de España, SA, Genentech Inc, Janssen-Cilag International NV, Merck Health KGAA, Novartis Farmacéutica SA, PharmaMar SA, Sanofi-Aventis Recherche & Développement, Servier, Taiho Pharma USA, Inc, BeiGene, Boehringer Ingelheim, HalioDX SAS, Hutchinson Medipharma, MedImmune, Menarini, Merus N V, Pfizer, Mirati; Non-Financial Interests, Personal, Leadership Role, External Scientific Committee: IdiSNA –Universidad de Navarra, Institute for Health Research INCLIVA – Clinical Hospital of Valencia; Non-Financial Interests, Personal, Member of Board of Directors, Board of Directors: Cancer Core Europe, Spanish Association Against Cancer -AECC; Non-Financial Interests, Personal, Member of Board of Directors, General Assembly: Horizon Europe Cancer Mission; Non-Financial Interests, Personal, Leadership Role, Scientific Advisory Board: Spanish National Cancer Research Centre (CNIO); Non-Financial Interests, Personal, Advisory Role, International Scientific Evaluation Committee: Bosch Health Campus (BHC); Non-Financial Interests, Personal, Advisory Role, Review Board: National Decade Against Cancer (NCT) - German Consortium for Translational Cancer Research (DKTK); Non-Financial Interests, Personal, Advisory Role, Scientific Advisory Board: Karolinska Comprehensive Cancer Centre; Non-Financial Interests, Personal, Advisory Role, International Review Committee (IRC): Oncode Institute; Non-Financial Interests, Personal, Advisory Role, Scientific Advisory Board (SAB): Oslo University Hospital Comprehensive Cancer Centre (OUH CCC); Non-Financial Interests, Personal, Leadership Role, Governance Advisory Committee: European Organization for Research and Treatment of Cancer -EORTC; Non-Financial Interests, Personal, Leadership Role, Vice Chairman: World Innovative Networking (WIN) Consortium in Personalized Cancer Medicine; Non-Financial Interests, Personal, Other, Coordinating PI & Steering Committee Member. Clinical Trials & Research: AstraZeneca Pharmaceutical LP, Array Biopharma Inc., Boehringer Ingelheim, MedImmune, Menarini, Merck Healthcare KGAA, Merck, Sharp & Dohme de España SA, Pfizer, Servier; Non-Financial Interests, Personal, Principal Investigator, Clinical Trials & Research: Array Biopharma Inc., AstraZeneca Pharmaceutics LP, BeiGene, Boehringer Ingelheim, F. Hoffmann-La Roche Ltd, Debiopharm International SA, Bristol Myers Squibb International Corporation, Celgene International SARL, Genentech Inc., HalioDX SAS, Hutchinson Medipharma, Menarini, Janssen-Cilag International NV, MedImmune, Merck Healthcare KGAA, Merck, Sharp & Dohme de España SA, Mirati, Merus NV, Novartis Farmacéutica SA, Pfizer, Sanofi-Aventis Recherche & Développement, Servier, Taiho Pharma USA Inc; Non-Financial Interests, Personal, Other, Steering Committee Member. Clinical Trials & Research: Debiopharm International SA, F. Hoffmann-La Roche Ltd, Genentech Inc., HalioDX SAS, Hutchinson Medipharma, Janssen-Cilag International NV, Merus NV, Taiho Pharma USA Inc; Non-Financial Interests, Personal, Other, Coordinating PI. Clinical Trials & Research: Mirati; Non-Financial Interests, Personal, Member: AACR, ASCO, EACR, EORTC, SEOM; Other, Personal, Other, Advisory Committee: Advisory Committee on Pharmaceutical Provision Financing under the Spanish National Health System; Other, Personal, Other, President: Oncology Master Plan – Catalonia Department of Health. H.J. Lenz: Financial Interests, Personal, Advisory Board, Advisory Role and Lectures: Bayer; Financial Interests, Personal, Advisory Board, Advisory ROle and Lectures: Merck; Financial Interests, Personal, Advisory Board, Advisory Role and lecture: ROche; Financial Interests, Personal, Advisory Board: Jazz Pharmaceuticals, oncocyte, Orion, Astellas, BMS; Financial Interests, Personal, Advisory Board, Scientific Advisory Board: 3T Biosscience; Financial Interests, Personal, Advisory Board, Advisory Board: Fulgent, G1 Therapeutics, BioNtech; Financial Interests, Personal, Advisory Board, advisory board: Cardiff; Financial Interests, Personal, Stocks/Shares, Advisory Board: Fulgent. M. Hidalgo: Financial Interests, Personal, Research Grant: PanCan, Rank Therapeutics; Financial Interests, Personal, Licencing Fees or royalty for IP: Miriad, Khar; Financial Interests, Personal, Speaker, Consultant, Advisor: MinKi, Oncomatrix, Inxmed, Peaches, Khar; Financial Interests, Personal, Funding: Agenus; Financial Interests, Personal, Other, Board of Directors: BMS; Financial Interests, Personal, Stocks/Shares: Champions Oncology, Nelum, Inxmed. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

127P - REGN5668 (MUC16xCD28 bispecific antibody) with cemiplimab (anti-PD-1 antibody) in recurrent ovarian cancer: Phase 1 dose-escalation study

Presentation Number
127P
Lecture Time
05:00 - 05:00
Speakers
  • Ira S. Winer (Detroit, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

REGN5668 (R5668) is a mucin16 (MUC16) x CD28 bispecific antibody (bsAb). R5668 provides “signal 2” T-cell costimulation through CD28 in the presence of MUC16 tumor antigen. CD28 bispecifics augment anti-tumor activity of the anti-PD-1 monoclonal Ab cemiplimab (cemi) in preclinical studies. We present the phase 1 dose-escalation results of intravenous (IV) R5668 combined with cemi in patients (pts) with recurrent platinum-experienced ovarian cancer (OC).

Methods

Pts received weekly R5668 IV at a dose range of 0.3-300 mg. Cemi 350 mg IV every 3 weeks was added beginning Day 21-28. Primary endpoints were safety and R5668 PK. Secondary endpoints included confirmed objective response rate (RECIST 1.1) and CA-125 response (Gynecologic Cancer InterGroup). Multiplex cytokine profiling was exploratory.

Results

28 pts were enrolled; 22 (79%) received ≥1 dose of cemi. Median number of prior therapies was 3.5 (range 1−10). Median duration of R5668 and cemi exposure was 7.5 (range 1.9-39.0) and 6.1 (2.4−36.0) weeks, respectively. Fatigue (32%), nausea (29%), and pain (18%) were the most common treatment-related adverse events (TRAEs). Infusion-related reactions/cytokine release syndrome (all Grade [G]1/2) occurred in 14% of pts. One pt had a G≥3 TRAE (fatigue). There were no adverse events resulting in death or study drug discontinuation. No DLTs were observed and opening of the next cohort (1000 mg) is planned. Across all pts treated to date, 1 confirmed partial response (-59% target lesion reduction from baseline) and 1 CA-125 response were observed, both in a single pt at 300 mg. 6 (21%) pts had stable disease. There was a dose-dependent increase in R5668 exposure between 1 mg and 300 mg IV QW dosing. Cytokine analyses revealed no apparent elevations after R5668 dosing, while upon cemi addition, slight increases in IFNγ and IP10 were observed.

Conclusions

In 28 heavily pretreated OC pts, an acceptable safety profile, low rates of CRS, and early activity were observed with R5668 + cemi. R5668 dose escalation with cemi or ubamatamab (MUC16xCD3 bsAb) is ongoing.

Clinical trial identification

NCT04590326.

Editorial acknowledgement

Writing assistance was provided by Brian Head, PhD, of Regeneron Pharmaceuticals, Inc.

Legal entity responsible for the study

Regeneron Pharmaceuticals, Inc.

Funding

Regeneron Pharmaceuticals, Inc.

Disclosure

I.S. Winer: Financial Interests, Personal, Research Funding: Oncoceutics. R.E. O'Cearbhaill: Financial Interests, Personal, Other, Personal Fees: Bayer, Curio, Fresenius Kabi, Immunogen, MJH, Seattle Genetics and Tesaro/GSK, Miltenyi, 2seventybio; Financial Interests, Institutional, Other, Personal Fees: Regeneron; Financial Interests, Personal, Advisory Board: AbbVie/StemCentrx, Atara , Biotherapeutics, Bayer/Celgene/Juno, Genentech, Genmab/Seagen Therapeutics, Gynecologic OncologyFoundation, Kite Pharma, Ludwig Cancer Institute, Merck, Regeneron, Sellas Therapeutics, Syndax Pharmaceuticals, TCR2 Therapeutics, Lyell Therapeutics, Arsenal-Bio, Tesaro/GSK, GSK; Financial Interests, Personal, Steering Committee Member, Non-Compensated: PRIMA, Moonstone (Tesaro/GSK), and DUO-O (AstraZeneca) studies; Financial Interests, Personal, Advisory Board, Non-Compensated: Carina Biotech; Financial Interests, Personal, Other, Travel for meeting: Hitech Health. S. Bouberhan: Financial Interests, Personal, Other, Consulting Fees: ImmunoGen. J.L. Hays: Financial Interests, Personal, Advisory Role: AstraZeneca, Merck, Tesaro, Clovis Oncology, Deciphera, and Ipsen; Financial Interests, Personal, Other, Travel, accommodations, and expenses: Tesaro, Merck, and AstraZeneca. D.R. Roque: Financial Interests, Personal, Other, Honoraria: Intuitive Surgical; Financial Interests, Personal, Advisory Role: Myriad Genetics; Financial Interests, Personal, Speaker’s Bureau: GSK; Financial Interests, Personal, Other, Travel, accommodations, and expenses: Surgical. O.O. Yeku: Financial Interests, Personal, Advisory Board: TigaTx, Celldex, GIMV NV, hC Bioscience. J.F. Liu: Financial Interests, Institutional, Research Funding: Zentalis, Vigeo Therapeutics, Tesaro, Surface Oncology, Regeneron, Impact Therapeutics, GSK, CytomX Therapeutics, Clovis Oncology, Bristol Myers Squibb, AstraZeneca, Arch Oncology, Aravive, 2X Oncology; Financial Interests, Personal, Advisory Board: AstraZeneca, Bristol Myers Squibb, Clovis Oncology, Daiichi, Eisai, EpsilaBio, Genentech/Roche, GSK, Regeneron Pharmaceuticals Inc., Zentalis. B. Wang, S. Yoo, S. Govindraj, M. Zhu, J. Brouwer-Visser, M.J. Peterman, B. Barnes, I. Lowy, D. Knorr, T.S. Uldrick, E.A. Miller: Financial Interests, Personal, Full or part-time Employment: Regeneron Pharmaceuticals, Inc.; Financial Interests, Personal, Stocks/Shares: Regeneron Pharmaceuticals, Inc. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

128P - A phase I dose escalation and expansion trial of LYT-200, a Galectin-9 antibody +/- tislelizumab

Presentation Number
128P
Lecture Time
05:00 - 05:00
Speakers
  • Gerald S. Falchook (Denver, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Galectin-9 (gal9) is a promising novel target for the treatment of both solid tumors and hematologic malignancies. When overexpressed, gal9 has broad immunosuppressor effects disabling immune-mediated cancer attack via T cell modulation, macrophages and other immune functions. LYT-200 is a fully human, anti-gal9, IgG4 monoclonal antibody that blocks gal9 mediated immunosuppression and protumor function.

Methods

Safety, pharmacokinetics (PK), pharmacodynamics (PD) and efficacy of LYT-200 are being assessed in this Phase 1/2 trial (NCT04666688) in advanced solid tumors as monotherapy and in combination with a PD-1 inhibitor, tislelizumab (TIS) in patients (pts) with relapsed, refractory head and neck squamous cell carcinoma (HNSCC) or urothelial cancer (UC) where gal9’s role is thought to be prominent.

Results

In monotherapy, 20 pts received IV LYT-200 in 7 escalation dose cohorts of 0.2 mg/kg to 16 mg/kg Q2W or 10 mg/kg QW. Median prior regimens (MPR) were 4 (1-7). No DLTs or LYT-200 related SAEs or G≥3 AEs were seen. LYT-200 related AEs of G≤2 were seen in 5 pts. 2 infusion related reactions (IRR) occurred in pts with IRRs on prior treatment. Single-agent cohorts have completed with 10 mg/kg QW selected as clinically relevant dose. Of 19 RECIST evaluable pts, 3 had stable disease (SD): 2 pancreatic cancer pts at 2 mg/kg QW and 6.3 mg Q2W with SD for 18 and 4 months (mo), and a colorectal cancer pt for 13 mo at 10 mg/kg QW. In the initial LYT-200/TIS combination cohort (LYT-200 6.3 mg/kg QW + TIS 300 mg Q4W) 6 pts have started treatment with 4 RECIST evaluable to date. MPR was 3 (1-5). No DLTs, irAEs or treatment related SAEs or G≥3 AEs. Of the 3 evaluable HNSCC pts, there have been 2 responses: 1 CR on treatment 9+mo and 1 PR on treatment 8+ mo. The 1 evaluable UC pt has had SD for 4+ mo, with near resolution of pleural effusion and ascites. LYT-200 has linear, dose-proportional PK. Geometric mean LYT-200 half-life is 6.6 days, supporting QW dosing.

Conclusions

LYT-200 has an acceptable safety profile in monotherapy and TIS combination, with observed antitumor activity in R/R pts who historically have very low response rates to an anti-PD1 agent alone. Enrollment into LYT-200 + TIS arms continues. The presentation will include additional safety, PK, PD and efficacy data.

Clinical trial identification

NCT04666688.

Editorial acknowledgement

The authors.

Legal entity responsible for the study

PureTech Health.

Funding

PureTech Health.

Disclosure

C. Korth: Financial Interests, Institutional, Advisory Board, works for Sponsor: Puretech health. A. Filipovic: Other, Institutional, Other, Head of Oncology at PureTech Health who is the Sponsor of the study: PureTech health. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

129P - Naxitamab efficacy in patients with refractory/relapsed high-risk neuroblastoma and bone metastases as assessed by Curie score

Presentation Number
129P
Lecture Time
05:00 - 05:00
Speakers
  • Brian Kushner (New York, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

High-risk neuroblastoma (HR-NB) commonly metastasizes to the bone, a frequent site of resistant disease. Naxitamab, a humanized GD2-binding monoclonal antibody, in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF), has previously been shown to be effective in the treatment of bone metastases. Here we report the maximum change in Curie score (CS) from baseline based on Trial 201 prespecified interim analysis.

Methods

Trial 201 (NCT03363373, data cutoff 31 Dec 2021) is a Phase 2 study evaluating naxitamab in combination with GM-CSF in patients (pts) with HR-NB with an incomplete response in bone/bone marrow after induction or relapse therapy. Naxitamab was administered intravenously (3 mg/kg/day) on days 1, 3, and 5, and GM-CSF was administered subcutaneously on days -4 to 5 in 4-week cycles. CS and treatment response were evaluated by use of 123I-MIBG scans per independent review in accordance with the International Neuroblastoma Response Criteria. The current analysis reports on the maximum change in CS from baseline in the overall efficacy population and in relapsed and refractory subgroups.

Results

A total of 48 pts with evaluable disease in bone and at least 1 postbaseline CS were eligible for assessment. A summary of baseline and maximum CS change for the overall and relapsed/refractory subgroups are presented in the Table. Among the 20 (42%) pts with a 100% decrease in CS, the median and mean baseline CSs were 2.5 and 4.8, respectively. Notably, 46/48 (96%) pts achieved a decreased or stable CS. Related grade ≥3 adverse events reported in ≥10% of pts included pain, hypotension, urticaria, and bronchospasm and were manageable with appropriate interventions in place.

Overall (n=48) Refractory to induction (n=25) Refractory to relapse therapy (n=23)
Baseline CS Median (range) 3 (1-20) 3 (1-20) 3 (1-19)
Mean 5.5 5.4 5.7
Maximum CS change Median (range) -2 (-18 to 14) -2 (-18 to 0) -1 (-17 to 14)
Mean -3.3 -4.3 -2.3
Maximum % CS change Median -69 -85 -67
Mean (SD) -55 (59.2) -68 (37.5) -41 (74.5)

Conclusions

The maximum change in CS provides a measure of best bone response and demonstrates that naxitamab has the potential to significantly reduce the disease burden in the bones of pts with HR-NB. Safety was in line with prior observations.

Clinical trial identification

NCT03363373.

Legal entity responsible for the study

Y-mAbs Therapeutics.

Funding

Y-mAbs Therapeutics.

Disclosure

J. Mora: Financial Interests, Personal, Speaker, Consultant, Advisor: Y-mAbs. D.A. Morgenstern: Financial Interests, Personal, Speaker, Consultant, Advisor: Y-mAbs, Clarity Pharmaceuticals, Oncoheroes Biosciences, Razyebio, Regeneron, AbbVie. K. Nysom: Financial Interests, Personal, Advisory Board: Y-mAbs, Bayer, EUSA. J. Faber: Financial Interests, Personal, Local PI: Y-mAbs. A. Wingerter: Financial Interests, Personal, Advisory Board: Y-mAbs, EUSA. M. Bear: Financial Interests, Personal, Advisory Board: Y-mAbs. M. During, K. Tornøe: Financial Interests, Personal, Full or part-time Employment: Y-mAbs. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

131P - Safety and clinical efficacy of Roginolisib (IOA-244), the first oral allosteric modulator of phosphoinositide 3-kinase inhibitor delta (PI3K_)

Presentation Number
131P
Lecture Time
05:00 - 05:00
Speakers
  • Anna M. Di Giacomo (Siena, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The highly selective oral allosteric modulator of PI3Kδ, roginolisib, has a unique safety and pharmacodynamic profile showing immune modulation in patients with solid tumours.

Methods

IOA-244 was investigated in a two-part FIH study: Part A explored daily dosing of roginolisib at 10, 20, 40 and 80 mg in patients (pts) with solid tumours and Follicular Lymphoma (FL). Part B confirmed the 80 mg QD as the recommended Phase 2 dose in uveal melanoma (UM) pts. Primary objective: Safety. Secondary objectives: PK; PD (e.g., inhibition of CD63 expression on basophils, changes in immune cell subsets in peripheral blood); radiographic responses (RECIST 1.1. or Lugano); PFS and OS. Exploratory studies: changes in circulating immune cells by mass cytometry (Cytometry by Time of Flight; CyTOF); response assessments by radiomics and blood-based proteins.

Results

Part A Solid Tumour (completed): Sixteen pts were treated in 4 cohorts with uveal (9/16; 56%), cutaneous melanoma (5/16; 31%) and pleural mesothelioma (2/16; 13%). Part A NHL-FL pts (completed): 8 pts at 20 mg (n=4) and 80 mg (n=4). At 80 mg, 2/4 pts (50%) had PR as per Lugano criteria. Safety: No treatment-emergent adverse events (TEAE) led to study drug discontinuation, immune related toxicity, or a Dose Limiting Toxicity. UM evaluation (recruitment completed): 29 pts (Part A: 9 pts; Part B1/2: 20 pts; total 29 pts), of which 23 pts were treated at 80 mg QD. Mean time on treatment: 9.6 mo (range: 1.5-35.3 mo). ORR (RECIST 1.1): PR: 1/29 (3%); SD: 20/29 (69%). Median OS for Part A: 20.8 mo (7/9 with 2 pts alive); Part B: not determined; 1-year OS rate: Part A: 66.7%; Part B1: 71.4%, B2: ongoing. Exploratory Investigations: Radiomics-based studies showed mixed responses in CT images. Pts with reduction of plasma soluble CTLA-4 exhibited a reduction in blood Treg cells, while increased plasma IL-15 levels accompanied an increase in CD8+ T and NK cells. Patients with RECIST 1.1 defined SD at Cycle 5 (∼4.5 mo of treatment): stable LDH levels, increased plasma levels of IFNg and IFNg-induced proteins.

Conclusions

Roginolisib is well tolerated at the 80 mg dose. Long term administration (>6 months) translates to encouraging OS in UM and lymphoma pts.

Clinical trial identification

NCT04328844.

Legal entity responsible for the study

iOnctura SA.

Funding

iOnctura SA.

Disclosure

A.M. Di Giacomo: Financial Interests, Personal, Advisory Board: Bristol Myers Squibb, MSD, Pierre Fabre, Novartis; Financial Interests, Personal, Invited Speaker: Sanofi. M. Simonelli: Financial Interests, Personal, Advisory Board: Incyte, Cytovia; Financial Interests, Personal, Invited Speaker: GSK, Bristol Myers Squibb; Financial Interests, Personal, Other, Data Monitoring Committee: Sanofi. M. Lahn: Financial Interests, Personal, Officer: iOnctura; Financial Interests, Personal, Stocks/Shares: iOnctura. G. Di Conza, T. Hammett, R. Zorrilla, P. Kaur: Financial Interests, Personal, Full or part-time Employment: iOnctura. T. Lakshmikanth: Financial Interests, Personal, Stocks/Shares, Cofounder and shareholder in Cytodelics AB, Sweden with no remuneration: Cytodelics AB. M. Occhipinti: Financial Interests, Personal, Full or part-time Employment: Radiomics. P. Spiliopoulou: Financial Interests, Personal, Invited Speaker: Pfizer. T.R.J. Evans: Financial Interests, Institutional, Advisory Board, Advisory Board for GI cancers and melanoma (immune checkpoint inhibitors): Bristol Myers Squibb; Financial Interests, Institutional, Invited Speaker, Invited speaker - GI cancers, melanoma, immunotherapy: Bristol Myers Squibb; Financial Interests, Institutional, Advisory Board, Advisory Boards - GI cancers, melanoma: Roche / Genentech; Financial Interests, Institutional, Invited Speaker, Invited speaker GI cancer, melanoma: Roche / Genentech; Financial Interests, Institutional, Advisory Board, Advisory Board (Lenvatinib): Eisai; Financial Interests, Institutional, Invited Speaker, Speaker's fees (lenvatinib): Eisai; Financial Interests, Institutional, Advisory Board, advisory board: MSD, AstraZeneca, Bayer, Bicycle Therapeutics, Clovis; Financial Interests, Institutional, Invited Speaker, Speaker's fees: MSD, AstraZeneca, Bayer; Financial Interests, Institutional, Advisory Board, Advisory board: Nucana; Financial Interests, Institutional, Invited Speaker, speaker's fees: Nucana; Financial Interests, Institutional, Advisory Board, advisory board; Chair of Scientific Advisory Council (HCC & MIV-818): Medivir; Financial Interests, Institutional, Invited Speaker, speaker's fees (and presentation to potential investors): Medivir; Financial Interests, Personal, Other, Support to attend international conferences: Bristol Myers Squibb, Roche / Genentech, MSD, Nucana, Bayer, Celgene, Pierre Fabre; Financial Interests, Institutional, Advisory Board, Advisory Board for Upper GI Cancer: Ascelia; Financial Interests, Institutional, Advisory Board, Advisory Board for Oesophageal Cancer: Seagen; Financial Interests, Institutional, Coordinating PI, Educational grant (supply of study agents) for investigator-led study and reimbursement of study costs for commercial studies: AstraZeneca; Financial Interests, Institutional, Local PI, reimbursement of study costs for commercial studies: Astellas, Bayer, Basilea, Celgene, GSK, Roche, Medivir, Starpharma, Immunocore, Novartis, Sapience Therapeutics, MiNa Therapeutics, CytomX, Lilly, Bicycle Therapeutics, Sierra, BeiGene, Pfizer, Johnson & Johnson, UCB, Avacta, Codiak, Nurix, T3P; Financial Interests, Institutional, Coordinating PI, reimbursement of study costs for commercial studies: Adaptimmune, Bristol Myers Squibb, Eisai, MSD, Nucana, Sanofi, iOnctura; Financial Interests, Institutional, Local PI, support for non-commercial investigator-led study: Verastem; Financial Interests, Institutional, Local PI, reimbursement for costs of commercial studies: Boehringer Ingelheim; Financial Interests, Institutional, Local PI, reimbursement of costs of commercial study: Seagen; Non-Financial Interests, Personal, Member, Cancer Society Member: American Society of Clinical Oncology, America Association for Cancer Research, British Association for Cancer Research, Association of Cancer Physicians (UK), European Association for Cancer Research, International Liver Cancer Association; Non-Financial Interests, Personal, Other, Member of Scientific Advisory Panel: Pancreatic Cancer Research Fund; Non-Financial Interests, Personal, Other, Annual Meeting abstracts committee: International Liver Cancer Association; Non-Financial Interests, Institutional, Product Samples, Supply of investigational and licensed compounds for a non-commercial study for which I'm Chief Investigator: AstraZeneca; Other, Personal, Other, Editor-in-Chief: British Journal of Cancer; Other, Personal, Other, Chair of Independent Data Monitoring Committee for a phase 1 trial - honorarium payable to the employing institution: Genmab; Other, Personal, Other, Chair and panel member, Scientific Evaluation Committee: early phase trials (Amgen, Merck, AstraZeneca): Institut National du Cancer (France). M. Maio: Financial Interests, Personal, Advisory Board: BMS, Roche, GSK, Sanofi, Alfasigma, Amgen, Sciclone, Eli Lilly, MSD, Incyte, Pierre Fabre, AstraZeneca; Financial Interests, Personal, Stocks/Shares: Epigen, Theravance. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

132P - A phase I clinical trial of QLS31905 in advanced solid tumors

Presentation Number
132P
Lecture Time
05:00 - 05:00
Speakers
  • Yakun Wang (Beijing, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

QLS31905 is a novel Claudin18.2/CD3 bi-specific antibody. This study aimed to evaluate its safety, tolerability, and antitumor activity in advanced solid tumors.

Methods

This phase I trial (NCT05278832) recruited pts with advanced solid tumors who failed standard treatment, or were inapplicable to or had no standard treatment. The dose-escalation stage, adopting accelerated titration and interval 3+3 design, recruited pts regardless of Claudin 18.2 (CLDN18.2) expression. QLS31905 was administered in nine sequential single doses (0.5, 1.5, 5, 15, 45, 100, 200, 350, and 500 μg/kg qw or q2w) with/without priming dose. The dose-expansion stage recruited CLDN18.2-positive pts (expression ≥1% of tumor cells). The primary endpoint was dose limiting toxicities (DLT) and maximum tolerated dose (MTD) in dose-escalation stage, and was objective response rate (ORR) in dose-expansion stage.

Results

As of July 17, 2023, 52 pts were included. In dose-escalation stage, 22 pts were included from 0.5 to 350 μg/kg qw, and 500 μg/kg q2w cohort is ongoing. Two dose-expansion cohorts, 200 μg/kg qw or 350 μg/kg q2w, included 30 pts. Of 52 pts, 31 had gastric or gastro-esophageal junction cancer and 12 had pancreatic cancer (PC). DLT did not occur. MTD was not reached. Treatment-related adverse events (TRAEs) occurred in 51 pts (98.08%), of which 21 (40.38%) were ≥grade 3. Treatment-related serious adverse events (AEs) occurred in 10 (19.23%) pts. Two pts (3.85%) discontinued treatment due to AEs. The most common TRAE was fever (30, 57.69%), nausea (26, 50.00%), and decreased white blood cell count (18, 34.62%). Two pts had ≥grade 3 cytokine release syndrome, both in the 350 μg/kg qw cohort. Among pts in 200 μg/kg qw cohorts or above dose levels, ORR was 11.1% (3/27) and disease control rate (DCR) was 63.0% (17/27). Three pts with partial responses (2 in PC and 1 in gallbladder cancer) showed intermediate (≥40% of tumor cells) or high (≥70% of tumor cells) CLDN18.2 expression. Of 14 pts with SD (9 in gastric cancer, 4 in PC, and 1 in duodenal cancer), eight had target lesion reduction, among whom seven showed intermediate or high CLDN18.2 expression.

Conclusions

QLS31905 showed safety, tolerability, and preliminary antitumor activity in advanced solid tumors.

Clinical trial identification

NCT05278832.

Legal entity responsible for the study

Qilu Pharmaceutical Co., Ltd.

Funding

Qilu Pharmaceutical Co., Ltd.

Disclosure

Y. Li, L.Gu, L.Li, X.Kang: Other, Personal, Full or part-time Employment: Qilu Pharmaceutical. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

133P - Phase 1/2 study of XTX101, a masked, tumor-activated Fc-enhanced anti-CTLA-4, in patients with advanced solid tumors

Presentation Number
133P
Lecture Time
05:00 - 05:00
Speakers
  • Diwakar Davar (Pittsburgh, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

XTX101 is a tumor-activated, Fc-enhanced, high affinity anti-CTLA-4 monoclonal antibody designed with masking peptides that block the CTLA-4 antigen-binding regions. The masking peptides, covalently linked, are cleaved by proteases upregulated in the tumor microenvironment compared to healthy tissues. XTX101 contains 2 mutations that facilitate Fcγ receptor binding and improve effector function.

Methods

XTX101-01/02-001 (NCT04896697) Part 1A examined XTX101 in 3+3 dose escalation in patients (pts) with advanced solid tumors. Part 1B examined pharmacodynamic (PD) biomarkers.

Results

As of August 3, 2023, 27 pts were evaluable for safety, median age 67, median 4 prior lines of therapy (1-12). In Part 1A, 5 dose levels (DL) were evaluated. Among 6 pts treated at 60 mg every 3 weeks (Q3W), 1 dose limiting toxicity (DLT) of colitis was seen. Among 6 pts treated at 180 mg Q3W, 2 DLTs of colitis were seen. At 150 mg every 6 weeks (Q6W) there were no DLTs and this was chosen as the recommended regimen[CF1] . There were no grade (G) 4/5 treatment-related adverse events (TRAEs), and no endocrine or liver-related TRAEs on the study. Among 18 pts treated with XTX101 at Q3W schedule (DL 7-180 mg), G3 TRAEs were colitis (n = 4), infusion reaction (n = 3), lymphopenia/lymph count decreased (n = 2), and diarrhea (n = 1). In 9 pts treated at 150 mg Q6W, the only G3 TRAEs were diarrhea and dermatitis (1 each). XTX101 exposure at 150 mg Q6W was comparable to 2 cycles of 60 mg Q3W. One pt, at 150 mg Q6W, with PD-L1 negative non-small cell lung cancer had a confirmed partial response (PR) ongoing at 36 weeks, with resolution of hepatic metastases and only TRAE of G1 fatigue. Supporting tumor-selective activity of XTX101, this pt had minimal changes in peripheral PD markers. Data from tumor biopsies from 2 other pts showed >70% activated molecule in tumor vs 13% in plasma.

Conclusions

XTX101, an Fc-enhanced, tumor-activated, anti-CTLA-4, has a differentiated safety profile from systemically active anti-CTLA4 and showed evidence of monotherapy clinical activity with a confirmed PR. Part 1B is ongoing at 150 mg Q6W. Development of XTX101 is planned in combination with atezolizumab for pts with microsatellite stable colorectal cancer.

Clinical trial identification

NCT04896697.

Legal entity responsible for the study

Xilio Development, Inc.

Funding

Xilio Therapeutics, Inc.

Disclosure

D. Davar: Financial Interests, Institutional, Research Grant: Arcus, CellSight Technologies, Immunocore, Merck, Regeneron Pharmaceuticals Inc., Tesaro/GSK; Financial Interests, Personal, Speaker, Consultant, Advisor: ACM Bio, Ascendis Pharma, Clinical Care Options (CCO), Gerson Lehrman Group (GLG), Merck, Medical Learning Group (MLG), Xilio Therapeutics Inc.; Financial Interests, Personal, Speaker’s Bureau: Castle Bioscience; Other, Personal, Other, US Patent 63/124,231: University of Pittsburgh Cancer Institute; Other, Personal, Officer, US Patent 63/208,719: University of Pittsburgh Cancer Institute. J.G. Knecht: Financial Interests, Personal, Full or part-time Employment: Cardinal Health, CLS Health, Incyte; Financial Interests, Personal, Leadership Role: Kindred Hospital Clear Lake; Financial Interests, Personal, Stocks/Shares: Beckton Dickinson, Checkpoint Therapeutics, Fortress Biotech, Myovant Sciences, Reviva Pharmaceuticals; Financial Interests, Personal, Advisory Role, Honoraria: OMNI Health Media; Financial Interests, Personal, Research Funding: Atossa Therapeutics, Cosmo Pharmaceuticals, EQRx, Phanes Therapeutics, Takeda, Xilio Therapeutics, Inc.; Financial Interests, Personal, Other, Travel, Accommodations, Expenses: Cardinal Health. A.L. Vandross: Financial Interests, Personal, Full or part-time Employment: Next Oncology, Texas Oncology; Financial Interests, Institutional, Research Funding: AbbVie, Ascentage Pharma, BiOneCure, Chugai Pharma, Deciphera, Exelixis, Immunomedics, Lyvgen Biopharma, Medikine, Nanjing, NGM Biopharmaceuticals, Nitto BioPharma, OncoResponse, PMV Pharma, Sirnaomics, Tachyon Therapeutics, Teon Therapeutics, Xilio Therapeutics, Inc. C.A. Perez: Financial Interests, Personal, Advisory Board: EMD Serono; Financial Interests, Personal, Other, Steering Committee: Kinnate Biopharma Inc., BeiGene; Financial Interests, Institutional, Local PI: Accutar Biotech, Kinnate Biopharma, Relay Therapeutics, Seagen Inc, Kura Oncology, Hyamab Inc, Xilio Therapeutics, Elucida Oncology, Tallac Therapeutics, Ribbon Therapeutics, Mirati Therpeutics, Elpiscience Biopharmaceuticals, Dracen Pharmaceuticals, Zhuhai Yufan Biotechnologies Co., Genentech, Inc., Jazz Pharmaceuticals, Artios Pharma, Ayala Pharmaceuticals, Elevation Oncology, Merus. D.M. Miller: Financial Interests, Personal, Stocks or ownership: Checkpoint Therapeutics; Financial Interests, Personal, Advisory Role: Castle Biosciences, Checkpoint Therapeutics, Merck Sharp & Dohme, Pfizer/EMD Serono, Regeneron, Sanofi/Regeneron; Financial Interests, Personal, Research Funding: Incyte; Financial Interests, Personal, Other, Travel, Accommodations, Expenses: Merck Sharp & Dohm, Pfizer, Regeneron, Sanofi/Aventis. J. Powderly: Financial Interests, Personal, Other, Consulting: Boxer Capital; Financial Interests, Personal, Writing Engagement, Consulting: Aavocyte; Financial Interests, Personal, Member of Board of Directors, Founder and Owner: Carolina BioOncology Institute, PLLC, BioCytics Inc.; Financial Interests, Personal, Ownership Interest, Founder and Owner: BioCytics Inc.; Financial Interests, Personal, Ownership Interest, Founder and Owner of phase 1 cancer research clinic.: Carolina BioOncology Institute, PLLC; Financial Interests, Personal, Other, Founder and Owner, developing intellectual property for cellular therapies: BioCytics Inc; Financial Interests, Personal and Institutional, Local PI: Bristol Myers Squibb, Cullinan, Genentech/Roche, AstraZeneca/MedImmune, EMD Serono, Macrogenics, InCyte, Top Alliance BioSciience, Seattle Genetics, AbbVie, FLX Bio, Alkermes, Arcus BioSciences, Tempest Therapeutics, Calico Life Sciences, Apros, Jounce Therapeutics, Atreca, Sequenom, Repertoire Immune Medicines, Molecular Templates, I-MAB Pharma, NexCure, Xilio Therapeutics, Immune-Onc, Trethera, Zenshine Pharma, Adagene, BJ BioScience, Fate Therapeutics, Conjupro BioTherapeutics, PEEL Therapeutics, CUE BioPharma, Pieris Pharmaceuticals, RiboScience, Moderna TX, Simcere, Phanes Therapeutics, SK Life Science, Harbour BioMed, Allarity, Aulos, GI Innovation, IGM BioSciences, Aptevo, Medikine, IconOVir Bio, Qurgen; Financial Interests, Institutional, Funding: Precision for Medicine, MT Group, Stemcell Technologies, Replimmune, Merck, Xilis; Financial Interests, Personal and Institutional, Funding: Pioma; Financial Interests, Personal and Institutional, Local PI, Also funding for contract laboratory services: Nuvation; Financial Interests, Personal and Institutional, Funding, Wugen is sponsor of contract laboratory translational research: Wugen; Financial Interests, Personal and Institutional, Other, AavoCyte & AavoBioCytics are jointly developing cellular therapies with BioCytics Human Applications Lab for point of care manufacturing: AavoCyte; Other, Personal, Other, As Founder and Owner of BioCytics Inc. developing immune cellular therapy.: BioCytics Inc. K. Montazeri: Financial Interests, Personal, Advisory Role, Consultant: Immunocore. M.J. Mooradian: Financial Interests, Personal, Advisory Role, Consultant/Honorarium: AstraZeneca, Bristol Myers Squibb, Regeneron Pharmaceuticals; Financial Interests, Personal, Research Funding: Foundation Medicine. A. Gupta: Financial Interests, Personal, Full or part-time Employment: Xilio Therapeutics, Inc.; Financial Interests, Personal, Stocks/Shares: Xilio Therapeutics, Inc. E. Patel: Financial Interests, Personal, Full or part-time Employment: Xilio Therapeutics, Inc.; Financial Interests, Personal, Stocks/Shares: Xilio Therapeutics, Inc.; Non-Financial Interests, Personal, Other, US Patent: TCR2 Therapeutics; Non-Financial Interests, Personal, Officer, US Patent: Mustang Bio. D. Fantini: Financial Interests, Personal, Full or part-time Employment: Xilio Therapeutics, Inc., Merck, ELi Lilly and Company; Financial Interests, Personal, Stocks/Shares: Xilio Therapeutics, Inc. , Merck, Eli Lilly and Company. S. Paramasivan: Financial Interests, Personal, Full or part-time Employment: Xilio Therapeutics, Inc.; Financial Interests, Personal, Stocks/Shares: Xilio Therapeutics, Inc., ADMA Biologics, Allogene Therapeutics, Amylyx Pharmaceuticals, Avadel Pharmaceuticals, Aurinia Pharmaceuticals, Iovance Therapeutics, Voyager Therapeutics, Deciphera Pharmaceuticals, Baudax Bio Inc., CRISPR Therapeutics, GRI Bio Inc., GSK, Novartis, Roche, Selecta Biosciences. D. Crowe: Financial Interests, Personal, Stocks/Shares: Xilio Therapeutics Inc. M. Duncan, S. Uptain, K. Luptakova: Financial Interests, Personal, Full or part-time Employment: Xilio Therapeutics, Inc.; Financial Interests, Personal, Stocks/Shares: Xilio Therapeutics, Inc. R. Sullivan: Financial Interests, Personal, Research Funding: Merck; Financial Interests, Personal, Advisory Board, Consulting: BMS, Marengo, Merck, Novartis, Pfizer, Replimune. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

134P - A Phase 1 Study Exploring the Safety and Tolerability of the Small Molecule PD-L1 Inhibitor INCB099318 in Select Advanced Solid Tumors

Presentation Number
134P
Lecture Time
05:00 - 05:00
Speakers
  • David J. Pinato (London, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

INCB099318, an oral, programmed death ligand-1 (PD-L1) inhibitor, has shown preliminary efficacy and acceptable safety in an ongoing, Phase 1, open-label, multicenter study in patients with advanced solid tumors (Pinato, et al. SITC 2022). Here we present updated study results.

Methods

Eligible patients were ≥18 years with ECOG PS ≤1, who had disease progression after available treatment or were ineligible for/without access to standard treatment. In Part 1, doses were escalated from 100 mg BID using a Bayesian optimal interval design. Select doses were to be expanded in Part 2. Primary endpoints are INCB099318 safety, tolerability, and determination of pharmacologically active/maximum tolerated dose (MTD). Secondary and exploratory endpoints include pharmacokinetics, objective response rate per RECIST v1.1, and biomarkers of pharmacologic activity.

Safety

TEAEs, n (%) Total patients (N=101)
Any grade Grade ≥3
All cause TEAEs 94 (93.1) 41 (40.6)
Occurring in >10% of patients
Fatigue 33 (32.7) 4 (4.0)
Constipation 24 (23.8) 0
Nausea 23 (22.8) 2 (2.0)
Anemia 17 (16.8) 9 (8.9)
Decreased appetite 15 (14.9) 2 (2.0)
Diarrhea 15 (14.9) 2 (2.0)
Cough 13 (12.9) 0
Dyspnea 13 (12.9) 1 (1.0)
Vomiting 12 (11.9) 1 (1.0)
Alanine aminotransferase increased 11 (10.9) 2 (2.0)
Immune-related TEAEs 11 (10.9) 4 (4.0)
Serious TEAEs 25 (24.8)
Occurring in >1 patient
Anemia 2 (2.0)
Cerebrovascular accident 2 (2.0)
Grade ≥3 Treatment-related TEAEs 11 (10.9)
Occurring in >1 patient
Fatigue 3 (3.0)
Anemia 2 (2.0)

Results

As of June 23, 2023, 101 patients had received INCB099318 at doses from 100 to 800 mg QD or BID (Part 1, n=64; Part 2, n=37). Median age was 58 years (range, 29-89), 59.4% were women, 86.1% were White, 68.3% had ≥2 prior lines of treatment, and 9.9% had prior IO. Most common tumor types were cervical (17.8%), ovarian (10.9%), and colorectal (5.9%). No dose-limiting toxicities were observed and MTD was not reached. In Part 2, 3 dose levels were expanded (400 mg BID, n=18; 800 mg BID n=14, 800 mg QD, n=5). Overall, 82 patients (81.2%) discontinued treatment, 69 (68.3%) due to disease progression. 94 (93.1%) patients had treatment-emergent adverse events (TEAEs) (Table). Several responses have been observed, and updated results will be presented.

Conclusions

INCB099318 was generally well tolerated at all doses tested. Preliminary safety and response outcomes support future development of INCB099318 for the treatment of select advanced solid tumors.

Clinical trial identification

NCT04272034.

Editorial acknowledgement

Editorial assistance was provided by Emily Sun and Andrew Marson-Neep of Envision Pharma Group (Philadelphia PA, USA).

Legal entity responsible for the study

Incyte Corporation, Wilmington, DE.

Funding

Incyte Corporation, Wilmington, DE.

Disclosure

D.J. Pinato: Financial Interests, Personal, Other, travel grant and advisory/speaker fees: AstraZeneca, Avammune, Bayer, Bristol Myers Squibb, Da Volterra, Eisai, Falk Pharma, H3 Biomedicine, Ipsen, Lift Biosciences, Mina Therapeutics, MSD Oncology, Mursla Bio, Roche, Roche/Genentech, Starpharma, ViiV Healthcare; Financial Interests, Institutional, Research Funding: Bristol Myers Squibb, GSK, MSD Oncology; Financial Interests, Personal, Other: Wiley. U. Banerji: Financial Interests, Personal, Other, travel grant and advisory fees: Bayer, Boehringer Ingelheim, Janssen, Pegascy, Sierra Oncology; Financial Interests, Institutional, Research Funding: AstraZeneca, BTG, Carrick Therapeutics, Chugai Pharma, Onyx, Verastem. S. Rottey: Financial Interests, Personal, Other, travel grants and advisory/speaker fees: Novartis, Roche, Pfizer, MSD, Bristol Myers Squibb, Ipsen; Financial Interests, Personal, Research Grant: Roche, MSD. K. Peltola: Financial Interests, Personal, Advisory Board: MSD, Ipsen, Roche, BMS, Pfizer, Lilly, Novartis, Bayer; Financial Interests, Personal, Stocks/Shares: Faron Pharmaceuticals; Financial Interests, Institutional, Local PI, Conduct of sponsored clinical trial: Novartis; Financial Interests, Institutional, Local PI, Sponsored clinical trial: Exelixis; Financial Interests, Institutional, Local PI, Several clinical trials: BMS, MSD, Roche; Financial Interests, Institutional, Local PI, clinical trials: Incyte; Financial Interests, Institutional, Local PI, Conduct of clinical trials: Pfizer; Financial Interests, Institutional, Local PI, Conduct of clinical trial: Bayer. R. Kristeleit: Financial Interests, Personal, Other, Honoraria: AstraZeneca, Clovis Oncology, GSK and Incyte ; Financial Interests, Personal, Other, Travel grant: AstraZeneca, GSK and Sierra Oncology ; Financial Interests, Personal, Research Grant: MSD; Financial Interests, Personal, Speaker, Consultant, Advisor: Basilea Pharmaceutica and Shattuck Pharma. M. Gutierrez: Financial Interests, Personal, Other, travel grand and speaker fees: BMS, Guardant Health, Lilly, Merck; Financial Interests, Personal, Stocks or ownership: Cota Healthcare; Financial Interests, Institutional, Research Funding: Acerta Pharma, Adlai Nortye, Arcus Biosciences, Array BioPharma, Bayer, Bellicum Pharmaceuticals, BMS, Boehringer Ingelheim, Celgene, Checkpoint Therapeutics, Compass Therapeutics, Constellation Pharmaceuticals, Cullinan Oncology, Cyteir, Daiichi Sankyo. S.N. Symeonides: Financial Interests, Institutional, Speaker, Consultant, Advisor: Bicycle Therapeutics, Boxer Capital, Bristol Myers Squibb, Duke Street Bio, Eisai, Ellipses Pharma, Eugit Therapeutics, EUSA Pharma, Exscientia, Ipsen, MedAnnex, MSD, Pfizer/EMD Serono, Vaccitech; Financial Interests, Personal, Other, Travel Grant: BioNTech, Bristol Myers Squibb, EUSA Pharma, Ipsen, MSD; Financial Interests, Institutional, Research Funding: MSD and Verastem. M. Hoejgaard: Financial Interests, Institutional, Research Funding: Puma Biotechnology, Roche/Genentech, AstraZeneca, Incyte, Pfizer, Orion Pharma, MSD, Merck, Bristol Myers Squibb, Novartis, Lilly Pharmaceuticals/ Loxo Oncology, Bayer/Loxo Oncology, Amgen, Repare Therapeutics, Genmab, Kinnate Biopharma; Financial Interests, Personal, Stocks/Shares: Bavarian Nordic, Agilent, Illumina, Pacific Biosciences; Financial Interests, Personal, Other, Board Member: Danish Medicines Council. V. Ebiana, J. Daniel, J. Pulini: Financial Interests, Personal, Full or part-time Employment: Incyte Corporation; Financial Interests, Personal, Stocks or ownership: Incyte Corporation. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

135P - Isunakinra as Monotherapy and Combined with Nivolumab for Treatment Resistant Advanced Solid Tumours: Exploratory Effect Data, Tolerability, and Pharmacokinetics from a Dose Escalation Trial

Presentation Number
135P
Lecture Time
05:00 - 05:00
Speakers
  • Carlos Becerra (Dallas, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Inflammatory signalling by interleukin-1 receptor (IL-1R) family members promote neoplastic transformation, tumour growth, dissemination, and supportive cell recruitment. IL-1R1-inhibition is tumor suppressive and optimizes PD-1-inhibition in animals. Isunakinra, an anakinra-like IL-1R1 antagonist, blocks IL-1α/IL-1β 10-20x more potently in various assays. A safety, dose escalation trial with exploratory endpoints of isunakinra w/wo nivolumab in patients with advanced solid tumors is presented.

Methods

15 subjects recruited to escalating dose groups (3+3 per Dose-Limiting Toxicity criteria) with daily isunakinra SC at 15, 25 or 50 mg for 3 weeks was followed by combination treatment with flat dose nivolumab every 4 weeks for up to 28 weeks.

Results

15 patients accrued to the study, exposure totalling 1.584 doses. No DLT occurred, and no PD-1 immune reactions developed. Five mild, transient injection site reactions were reported. Ten serious events (hospitalizations) in 7 subjects were related to tumor progression. Decrease in circulating tumor cells: 35% (N=13) at week 4, 53% (N=10) at week 8 and 80% (N=5) at week 12. Best Overall Response (BoR) per RECIST 1.1 and ( irRC) assessable in 12 subjects: SD in 10 (8), PR in 1 (2), and PD in 1(2). Four subjects completed the trial with SD/SD or SD/PR. One subject (colon cancer, k-ras mut, MSS) with a pelvic mass close to CR at W28 with PR /PR granted extended study therapy continued to W64 without safety issues and the target lesion stable at 16 mm from W44. Baseline MDSC compositions varied at baseline. Isunakinra alone (W1 - W4) clearly impacted key naïve cell types in all patients. With nivolumab added (W4), equally clear but often contrasting effects were observed at W8 and W12. Mature cells were unaffected.

Conclusions

The RP2D for isunakinra + nivolumab is 50 mg SC per day. No serious events related to the combination therapy. Encouraging exploratory data with signs of tumour control in heavily pre-treated subjects along with currently obtained pharmacokinetic and safety profiles justifies exploring further CPI combinations in planned phase 2 studies.

Clinical trial identification

NCT04121442.

Legal entity responsible for the study

Buzzard Pharmaceuticals AB Buzzard Pharmaceuticals AB.

Funding

Buzzard Pharmaceuticals.

Disclosure

C. Becerra: Non-Financial Interests, Institutional, Principal Investigator: Buzzard Pharmaceuticals. H. Olivecrona: Non-Financial Interests, Personal, Coordinating PI: Buzzard Pharmaceuticals. M. de Château: Financial Interests, Institutional, Full or part-time Employment: Buzzard Pharmaceuticals. S.A. Paulson: Non-Financial Interests, Institutional, Local PI: Buzzard Pharmaceuticals.

Collapse
Poster Display (ID 34) Poster Display

136P - Circadian rhythm positioned chronomodulated-SBRT with Cancer associated fibroblast elimination theranostic treatment to harmonize resistant TFG-Beta stromal microenvironment in conjunction with PDL-1 based immunotherapy in disseminated cancers-Concept randomised study.

Presentation Number
136P
Lecture Time
05:00 - 05:00
Speakers
  • G Lohith (Gulbarga, India)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Intrinsic links of variation in immune function and phases of circadian rhythm is the basis of chronotherapy. Modulating immune potentiating nature of modern anti-cancer treatment combinations such as PDL1 inhibitors, Stereotactic body radiotherapy (SBRT), stromal targeted theranostic using chronobiology is an unfathomed area of research. SBRT releases tumour neo-antigens, the anti PDL1 therapy aids in effective cytotoxic T cell activity and anti-stromal theranostic modulates the resistant tumour stroma with an anti TGF Beta activity in an immune favouring way. Chrono-phased SBRT -Timing the SBRT at the best immune functioning part of the day could be an effective synergism to the amalgam.

Methods

26 disseminated cancer patients eligible for anti-PDL1 drug were treated with Actinium-225 tagged fibroblast activated protein (FAP) based Alpha theranostic to target the cancer associated fibroblasts (CAFs) forming the tumor stroma and were randomized to receive SBRT at any given part of the day versus a chrono-phased SBRT-7am to 9am after verifying the circadian rhythm.Intactness of the circadian rhythm in these patients were assessed using sleep log. Baseline cytokine profile, WBC, ALC and NL ratio were compared to immediate therapy level and post 3-months. Response to therapy was radiologically assessed using RECIST 1.1 criteria.

Results

Response Regular SBRT (13 cases) Chrono-SBRT (13 cases)
Partial (PR) 3 4
Complete (CR) 0 1
Progression (PD) 2 1
Mixed Type-1 (PR+CR) 3 5
Mixed Type-2 (PD+PR) 5 2

Conclusions

While augmenting immune responses to cancer immune-modulatory therapies is an area of active probing, using the immune sensitive phase of circadian rhythm to obtain therapeutic benefit seams beneficial. Our study suggests that the technique is feasible, and our early results are encouraging with better quality of life with chrono-sbrt.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

137P - First-in-human results from a Phase I dose-escalation study of VSV-GP (BI 1831169) in patients with advanced solid tumors

Presentation Number
137P
Lecture Time
05:00 - 05:00
Speakers
  • Stephane Champiat (Villejuif, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

VSV-GP (BI 1831169) is a selective oncolytic virus that can mediate potent oncolysis in many solid cancer cell types and initiate adaptive antitumor immunity in animal models. Here we report the preliminary first-in-human results of VSV-GP monotherapy administered intratumorally (IT) in patients with advanced solid tumors.

Methods

In this phase I, open-label, dose-escalation study, VSV-GP is administered IT, intravenously (IV) or IT+IV as monotherapy (Part 1) or with anti-programmed cell death 1 compound ezabenlimab (Part 2). VSV-GP is given on Days 1 and 4 of Cycle 1 and Day 1 of Cycles 2–4 (21-day cycles). Dose finding is guided by the Bayesian Optimal Interval design. The primary endpoint is the number of patients with dose-limiting toxicities (DLTs) during the maximum tolerated dose (MTD) evaluation period in order to determine the MTD and/or recommended Phase II dose (RP2D). Further safety, efficacy, pharmacokinetics, shedding and immunogenicity results will be evaluated.

Results

As of August 2, 2023, eight patients have received VSV-GP monotherapy IT, four at dose level (DL) 1 (5 x 107 TCID50) and four at DL2 (5 x 108 TCID50). At DL1, all patients discontinued treatment (progressive disease n=3, unrelated Grade [G] 3 E. coli sepsis n=1). A G1 classified cytokine release syndrome (presenting as fever and low oxygen saturation) was reported in one patient at DL2 following the first dose. The maximum number of cycles received was three. At DL2, one patient discontinued treatment due to related G3 neutropenia after the first administration and three patients are ongoing. One per protocol DLT (G3 fatigue) was reported at DL2 and three additional patients will be recruited. The most frequent related adverse events reported were pyrexia, chills and decreased lymphocytes (Table).

Overall summary of adverse events (AEs), n (%) DL1 (n=4) DL2 (n=4) Total (n=8)
Any AE 4 (100) 4 (100) 8 (100)
AEs leading to treatment discontinuation 1 (25) 1 (25) 2 (25)
Patients with serious AEs 1 (25) 2 (50) 3 (37.5)
Treatment-related AEs 3 (75) 4 (100) 7 (87.5)
Treatment-related AEs occurring in ≥30% of patients
Pyrexia 2 (50) 2 (50) 4 (50)
Chills 1 (25) 2 (50) 3 (37.5)
Decreased lymphocyte count 2 (50) 1 (25) 3 (37.5)

Conclusions

The study is ongoing to evaluate the safety and determine the RP2D of VSV-GP administered IT, IV and IT+IV as monotherapy and in combination with ezabenlimab.

Clinical trial identification

NCT05155332.

Editorial acknowledgement

Katerina Douka, PhD and Devon Else, BSc, of MediTech Media provided writing and editorial support which was contracted by Boehringer Ingelheim International GmbH (BI). BI was given the opportunity to review the abstract for medical and scientific accuracy as well as intellectual property considerations.

Legal entity responsible for the study

Boehringer Ingelheim International GmbH.

Funding

Boehringer Ingelheim International GmbH.

Disclosure

S. Champiat: Financial Interests, Personal, Principal Investigator: AbbVie, Amgen, Boehringer Ingelheim, Cytovation, Eisai, GSK, Imcheck Therapeutics, Immunocore, Molecular Partners Ag, MSD, Ose Immunotherapeutics, Pierre Fabre, Replimune, Roche, Sanofi Aventis, Seagen, Sotio A.S, Transgene; Financial Interests, Personal, Advisory Board: Alderaan Biotechnology, Amgen, AstraZeneca, Avacta, BioNTech, Celanese, Domain Therapeutics, Ellipses Pharma, Genmab, Immunicom, Inc., Nanobiotix, Nextcure, Oncovita, Pierre Fabre, Seagen, Takeda, Tatum Bioscience, Tollys, UltraHuman8; Financial Interests, Personal, Other, Travel and congress: Amgen, AstraZeneca, Bristol Myers Squibb, MSD, Ose Immunotherapeutics, Roche, Sotio; Financial Interests, Personal, Other, Honoraria: Amgen, Astellas, AstraZeneca, Bristol Myers Squibb, Eisai, Genmab, Janssen, Merck KGaA, MSD, Novartis, Roche and Servier; Financial Interests, Institutional, Principal Investigator, As part of the Drug Development Department (DITEP): AbbVie, Adaptimmune, Adlai Nortye USA Inc, Aduro Biotech, Agios Pharmaceuticals, Amgen, Astex Pharmaceuticals, AstraZeneca Ab, Aveo, Basilea Pharmaceutica International Ltd, Bayer Healthcare Ag, Bbb Technologies Bv, BeiGene, BicycleTx Ltd, Blueprint Medi; Financial Interests, Institutional, Research Grant, As part of the Drug Development Department (DITEP): AstraZeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Pfizer, Roche, Sanofi; Non-Financial Interests, Institutional, Non-financial benefits, Drug supplied: AstraZeneca, BMS, Boehringer Ingelheim, GSK, MedImmune, Merck, NH TherAGuiX, Pfizer, Roche. A. Italiano: Financial Interests, Personal, Advisory Board: Bayer, Roche, Philips, Chugai, GSK; Financial Interests, Institutional, Coordinating PI: Bayer, AstraZeneca, Roche, MSD, Ipsen, Merck. N. Wagle: Financial Interests, Institutional, Research Funding: Bavarian Nordic, Bayer, Biocept, Boehringer Ingelheim, Caris MPI, CNS Pharmaceuticals, EpicentRx, Novocure, Oblato, Pyramid Biosciences, Stemedica Cell Technologies, xCures, Xoft. U.M. Lauer: Financial Interests, Institutional, Research Funding: Boehringer Ingelheim; Financial Interests, Institutional, Research Grant: Boehringer Ingelheim; Financial Interests, Personal, Speaker, Consultant, Advisor: Boehringer Ingelheim, Abalos Therapeutics; Financial Interests, Personal, Advisory Board: Asgard Therapeutics. A. Quinson, S. Luecke, V. Hern: Financial Interests, Personal, Full or part-time Employment: Boehringer Ingelheim. M. Porosnicu: Financial Interests, Personal, Speaker, Consultant, Advisor: Boehringer Ingelheim; Financial Interests, Personal, Research Funding: Boehringer Ingelheim, AstraZeneca, Eli Lilly, Astellas, Sanofi-Aventis.

Collapse
Poster Display (ID 34) Poster Display

138P - Generation of frameshift mutated TGF_R2-specific T cells in healthy subjects following administration with cancer vaccine candidate FMPV-1/GM-CSF

Presentation Number
138P
Lecture Time
05:00 - 05:00
Speakers
  • Else M. Inderberg (Oslo, Norway)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Generation of specific and durable T cell response is the basis of antigen targeted cancer vaccine strategies. A study in healthy male subjects conducted over a one-year period investigated the generation of antigen specific T cell responses from a peptide vaccine FMPV-1, targeting frameshift mutated (mut) transforming growth factor receptor 2 (TGFβR2), commonly occurring in cancers with microsatellite instability (MSI-H). Granulocyte macrophage colony stimulating factor (GM-CSF) was used as adjuvant.

Methods

An open label study was conducted in 16 healthy male subjects who received intradermal FMPV-1 (0.15 mg/injection) preceded by GM-CSF (0.03mg/injection) on Days 1, 8, 16, 29 and 43. Immune responses were assessed by in vivo delayed type hypersensitivity (DTH) in all subjects on Days 1, 29 and 43 and in 7 subjects after 6 months, and in vitro assessment of induced FMPV-1 antigen specific T cells at Days 1, 57, 80, 6 months and 12 months, with Stimulation Index (SI) values ≥ 2 considered positive. Plasma samples were assayed for the presence of mut (A9) TGFβR2 circulating cell-free DNA (cfDNA) fragments and wildtype (wt) (A10) TGFβR2 cfDNA fragments. Subjects were assessed for safety and tolerability.

Results

None of the 16 subjects had a positive DTH before vaccination. After 3 and 4 vaccinations, 8/16 and 15/16 subjects had a positive DTH response respectively. At 6 months 5/7 subjects still had a positive DTH response. FMPV-1 antigen-specific T cells were detected shortly after the vaccination period and at 6 and 12 months. Neither mut nor wt TGFβR2 cfDNA fragments were detected in any subject. Vaccination with FMPV-1/GM-CSF was well tolerated out to 1 year following initial dosing.

Conclusions

The FMPV-1 peptide vaccine is immunogenic and induces frameshift mutant TGFβR2 specific T cells after a short vaccination period in healthy subjects. This provides a rationale for FMPV-1 treatment of MSI-H cancer patients in combination with immune checkpoint inhibitors. With the favourable safety profile and ability to induce frameshift mut TGFβR2 specific memory T cells, FMPV-1/GM-CSF may play an important role in both therapeutic and prophylactic settings of cancer.

Clinical trial identification

EudraCT 2020-004363-80.

Legal entity responsible for the study

Quotient Sciences.

Funding

Hubro Therapeutics AS.

Disclosure

S.J. Arbe-Barnes: Financial Interests, Personal, Full or part-time Employment: Hubro Therapeutics AS. J.A. Eriksen: Financial Interests, Personal and Institutional, Stocks/Shares: Hubro Therapeutics AS; Financial Interests, Personal, Officer: Hubro Therapeutics AS. B. Iverson, H. Kvalheim, R.M. Miller, K. Risberg Handeland: Financial Interests, Institutional, Full or part-time Employment: Hubro Therapeutics AS. N. Singh: Other, Institutional, Local PI: Quotient Sciences. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

139P - Safety and clinical activity of a novel anti-CCR8 antibody (LM-108) as a single agent or in combination with pembrolizumab in patients with advanced solid tumors: Results of phase 1 study

Presentation Number
139P
Lecture Time
05:00 - 05:00
Speakers
  • Alexander Starodub (Cincinnati, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Tumor-infiltrating regulatory T cells (TITR) specifically demonstrate high expression of chemokine receptor 8 (CCR8). LM-108 is an anti-CCR8 monoclonal antibody that deplete TITR cells with enhanced antibody-dependent cell-mediated cytotoxicity. LM-108 alone or in combination with anti-PD-1 antibody has demonstrated excellent activity and safety profile in animal models. This phase 1/2 study (NCT05255484) evaluate LM-108 as single agent or in combination with pembrolizumab in patients with advanced solid tumor. Here, we present the results of phase 1 part.

Methods

This first-in-human, multicenter, open-label, phase 1/2 study enrolled patients aged ≥ 18 years with advanced solid tumors who had progressed on standard therapy, or were intolerable to the available standard therapy, or had no available standard therapy for treatment. For monotherapy, LM-108 were administered intravenously Q3W at dose levels of 0.3 mg/kg, 1 mg/kg, 3 mg/kg, 10 mg/kg, and 20 mg/kg as per traditional 3+3 design. Combination therapy was evaluated at dose level of 10 mg/kg of LM-108 combined with 200 mg pembrolizumab Q3W.

Results

As of July 3, 2023, 24 patients (21 from monotherapy and 3 from combination therapy) were enrolled, no DLT was observed at all dose levels in monotherapy or combination therapy. MTD was not reached. The most frequent adverse events (AEs) related to the study drug were rash (8/24, Grade 1-2) and diarrhea (3/24, including 1 Grade 3). Out of 19 patients from monotherapy, 1 patient achieved partial response (PR) and 11 patients achieved stable disease (SD) with a disease control rate of 63%. One patient achieved PR and one patient achieved SD out of 2 evaluable patients from combination group. The two PR patients had duration of response over 6 months.

Conclusions

LM-108 monotherapy or in combination with pembrolizumab showed excellent safety profile and promising anti-tumor activity in patients with advanced solid tumors.

Legal entity responsible for the study

LaNova Medicines Limited.

Funding

LaNova Medicines Limited.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

140P - Eliciting mAbs targeting MHC-bound peptides with a novel antibody discovery platform

Presentation Number
140P
Lecture Time
05:00 - 05:00
Speakers
  • Elli Sandberg (Heidelberg, Germany)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The cell membrane of the trypanosomiasis-causing parasite Trypanosoma brucei is densely coated with 10 million copies of Variant Surface Glycoproteins (VSGs). VSG is highly immunogenic and can elicit a robust and long-lasting humoral immune response from the host organism.

Methods

Our laboratory has adapted the T. brucei coat into a uniquely powerful immunization platform (VSG-immunogen Array by Sortase Tagging) that has the capacity to elicit antibody responses against previously untargetable antigen classes. The antigens of interest are covalently coupled to the VSG proteins on the membrane by a sortase reaction. Sortase is a bacterial enzyme that can covalently link any polypeptides to each other, provided that they contain the sortase recognition peptides. We have engineered trypanosomes whose VSGs bear the N-terminal sortase recognition motif, and we generate antigens that display the cognate C-terminal motif. This platform has elicited high-affinity monoclonal antibodies against several immunogens of interest, including small molecules and peptides.

Results

A current obstacle in the treatment of many diseases is the difficulty of targeting proteins within the target cells. TCR-like monoclonal antibodies (TCR-like-mAbs) present a unique solution by targeting MHC receptors presenting such intracellular proteins. Currently, we are developing antibodies against intracellular protein targets that are presented on MHC after processing by the proteasome machinery, for example the MART-1 peptide. MART-1 is a protein expressed in metastatic melanoma cells. The protein is further processed by proteasome machinery, leading to a stretch of peptide from amino acids 27-35 (EAAGIGILTV) being presented on the cell surface via MHC class I.

Conclusions

To target peptides such as MART-1, we are using the VAST platform to elicit high-affinity TCR-like antibodies that can recognize peptides within the context of MHC, but in an MHC agnostic manner.

Legal entity responsible for the study

Panosome GmbH, Deutsches Krebsforschungszentrum.

Funding

Panosome GmbH.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

141P - An IgE antibody targeting the melanoma-associated Chondroitin Sulfate Proteoglycan 4

Presentation Number
141P
Lecture Time
05:00 - 05:00
Speakers
  • Lais Cristina G. Palhares (London, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

In antibody-mediated oncotherapy, IgG monoclonal antibodies have been the class of choice for the treatment of different cancers. However, tumour-antigen specific IgE antibodies are emerging as a new avenue for cancer therapy. This class of antibody have a high affinity for cognate Fcε receptors expressed on tumour-resident immune cells, such as macrophages and mast cells, and a lack of inhibitory Fc receptors, offering the chance to activate anti-tumour responses in tissues. Recently, a tolerable safety profile and anti-tumour efficacy of an IgE antibody recognizing the tumour-associated antigen, folate receptor α (FRα), has been shown in the first-in-class clinical trial of this agent.

Methods

We engineered a monoclonal IgE antibody, with human constant domains, recognising CSPG4 to target human melanoma. In vitro Fab-mediated antitumour effects, and mediated antibody-dependent cellular cytotoxicity (ADCC) against melanoma cells by immune effector cells were evaluated. CSPG4 antigen may be cleaved and released from the surface of tumour cells, and may bind to anti-CSPG4 antibodies, preventing them from engaging tumour cells and impairing their anti-tumour efficacy; or complexes of shed CSPG4 may cross-link anti-CSPG4 IgE engaged with basophils, which could potentiate degranulation and type I hypersensitivity. CSPG4 antigen shedding was measured in melanoma cell supernatant and in human sera. As an early evaluation of safety, we tested the propensity for CSPG4 IgE to induce RBL-SX38 degranulation in the presence of melanoma patient sera, and in unfractionated patient blood in basophil activation tests (BAT).

Results

CSPG4 IgE bound to melanoma tumours, induced Fab-mediated antitumour effects, and mediated ADCC against melanoma cells. We measured minimal shedding of the CSPG4 antigen in melanoma cell supernatants. Low levels of CSPG4 were detected in the circulation of melanoma patients and healthy subjects, with no significant difference in levels between the groups. CSPG4 IgE did not trigger RBL-SX38 degranulation and did not activate basophils in the BAT assay, suggesting lack of propensity to trigger anaphylaxis.

Conclusions

Our findings suggest that CSPG4 IgE may be an efficacious and safe immunotherapy for melanoma.

Legal entity responsible for the study

The authors.

Funding

Epsilogen Ltd.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

142P - Identifying novel immunotherapy targets using machine learning and ex vivo validation

Presentation Number
142P
Lecture Time
05:00 - 05:00
Speakers
  • Marcellus Augustine (London, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immunotherapy has transformed the cancer therapy landscape. However, since most patients do not respond, new alternatives are needed. Traditional immune-oncology (IO) target identification uses preclinical models with limited translation between species and an inability to fully recapitulate human tumour complexity. Large-scale multimodal molecular data from patient samples offers utility to discover new generations of targets with higher probability of successful translation into the clinic. Increasing attention is thus focussed on harnessing rich patient molecular data and modern machine learning approaches.

Methods

We profiled tumour-immune relationships to create a rich dataset for IO target discovery. Bulk exome and transcriptome data from n=1,317 immune checkpoint inhibitor-treated patients (Litchfield et al., Cell 2021) were integrated with single cell transcriptomic (scRNAseq) atlases from n=350 samples. We extended this to profile the immunopeptidome of 60 patients. Causal information regarding immune responses to genetic perturbation derived from n=7 genome-wide CRISPR tumour-T cell co-cultures and n=15,442 SNP-phenotype associations. Further biological context was provided by n=6,387 gene-disease, n=147,164 protein-protein and n=265,672 gene-regulatory interactions (Himmelstein et al., eLife 2017).

Results

We developed an ensemble approach comprising XGBoost, random forest, support vector machine and logistic regression models. This framework achieved held-out ROC-AUC>0.75 for classifying gene IO target status (whether a gene was addressed in IO clinical trials), and successfully ranked targets by clinical trial phase (p<0.001), despite being agnostic to these annotations. Interpretability analysis showed strong links between autoimmunity and IO targets. 41 targets were reviewed based on plausibility, tractability and commercial landscape. 4 were selected for immediate ex vivo validation in patient-derived explants and organoid co-cultures, with 5 reserved for potential future work.

Conclusions

Here we will present results from this study in progress and share learnings on the feasibility of using patient data and machine learning to discover new IO drug targets.

Legal entity responsible for the study

UCL.

Funding

Cancer Research UK Cancer Research Horizons.

Disclosure

C. Swanton: Financial Interests, Personal, Invited Speaker, Activity took place in 2016.: Pfizer, Celgene; Financial Interests, Personal, Invited Speaker, October 26th 2020: Novartis; Financial Interests, Personal, Invited Speaker: Roche/Ventana, BMS, AstraZeneca, MSD, Illumina, GSK; Financial Interests, Personal, Advisory Board, AdBoard - November 12th, 2020: Amgen; Financial Interests, Personal, Advisory Board, Current - since 2018: Genentech; Financial Interests, Personal, Advisory Board: Sarah Canon Research Institute; Financial Interests, Personal, Advisory Board, Joined October 2020. Also have stock options: Bicycle Therapeutics; Financial Interests, Personal, Other, Consultancy: Medicxi; Financial Interests, Personal, Advisory Board, Member of the Science Advisory Board. Also had stock options until June 2021: GRAIL; Financial Interests, Personal, Other, Consultancy agreement: Roche Innovation Centre Shanghai; Financial Interests, Personal, Advisory Board, 29 November - 1 December 2022: Novartis; Financial Interests, Personal, Invited Speaker, Oncology Collective - 2nd Nov - 4 Nov 2022 - Atlanta, USA: Roche; Financial Interests, Personal, Advisory Board, ctDNA advisory Board - 24th March 2023: AstraZeneca; Financial Interests, Personal, Invited Speaker, Pfizer Oncology 'Leading the revolution for the future: Pfizer; Financial Interests, Personal, Full or part-time Employment, Chief Clinician since October 2017: Cancer Research UK; Financial Interests, Personal, Ownership Interest, Co-Founder of Achilles Therapeutics. Also, have stock options in this company.: Achilles Therapeutics; Financial Interests, Personal, Stocks/Shares, Stocks owned until June 2021: GRAIL, Apogen Biotechnologies; Financial Interests, Personal, Stocks/Shares: Epic Biosciences, Bicycle Therapeutics; Financial Interests, Institutional, Research Grant, Funded RUBICON grant - October 2018 - April 2021: Bristol Myers Squibb; Financial Interests, Institutional, Research Grant, Collaboration in minimal residual disease sequencing technologies.: Archer Dx Inc; Financial Interests, Institutional, Research Grant: Pfizer, Boehringer Ingelheim; Financial Interests, Institutional, Trial Chair, Chief Investigator for the MeRmaiD 1and 2 clinical trials and chair of the steering committee.: AstraZeneca; Financial Interests, Institutional, Research Grant, Research grant from Oct 2019 - July 2023 - Genetics of CIN and SCNAs for Targeted Discovery (SCEPTRE): Ono Pharmaceutical; Financial Interests, Institutional, Research Grant, Research Grants from 2015: Roche; Financial Interests, Personal, Other, Co-chief investigator: NHS-Galleri Clinical Trial; Financial Interests, Institutional, Research Grant, from October 2022: Personalis; Non-Financial Interests, Personal, Principal Investigator, Chief Investigator for MeRmaiD 1and 2 clinical trials: AstraZeneca; Non-Financial Interests, Personal, Member of Board of Directors, From 2019-2022: AACR; Non-Financial Interests, Personal, Other, Board of Directors: AACR; Non-Financial Interests, Personal, Advisory Role, EACR Advisory Council member: EACR. K.R. Litchfield: Financial Interests, Personal, Invited Speaker: Roche Tissue Diagnostics; Financial Interests, Personal, Other, Consulting work: Kynos Therapeutics, Monopteros Therapeutics, Tempus; Financial Interests, Personal, Invited Speaker, Invited speaker: Ellipses Pharma; Financial Interests, Institutional, Research Grant: Ono/LifeArc; Financial Interests, Institutional, Research Grant, Research funding: Genesis Therapeutics; Non-Financial Interests, Institutional, Proprietary Information, Collaboration on data analysis: Bms. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

143P - Advancing Cancer Immunotherapy via HLA-G Pathway Modulation with UCB4594

Presentation Number
143P
Lecture Time
05:00 - 05:00
Speakers
  • Ann L. WHITE (Slough, United Kingdom)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Human leukocyte antigen-G (HLA-G), a tolerogenic protein usually restricted to the placenta, is upregulated by many solid cancers where it promotes tumour immune evasion by engaging inhibitory receptors immunoglobin-like transcript (ILT)2 and ILT4 on lymphoid and myeloid cells.UCB4594, a humanized afucosylated IgG1 HLA-G antibody, was developed to promote therapeutic tumour immune rejection by blocking interaction of HLA-G with ILT2 and ILT4 as well as direct induction of tumour cell death via natural killer (NK) cells, macrophages and complement.

Methods

Functional properties of UCB4594 were evaluated using a suite of in vitro assays including assessment of NK cell killing, complement dependent lysis, and antibody-dependent cellular phagocytosis (ADCP) of tumour targets. To further support its therapeutic potential, UCB4594-induced tumour cell killing dependent on tumour infiltrating immune cells was evaluated in 20 primary solid human tumours (10 renal and 10 colorectal) using an ex-vivo human tumoroid platform.

Results

UCB4594 exhibited strong affinity for HLA-G (4-6 nM for soluble HLA-G; <0.025 nM for cell expressed HLA-G), and high selectivity (no binding to other HLA-I molecules on transfected cells or human CD4 T-cells from 60 donors). UCB4594 effectively disrupted interaction of HLA-G with ILT2 and ILT4. UCB4594 induced NK cell killing of 85% and 40% of HLA-G transfected HCT116 cells and naturally HLA-G expressing JEG3 cells, respectively and activated potent complement-dependent lysis of HLA-G expressing Reh cells. UCB4594's dual mechanisms promoting phagocytosis are driven by Fc receptor-mediated ADCP and HLA-G receptor blocking (depletion of 22.0±10.8% of target cells at 0.1 μg/mL and 44.8±8.6% at 10 μg/mL), activity that was enhanced in the presence of anti-CD47. Across 20 human tumours, UCB4594 IgG1 led to 1.5-fold higher tumour cell death than the isotype control in 9 tumours, seen at 24h/72h post-treatment.

Conclusions

These findings support the development of UCB4594 as a novel cancer immunotherapy for application in several solid tumour indications.

Editorial acknowledgement

Medical writing support for this abstract was provided by Enago Life Sciences, India.

Legal entity responsible for the study

UCB Pharma.

Funding

UCB Pharma.

Disclosure

A.L. White, R. McElhone, G. Le Friec, T. Colley, V. O’Dowd: Financial Interests, Personal, Full or part-time Employment: UCB Pharma. C. Thompson, C. Berteau: Financial Interests, Personal, Full or part-time Employment: UCB Pharma; Financial Interests, Personal, Stocks/Shares: UCB Pharma.

Collapse
Poster Display (ID 34) Poster Display

144P - Discovery of CBO421, a first-in-class Drug Fc-Conjugate (DFC), targeting CD73 in Cancer

Presentation Number
144P
Lecture Time
05:00 - 05:00
Speakers
  • Simon Döhrmann (San Diego, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

CD73 (NT5E) contributes to immune evasion in solid tumors by producing immuno-suppressive adenosine. Herein, we describe CBO421, a CD73 targeting DFC, that is a multivalent conjugate of a novel small molecule inhibitor stably linked to a proprietary immune-silent human IgG1 Fc. CBO421 combines the strengths of small molecule inhibitors and monoclonal antibodies (mAbs) targeting CD73 that are in clinical development, with potential best-in-class activity.

Methods

CD73 inhibition was evaluated, and functional activity measured in a PBMC rescue assay with AMP. Binding to cancer cells and CD73 internalization was measured by flow cytometry. Tumor spheroid penetration was measured with CD73+ cancer cells. Efficacy of CBO421 was evaluated in syngeneic mouse models.

Results

CBO421 is a potent, AMP-competitive inhibitor of CD73. CD73 inhibition by CBO421 was comparable or superior to small molecules and anti-CD73 mAbs. Unlike most anti-CD73 mAbs, CBO421 demonstrated complete CD73 enzyme inhibition. CBO421 demonstrated potent reactivation of CD8+ T cells as measured by CD25+ and granzyme B+, that was comparable to small molecules and significantly more potent than anti-CD73 mAbs. Differentiated from small molecules, CBO421 triggered CD73 internalization as a second mechanism to reduce CD73 mediated adenosine production. The 2.3-fold smaller size and hydrodynamic radius of CBO421 compared to mAbs significantly improved tumor spheroid penetration. CBO421 demonstrated significant tumor growth inhibition (TGI) in multiple syngeneic mouse models with CD73- or CD73+ cancer cells. Combination therapy of CBO421 with an anti-PD-1 mAb resulted in significant increases in TGI and complete responses when compared with the respective monotherapy. Immunologic memory was demonstrated in complete responders by rejection of tumor upon re-challenge.

Conclusions

CBO421 demonstrated high potency, combined with multiple, distinct mechanisms of action that translated to potent antitumor activity as a monotherapy in syngeneic mouse models that was further improved in combination with PD-1 therapy. Based on these results and other emerging data, CBO421 is being advanced as a clinical development candidate for the treatment of solid cancers.

Legal entity responsible for the study

Cidara Therapeutics, Inc.

Funding

Cidara Therapeutics, Inc.

Disclosure

S. Döhrmann, J. Levin, N. Dedeic, A. Almaguer, D. Zuill, E. Abelovski, M. Hernandez, K. Amundson, M. Moniz, D. Panickar, T. Lam, T. Brady, A. Borchardt, J.N. Cole: Financial Interests, Personal, Full or part-time Employment: Cidara Therapeutics, Inc. Q. Zhao, H. Chen: Financial Interests, Personal, Advisory Board: Cidara Therapeutics, Inc. L.W. Tari: Financial Interests, Personal, Leadership Role: Cidara Therapeutics, Inc.

Collapse
Poster Display (ID 34) Poster Display

145P - An Engineered Ligand-Trap Biologic Targeting the CD47 Signaling Pathway for Cancer Treatment with Superb Efficacy and Safety Profiles

Presentation Number
145P
Lecture Time
05:00 - 05:00
Speakers
  • ZONG SEAN JUO (Taipei City, Taiwan)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Macrophages exert their functions mainly through SIRPa receptor, which interacts with CD47 on cellular targets. As many cancer cells overexpress CD47 to evade immune surveillance, blocking SIRPa-CD47 interaction represents a promising approach to control tumor progression. The clinical progress of anti-CD47 antibodies were hindered by either side effects or lacking appreciable efficacy. To overcome this dilemma, we engineered a SIRPa-fusion protein that exhibits superior efficacy against multiple tumor types while maintaining good safety profiles.

Methods

Using structure-guided protein engineering, we selected a SIRPa mutant that exhibited marked phagocytic abilities against tumors while maintaining good safety features on normal cells. To assess the efficacy of this molecule, it was tested in multiple xenograft mouse models alongside competing biologics currently in clinical trials. We also performed quantitative RNA transcriptional analysis to evaluate the changes in gene expression inside tumor and in the tumor microenvironment.

Results

Comparing to other clinical candidates, HCB101 triggered strong phagocytic reactions against tumor cells but not red blood cells. We have analyzed 14 human tumor xenograft models, HCB101 consistently showed excellent efficacy against heme and solid tumors, with tumor growth inhibition index ranging from 60-100% at the dose of 0.5-10mg/kg over placebo. We also observed an increase in M1/M2 macrophage ratio after the treatment with HCB101, which correlated with the observed anti-tumor efficacy. Quantitative RNA transcriptional analysis indicated that HCB101 triggered drastic changes in gene expression comparing to other competing molecules. This suggested a unique MOA underlying HCB101’s superior efficacy. There was no apparent adverse reaction observed during the toxicology studies, indicating a good safety profile.

Conclusions

Comparing to relevant clinical candidates, HCB101 exhibits superior efficacy in 14 different CDX models of hemotological and solid tumors while maintaining good safety profiles. It is a highly effective biologic with robust efficacy, both as monotherapy and in combination. Clinical trial of HCB101 is now in progress.

Clinical trial identification

NCT05892718.

Legal entity responsible for the study

HanchorBio, Inc.

Funding

HanchorBio, Inc.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

130P - A Phase I Study of a Novel BCMA_CD3 Bispecific Antibody EMB-06 in Relapsed or Refractory Multiple Myeloma (RRMM)

Presentation Number
130P
Lecture Time
05:00 - 05:00
Speakers
  • Peter Thian Guan Tan (Nedlands, Australia)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

146P - A Novel Allosteric Oral Immunotherapy Small Molecule Modulates Adenosine 2A Receptor Signaling and Restores Anti-Tumor Immune Responses

Presentation Number
146P
Lecture Time
05:00 - 05:00
Speakers
  • David Pejoski (Geneva, Switzerland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Improving survival of cancer patients could be achieved by enhancing anti-tumor immunity which is frequently suppressed. Adenosine, found at high concentrations in most solid tumors, is reported to be a key mediator of immune suppression. Current drugs that target a key immune system switch, the adenosine 2A receptor (A2AR), are confronted with developmental hurdles related to their conventional (orthosteric) molecular mode of action, which cannot modulate A2AR signaling in immunosuppressive solid tumor microenvironment without safety concerns.

Methods

A drug discovery campaign at University of Geneva identified, and improved the potency of, orally bioavailable, non-brain penetrant, small molecule negative allosteric modulators (NAM) of A2AR. Optimized lead drug candidates were evaluated for safety, pharmacokinetics, and pharmacodynamics (target engagement, immune response restoration and tumor growth control) using in vitro, ex vivo, and in vivo models.

Results

A2AR NAMs displayed an excellent safety profile using industry-standard in vitro assays, and when administered to mice. The drug candidates fully restored anti-tumor cytokine secretion in primary human PBMC subsets, outperforming existing A2AR antagonists which have been tested in phase I or II human trials. A2AR NAMs were detected in mouse plasma at therapeutic concentrations after administration via the oral or subcutaneous routes. When tested in high adenosine mouse tumor models such as MC38 and CT26, A2AR NAMs significantly reduced tumor growth as a single agent immunotherapy, outperforming PD-1 immune checkpoint blockade as well as a small molecule A2AR drug candidate currently in Ph-II clinical development.

Conclusions

A2AR NAMs have an excellent safety and efficacy profile in translational assays and biologically relevant mouse tumor models, presumably stemming from their novel allosteric molecular mode of action. The drug candidates represent a next generation approach to A2AR blockade, which are intended to enhance the immune system and favor positive responses in solid tumor patients, especially when used in combination with other standard and immune-based cancer therapies.

Legal entity responsible for the study

University of Geneva.

Funding

Innosuisse.

Disclosure

D. Pejoski, H. Hamed: Financial Interests, Institutional, Member of Board of Directors: Adoram Therapeutics. M. Boujut: Financial Interests, Institutional, Stocks/Shares: Adoram Therapeutics. L. Scapozza: Financial Interests, Institutional, Advisory Role: Adoram Therapeutics. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

147P - Pre-clinical evaluation and safety profile of the highly selective anti-VISTA antibody K01401-020

Presentation Number
147P
Lecture Time
05:00 - 05:00
Speakers
  • Geneviève Gueguen Dorbes (Toulouse, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

V-domain Immunoglobulin Suppressor of T cell Activation (VISTA) is an immune checkpoint regulator that is highly expressed in myeloid cells in the tumor microenvironment (TME). VISTA has been shown to maintain immunosuppressive conditions in the TME. Its expression is associated with poor prognosis in several cancers and with resistance to immune checkpoints inhibitors which makes it a very attractive target for cancer immunotherapy. K01401-020 is a humanized IgG antibody targeting monkey and human VISTA with a high affinity.

Methods

In vitro assays were performed using human recombinant proteins and peripheral blood mononuclear cells (PBMC) from healthy donors. Human VISTA knock-in (KI) mice were generated, engrafted with MC38 cells, and used for the assessment of anti-tumor activity. Non-Human Primate (NHP) were used in GLP 6- and 13-week repeat-dose toxicity studies.

Results

K01401-020 blocks VISTA binding to its main partners (PSGL-1, VSIG3 and VSIG8) independently of pH. In in vitro assay with PBMCs, K01401-020 stimulates monocytes’ activation and proliferation of NK cells and induces release of cytokines contributing to T cell activation. In vivo, K01401-020 treatment mediates single-agent antitumor activity in a syngeneic tumor model by activating the immune response in the tumors. Moreover, K01401-020 shows an enhanced antitumor response when combined with anti-PD-1 antibody. In the NHP 13-week toxicity study, minimal and reversible clinical observations and microscopic findings have only been observed at the highest dose (100 mg/kg/inj). Among the various cytokines tested, transient increases in IL-6, MCP-1 and IP-10 levels have been observed, in a dose-independent manner. At safe lower doses in NHP, exposures were 10-fold above those necessary in mice to observe antitumor activity, suggesting a favorable safety profile.

Conclusions

K01401-020 is a non-pH-sensitive antibody targeting VISTA, inducing immune cell activation and anti-tumor response, as single agent and in combination with anti PD-1 which is well tolerated in NHPs. A First-in-Human dose escalation study as single agent and in combination with pembrolizumab is currently ongoing in advanced solid tumors (NCT04564417).

Legal entity responsible for the study

Pierre Fabre Laboratories.

Funding

Pierre Fabre Laboratories.

Disclosure

G. Gueguen Dorbes, N. Loukili, A. Petain, J. Labbe, N. Boute, B. Akla, M. Broussas, P. Ferre, F. Hofmann: Financial Interests, Personal, Full or part-time Employment: Pierre Fabre.

Collapse
Poster Display (ID 34) Poster Display

148P - HexaBody-OX40, a novel Fc_ receptor crosslinking-independent OX40-targeting antibody, exhibits agonistic activity in vitro and antitumor activity in vivo

Presentation Number
148P
Lecture Time
05:00 - 05:00
Speakers
  • Kristel Kemper (Utrecht, Netherlands)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Clustering of the costimulatory TNF receptor superfamily member OX40 on activated T cells activates signaling pathways that enhance T-cell activation, survival, and proliferation. OX40 agonists in development requiring FcγR-mediated crosslinking to induce OX40 agonism have demonstrated limited clinical activity. We present the preclinical characterization of HexaBody-OX40 (GEN1055/BNT315), a novel OX40 agonist antibody designed to cluster OX40 independent of FcγR-mediated crosslinking to enhance antitumor T-cell responses.

Methods

Target binding characteristics and agonistic activity of HexaBody-OX40 were analyzed in vitro using binding assays, cell-based reporter assays, and functional assays using primary human T cells. Antitumor activity and pharmacodynamics of HexaBody-OX40 were assessed in vivo in MC38 tumor-bearing human OX40 knock-in mice.

Results

HexaBody-OX40 bound to activated OX40-expressing T cells. In contrast to other OX40 agonists, HexaBody-OX40 induced FcγR-independent OX40 agonist activity in a T-cell based reporter assay. In an assay using polyclonally stimulated human PBMCs, HexaBody-OX40 enhanced proliferation and activation of CD4+ and CD8+ T cells, and cytokine secretion. In this assay, OX40 agonistic activity in CD8+ T cells depended on the presence of CD4+ T cells. Furthermore, HexaBody-OX40 enhanced proliferation of antigen-specific CD8+ T cells overexpressing OX40. In vivo antitumor activity of HexaBody-OX40 in a syngeneic MC38 model in human OX40 knock-in mice was associated with peripheral T-cell proliferation and activation, increased percentages of tumor-specific CD8+ T cells, and an intratumoral increase in CD4+ T cells and Granzyme B+ cells.

Conclusions

In preclinical studies, HexaBody-OX40 exhibited FcγR-crosslinking-independent OX40 agonist activity, a unique mechanism of action that is distinct from other OX40 agonists. HexaBody-OX40 enhanced T-cell activation and proliferation in vitro and showed antitumor activity in vivo. A first-in-human clinical trial is planned to evaluate clinical safety and preliminary efficacy of HexaBody-OX40 in patients with advanced solid tumors.

Legal entity responsible for the study

The authors.

Funding

Genmab BV, BioNTech SE.

Disclosure

K. Kemper, M. van der Kroef, G. Zom, A. Gorlani, L. Guelen, D. Satijn, T. Ahmadi, E. Breij: Financial Interests, Personal, Full or part-time Employment: Genmab; Financial Interests, Personal, Stocks/Shares: Genmab BV. M. Köhne, K.B.B. Nuermberger, A. Krause, F. Gieseke, A. Muik, S. Fellermeier-Kopf, î Türeci, U. Sahin: Financial Interests, Personal, Full or part-time Employment: BioNtech SE; Financial Interests, Personal, Stocks/Shares: BioNTech SE. M. Russier, P. de Goeje: Financial Interests, Personal, Stocks/Shares: Genmab BV; Financial Interests, Personal, Full or part-time Employment: Genmab BV.

Collapse
Poster Display (ID 34) Poster Display

149P - HLA/SIRPa bispecifics-A novel multitarget therapeutic strategy to induce potent anti-tumor immune responses

Presentation Number
149P
Lecture Time
05:00 - 05:00
Speakers
  • Anahita Rafiei (Zurich, Switzerland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

To develop novel anti-cancer therapeutics, we have used a reverse rational approach and searched for human HLA class I molecules known to induce autoimmunity as a surrogate marker for potential anti-cancer activity. HLA-B57 is one example of an HLA class I molecule described to target LILRB receptors in these studies. Here we demonstrate the proof of concept of a bispecific optimised HLA-Fc fusion conjugated to a SIRPa protein which shows potent in vitro anti-tumor efficacy through its multimodal binding of LILRB1, LILRB2 and CD47 receptors. This novel modality of HLA/SIRPa Bispecifics has the unique characteristic of targeting myeloid cells and the ability to be directed to tumor sites by targeting the CD47 on cancer cells.

Methods

Rational guided protein engineering and transient protein production in CHO was conducted. Affinity determination was measured by SPR/BLI. Binding to cells expressing the receptors was assessed by flow cytometry. Potency was measured using CD47/SIRPa reporter cell lines. Evaluation of in vitro safety was performed by binding assessment to red blood cells and hemagglutination induction. In vitro efficacy studies were performed for macrophage phagocytosis and NK cell cytotoxicity against cancer cells. Ex-vivo efficacy is currently being assessed using primary NSCLC tumor biopsies and measuring tumor cell killing and activation of immune cell markers.

Results

HLA/SIRPa Bispecifics were expressed as IgG4 Fc-fusion proteins in a 2+2 format. Affinity to LILRB1/LILRB2 and CD47 receptors are in low nM range. By optimization of the affinity of the c-terminal SIRPa we can show an enhanced safety profile compared to competitor CD47 targeting molecules through reduced binding to red blood cells and reduced hemagglutination. Reporter cells demonstrate that bispecifics effectively inhibit the interaction of CD47 with SIRPa. In vitro efficacy data shows that macrophage phagocytosis was enhanced by optimizing specific properties of the SIRPa arm.

Conclusions

HLA/SIRPa bispecifics are novel multi-functional agents that potentiate anti-tumor immunity through the interaction of multiple immune checkpoint receptors on one side and may function to engage macrophages towards cancer cells on the other side.

Legal entity responsible for the study

ImmunOs Therapeutics AG.

Funding

ImmunOs Therapeutics AG.

Disclosure

A. Rafiei: Non-Financial Interests, Personal, Full or part-time Employment: ImmunOs Therapeutics AG. O. Marroquin Belaunzaran: Financial Interests, Personal, Officer: ImmunOs Therapeutics AG. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

150P - Chemotherapy in combination with Toll-like receptor agonism promoted antitumor immune response in triple negative breast cancer

Presentation Number
150P
Lecture Time
05:00 - 05:00
Speakers
  • Eunice Dotse (Kowloon, Hong Kong PRC)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immunotherapy represents a promising research area for triple negative breast cancers (TNBCs), a breast cancer subtype defined by lack of estrogen and progesterone receptors, and HER2 overexpression. Current treatment is often limited to chemotherapy which is toxic and short lived with imminent metastatic recurrence. TNBCs are more immunogenic, often characterized by high number of tumors infiltrating lymphocytes, a feature which can be harnessed to increase responsiveness to immunotherapy. Immune cells are known to express pathogen recognition receptors such as toll-like receptors (TLRs) and their engagement activates downstream pathways to elicit specific T cell antitumor immunity.

Methods

To utilize immune modulation strategy for TNBC treatment, the study evaluated the combined efficacy of Paclitaxel with intratumoral administration of TLR7/8 (Resiquimod) or TLR9 (CPG-ODN-2395) agonists in syngeneic TNBC mouse model. Tumor volume was measured, and percentages of tumor immune cell infiltrates were determined by flow cytometry at endpoint. Gene expression studies were also carried out on excised tumors using qPCR.

Results

Showed chemotherapy with Resiquimod or CPG-ODN 2395 promoted significant tumor regression compared to chemotherapy alone. Also, Paclitaxel/Resiquimod treatment significantly increased influx of B-cells, pDCs, helper and cytotoxic T-cells, and reduced regulatory T-cells tumor infiltrates compared to Paclitaxel/CPG-ODN 2395 and chemotherapy alone. Furthermore, Paclitaxel/Resiquimod treatment significantly upregulated gene expression of chemokines attracting cytotoxic T lymphocytes, type-1 helper, and NK cells as well as increased gene expression of IFN-γ, perforin, and granzyme B. Vimentin and Claudin 5, genes associated with epithelial mesenchymal transition and metastasis respectively were downregulated following Paclitaxel/Resiquimod treatment.

Conclusions

Overall, findings suggest that combination therapy of TLR7/8 agonist (Resiquimod) with Paclitaxel, promotes antitumor immunity and may represent a more effective treatment approach for TNBC.

Legal entity responsible for the study

The authors.

Funding

Croucher innovative award.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

151P - Tumor organoid-derived TIL therapy for colorectal cancer

Presentation Number
151P
Lecture Time
05:00 - 05:00
Speakers
  • Marc Leushacke (Palo Alto, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Colorectal cancer (CRC) represents ∼7% of new cancer cases and 11% of cancer deaths worldwide. Tumor-Infiltrating Lymphocyte (TIL) immunotherapy is efficacious against melanoma, but its potency in epithelial cancers such as CRC remains inconsistent. We hypothesized that growing tumor organoids in an Air-Liquid Interface (ALI) system prior to TIL expansion might select for the rare tumor-specific T cells that mediate antitumor activity.

Methods

ALI CRC organoids were generated by embedding intact tumor fragments within a collagen matrix on top of a permeable support membrane exposed to both air and culture medium. This configuration facilitates optimal oxygenation, supporting the growth of faithful mini replicas of original tissue that preserve stroma and tumor-infiltrating immune cells, including cytotoxic T cells. To characterize ALI TILs after ex vivo expansion, cell surface markers were analyzed by flow cytometry and immune gene expression by single-cell RNA sequencing. We also established submerged organoids in vitro and transplanted them as organoid derived xenografts (ODX) in mice to assess antitumor activity. Tumor reactivity and T cell cytotoxicity were measured as cytokine induction and tumor cell death upon in vitro co-culture, while preclinical in vivo efficacy was assessed as tumor growth inhibition.

Results

Our ALI process yielded high cell numbers, mostly comprised of CD4+ and CD8+ T cells of the effector and central memory subtypes. T cell receptor analysis revealed unique sets of polyclonal repertoires, suggestive of tumor specificity. Functionally, ALI TIL tumor reactivity and killing were demonstrated in vitro, that translated to a potent in vivo antitumor activity against autologous ODX models.

Conclusions

Our study presents an innovative TIL immunotherapy approach for CRC. The application of ALI organoid culture conditions prior to the ex vivo TIL expansion resulted in a cell product with appropriate phenotypic and functional features, warranting further development of the novel process.

Legal entity responsible for the study

The authors.

Funding

Khosla Ventures, Peregrine Ventures, Alexandria, Wilson Sonsini.

Disclosure

M. Leushacke: Financial Interests, Institutional, Principal Investigator: Nextvivo. M. Pari, J. Ju, P-Y. Lin, B. Shreshta: Financial Interests, Personal, Stocks/Shares: NextVivo. C. Chartier: Financial Interests, Institutional, Officer: NextVivo.

Collapse
Poster Display (ID 34) Poster Display

152P - Discovery of best-in-class dual-acting A2AR/A2BR antagonists that are functional in high adenosine environment

Presentation Number
152P
Lecture Time
05:00 - 05:00
Speakers
  • Nainesh Katagihallimath (Bengaluru, India)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Adenosine accumulates in the tumor microenvironment due to abnormal tumor metabolism and hypoxia. It binds to A2A and A2B receptors on immune cells and impairs their activation and cytotoxic ability, helping cancers to evade the immune system and resist cancer therapies. Adenosine receptor antagonists that act in pathological concentrations of adenosine can rescue T cell and innate immune suppression and improve the efficacy of existing treatments.

Methods

We used structure-guided medicinal chemistry to design adenosine antagonists and measured how well they bind to A2A and A2B receptors in cells using HTRF and Tag-lite technology. Their ability to block adenosine’s effect on primary immune cells was monitored by measuring CREB phosphorylation and cytokine production. In vitro coculture assays and syngeneic mouse models were used to check the effect of immune response and tumor growth. We also used a patient-derived ex vivo tumor platform to demonstrate clinical translatability.

Results

BWC2094 is a potent A2A receptor antagonist, while BWC2562 is a potent A2A/A2B dual antagonist in a high adenosine environment. They selectively bind A2A at picomolar concentrations and had >50-fold selectivity over A1 and A3 receptors. They reduced the phosphorylation of CREB and restored the production of cytokines, such as IL2, TNFα and IFNγ in adenosine-suppressed immune cells. The lead compounds exhibited a synergistic effect with standard of care drugs in co-culture assays of tumor cells and PBMC. Both, BWC2094 and BWC2562 demonstrated good oral bioavailability in rodents (>25 %F) and dogs (>50 %F) and are well tolerated for 14 days in rat acute toxicity study. At a dose of 3mg/kg, they reduce the tumor volume as monotherapy and enhanced the anti-tumor effects of anti-PD1/PDL1 and CTLA4 in CT26, MC38, and MCA205 murine tumor models. In a patient-derived ex vivo tumor model, Precision immunotherapy using Complementary approaches in Systems Oncology (PiCaSO), they reduced tumor density, decreased Ki67 expression and increased apoptosis/necrosis.

Conclusions

BWC2094 and BWC2562 can block the immunosuppressive effects of adenosine and improve the effectiveness of existing therapies in oncology.

Legal entity responsible for the study

The authors.

Funding

Bugworks Research Inc.

Disclosure

N. Katagihallimath, R. Nandishaiah, S. Sharma, S. Venkatesan, R. Rao, N. Bharatham, M. Gupta, D. Kundu, H. Kaushik Kotakonda, R. Ramalingam Kalainesan, S. Reddy, B. Tewary, L. Maitreyi, B. Venkatraman, S. V Rekha Sody, B. Majumder: Financial Interests, Personal and Institutional, Stocks/Shares: Bugworks. S. Datta, S.Hameed: Financial Interests, Personal and Institutional, Stocks or ownership: Bugworks. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

153P - Discovery of a Novel, Dual CD73 and PD-1 Targeting Multispecific Drug Fc-Conjugate (DFC) for the Treatment of Cancer

Presentation Number
153P
Lecture Time
05:00 - 05:00
Speakers
  • James Levin (San Diego, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The approval of several PD-1/L1 axis inhibitors has revolutionized cancer therapy and established a role for CD8+ T cells in tumor destruction. While anti-PD-1 therapy has demonstrated durable responses, only a subset of patients respond. To improve response rates, we developed a multispecific PD-1/CD73 targeting DFC. CD73 catalyzes the rate limiting step in production of immuno-suppressive adenosine, which inhibits immune cell activation through a mechanism distinct from PD-1. Herein, we describe a first-in-class dual targeting DFC, comprising a multivalent conjugate of a small molecule CD73 inhibitor to a proprietary human IgG1 Fc-fusion with a PD-1 inhibitor peptide. This dual-targeting DFC has potential for differentiation from approved PD-1/L1 inhibitors.

Methods

Binding of the dual targeting DFC to biotinylated hPD-1 was determined using a commercial kit and to human CD8+ T cells by flow cytometry. CD73 inhibition was determined in cell-free and cell-based assays. Efficacy was evaluated in transgenic mice expressing human PD-1/PD-L1 with MC-38 (hPD-L1) tumors (Genoway, France). Tumor volumes were recorded and statistical analysis was by t-test (Mann-Whitney) or two-way ANOVA.

Results

The dual targeting DFC demonstrated potent activity against both checkpoint targets: Binding to hPD-1 with an IC50 of < 1 nM and functional inhibition of CD73 with an EC50 of 9 nM. Efficacy was determined in a humanized mouse model against the colon cell line MC-38 (hPD-L1). The dual targeting DFC demonstrated a statistically significant reduction in tumor volume (∼55%) at a dose of 3 mg/kg (P<0.0001). Pembrolizumab biosimilar at 10 mg/kg did not result in a significant reduction in tumor volume.

Conclusions

This work describes a dual targeting DFC with potent activity against two validated immune checkpoint pathways. The in vitro activity translated to efficacy in a humanized mouse model at doses as low as 3 mg/kg. The relative contributions of each ligand to activity against different tumors is under investigation as Cidara’s first multispecific DFC advances through preclinical development.

Legal entity responsible for the study

Cidara Therapeutics.

Funding

Cidara Therapeutics.

Disclosure

J. Levin, S. Döhrmann, N. Dedeic, A. Almaguer, D. Zuill, E. Abelovski, J. Fortier, Q. Zhao, M. Hernandez, K. Amundson, M. Moniz, H. Chen, D. Panickar, T. Lam, T. Brady, A. Borchardt, J.N. Cole, G. Hough, J.B. Locke, L.W. Tari: Financial Interests, Personal, Full or part-time Employment: Cidara Therapeutics.

Collapse
Poster Display (ID 34) Poster Display

154P - Computer-aided drug design based on CLDN4 ligand and its biological evaluation in ovarian cancer

Presentation Number
154P
Lecture Time
05:00 - 05:00
Speakers
  • Yi Xu (Nanjing, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Numerous studies have consistently highlighted CLDN4 as a viable candidate for targeted therapy across various ovarian tumor types. Specifically, the C-terminal region of Clostridium perfringens enterotoxin (CPE) has demonstrated the ability to establish a stable complex with CLDN4. In our investigation, we harnessed computer-aided drug design (CADD) technology to craft peptides characterized by a specific and robust affinity for CLDN4. These peptides hold the potential to facilitate the development of self-assembling anti-tumor Peptide-Drug Conjugates (PDCs).

Methods

We employed virtual screening techniques, including alanine mutation, saturation mutation, and multi-point mutation, in conjunction with DS software to screen peptide sequences. To validate the binding capacity of the selected peptides with the CLDN4 protein, we conducted Surface Plasmon Resonance (SPR) and immunofluorescence colocalization experiments. Subsequently, we prepared Peptide-Drug Conjugates (PDCs), taking advantage of their inherent hydrophilic and hydrophobic properties, which promote their spontaneous assembly into nanofibrous structures. The anti-tumor efficacy of these formulations was rigorously assessed through both in vivo and in vitro experiments.

Results

SPR analysis revealed a noteworthy binding affinity between the targeted peptide and the CLDN4 protein, as evidenced by a Kd value of 5.343nM. Immunofluorescence co-localization experiments unequivocally demonstrated the co-localization of the targeted peptide with CLDN4. Furthermore, The PDC self-assembly group exhibited superior cytotoxicity compared to both the PDC non-self-assembly group and the camptothecin group.

Conclusions

In this study, we harnessed Computer-Aided Drug Design (CADD) technology to effectively engineer a targeting peptide characterized by a strong affinity for the CLDN4 protein. This peptide was subsequently employed in the self-assembly of Protein-Drug Conjugates (PDCs). The therapeutic potential of these PDCs for ovarian cancer was robustly substantiated through a comprehensive array of in vitro and in vivo experiments, unequivocally affirming their promise as a viable treatment option.

Legal entity responsible for the study

The authors.

Funding

National Natural Science Foundation of China.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

155P - A Phase 1 Study Exploring the Safety and Tolerability of the Small-Molecule PD-L1 Inhibitor INCB099280 in Select Advanced Solid Tumors

Presentation Number
155P
Lecture Time
05:00 - 05:00
Speakers
  • Hans Prenen (Edegem, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

INCB099280, an oral, programmed death ligand 1 (PD-L1) inhibitor, has shown preliminary efficacy and acceptable safety in an ongoing Phase 1, open-label, multicenter study in patients (pts) with advanced solid tumors (Prenen, et al. SITC 2022). Here we present updated results.

Methods

Eligible pts were ≥18 years with ECOG PS ≤1, and disease progression after available treatment (tx) or were ineligible for/without access to standard tx. In part 1, INCB099280 dose was escalated from 100 mg QD with a Bayesian optimal interval design. In part 2, 3 expansion cohorts with select tumor types were studied: 1) IO-naive pts, 2) IO-naive pts with MSI-H/dMMR tumors, 3) pts who progressed on anti-PD-1 mAb. Primary endpoints are INCB099280 safety, tolerability, and pharmacologically active/MTD determination. INCB099280 pharmacokinetics, objective response rate per RECIST v1.1, and biomarkers of pharmacologic activity were also analyzed.

Results

As of June 22, 2023, 172 pts had received INCB099280 at doses from 100 mg QD to 800 mg BID (median age, 63 years [range, 21–86]; ≥2 prior lines of tx, 64.0%; prior IO, 14.5%; most common tumor types: anal [14.5%], cervical [8.7%], and colorectal [7.6%]). Dose was escalated to 800 mg BID; MTD was not reached. 5 dose levels were expanded in part 2 up to 800 mg BID. Overall, 137 pts (79.7%) discontinued treatment, 119 (69.2%) due to disease progression; 95.3% of pts had ≥1 tx-emergent adverse event (TEAE) (Table). Several responses have been observed, and updated results will be presented. In pts with complete response (n=2), baseline tumor mutational burden scores were high (34–49 mut/Mb) and ctDNA levels at cycle 4 day 1/end of tx had decreased by 92.3% from baseline.

Safety

TEAEs, n (%) Total Pts (N=172)
Any grade Grade ≥3
All cause TEAEs 164 (95.3) 61 (35.5)
Occurring in >20% of pts
Asthenia 52 (30.2) 4 (2.3)
Decreased appetite 47 (27.3) 4 (2.3)
Nausea 43 (25.0) 2 (1.2)
Vomiting 39 (22.7) 2 (1.2)
Fatigue 35 (20.3) 3 (1.7)
Immune-related TEAEs 37 (21.5) 10 (5.8)
Serious TEAEs 45 (26.2)
Occurring in >1 pt
Pyrexia 4 (2.3)
Pneumonia 3 (1.7)
Sepsis 3 (1.7)
Anemia 2 (1.2)
Dyspnea 2 (1.2)
Hypercalcemia 2 (1.2)
Large intestinal obstruction 2 (1.2)
Pneumothorax 2 (1.2)
Urinary tract infection 2 (1.2)
Grade ≥3 tx-related TEAEs 21 (12.2)
Occurring in >1 pt
Increased ALT 4 (2.3)
Increased AST 3 (1.7)
Increased lipase 3 (1.7)
Anemia 2 (1.2)
Lymphopenia 2 (1.2)

Conclusions

INCB099280 was generally well tolerated at all doses tested. Updated results indicate promising antitumor activity and support future development of INCB099280 as monotherapy and in combination regimens for advanced solid tumors.

Clinical trial identification

NCT04242199.

Editorial acknowledgement

Editorial assistance was provided by Emily Sun and Andrew Marson-Neep of Envision Pharma Group (Philadelphia PA, USA).

Legal entity responsible for the study

Incyte Corporation, Wilmington, DE.

Funding

Incyte Corporation, Wilmington, DE.

Disclosure

H. Prenen: Financial Interests, Personal, Speaker, Consultant, Advisor: Biocartis, Cureteq; Financial Interests, Personal, Other, Honoraria: Amgen, AstraZeneca, Bayer, Roche, and Sanofi. T. Lesimple: Financial Interests, Personal, Speaker, Consultant, Advisor: BMS, MSD, Novartis, and Pierre Fabre. M. Robert: Financial Interests, Personal, Advisory Board: AstraZeneca and Eisai; Financial Interests, Personal, Other, Travel Grants: AstraZeneca. B.R. Delafontaine: Financial Interests, Personal, Speaker, Consultant, Advisor: Sanofi. P. Tomasini: Financial Interests, Personal, Speaker, Consultant, Advisor: Amgen, AstraZeneca, Bristol Myers SquibbFoundation, Janssen, Roche, and Takeda; Financial Interests, Institutional, Funding: AstraZeneca, Bristol Myers Squibb/Pfizer and Takeda. T. Meniawy: Financial Interests, Personal, Speaker, Consultant, Advisor: BMS GmbH & Co. KG, Eisai, GSK, MSD, Novartis, Regeneron, and Sanofi; Financial Interests, Institutional, Research Funding: AstraZeneca/MedImmune, Bayer, BeiGene, Bristol Myers Squibb, Incyte, Merck Serono, Regeneron, Roche/Genentech. E. Van Cutsem: Financial Interests, Personal, Speaker, Consultant, Advisor: Array, AstraZeneca, Bayer, Biocartis, Bristol Myers Squibb, Celgene, Eli Lilly, Ipsen, MSD, Merck KGaA, Novartis, Pierre Fabre, Roche, Servier, Sirtex, and Taiho; Financial Interests, Institutional, Research Funding: Amgen, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Eli Lilly, Ipsen, Merck, Merck KGaA, Novartis, Roche, and Servier. S.A. Piha-Paul: Financial Interests, Institutional, Research Funding: AbbVie, Inc., ABM Therapeutics, Inc., Acepodia, Inc, Alkermes, Aminex Therapeutics, Amphivena Therapeutics, Inc., BioMarin Pharmaceutical, Inc, Boehringer Ingelheim, Bristol Myers Squibb, Cerulean Pharma, Inc., Chugai Pharmaceutical Co., Ltd, Curis, Inc.; Financial Interests, Personal, Speaker, Consultant, Advisor: CRC Oncology. M.T. Schweizer: Financial Interests, Personal, Other, Consultancy/Honoraria: AstraZeneca, PharmaIn, Resverlogix, and Sanofi; Financial Interests, Institutional, Research Funding: Ambrix, Inc., AstraZeneca, Bristol Myers Squibb, Hoffman-La Roche, Immunomedics, Janssen, Madison Vaccines, Merck, Pfizer, SignalOne Bio, Tmunity, and Zenith Epigenetics. S. Gadgeel: Financial Interests, Personal, Other, Honoraria: Merck; Financial Interests, Personal, Speaker, Consultant, Advisor: AstraZeneca, Blueprint Medicines, Bristol Myers Squibb, Daichii Sanyko , Eisai, Genentech/Roche, Gilead Sciences, GSK, Janssen Oncology, Lilly; Merck, Mirati Therapeutics, Novartis, Pfizer, Takeda; Financial Interests, Institutional, Research Funding: Amgen (Inst), Astellas Pharma (Inst), AstraZeneca, AstraZeneca (Inst), BioMed Valley Discoveries (Inst), Blueprint Medicines (Inst), Calithera Biosciences (Inst), Daichii Sankyo (Inst), Daiichi Sankyo (Inst), Dragonfly Therapeutics (Inst), eFFECTOR Therap; Financial Interests, Personal, Other, travel, accommodations, expenses: Mirati Therapeutics; Financial Interests, Personal, Other: AstraZeneca. S. Kondo: Financial Interests, Personal, Speaker’s Bureau: Incyte; Financial Interests, Personal, Other, Honoraria: Chugai Pharma, Eisai, and Incyte; Financial Interests, Personal, Speaker, Consultant, Advisor: Takeda; Financial Interests, Institutional, Research Funding: AbbVie (Inst), AstraZeneca (Inst), Eisai (Inst), and Lilly (Inst). K. Ouali: Financial Interests, Personal, Speaker, Consultant, Advisor: Sotio; Financial Interests, Institutional, Funding: Amgen and Sotio. Y. Kuboki: Financial Interests, Institutional, Research Funding: Taiho, Takeda, AstraZeneca, Daiichi Sankyo, Boehringer Ingelheim, Amgen, Chugai, Genmab, GSK, and Incyte; Financial Interests, Personal, Funding: Taiho, Ono, Bayer, Lilly, Bristol Myers Squibb, and Merck Serono; Financial Interests, Personal, Advisory Role: Taiho, Takeda, and Amgen. J. Daniel, V. Ebiana, J. Howe, S. Spitz: Financial Interests, Personal, Full or part-time Employment: Incyte Corporation; Financial Interests, Personal, Stocks or ownership: Incyte Corporation. A. Italiano: Financial Interests, Personal, Advisory Board: AstraZeneca, Bayer, BMS, MSD, Parthenon, and Roche; Financial Interests, Institutional, Research Funding: AstraZeneca, Bayer, BMS, MSD, PharmaMar, and Roche. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

156TiP - The LUNGVAC-study; A randomized phase II, open-label, multicenter study investigating efficacy and safety of anti-PD-1/PD-L1 treatment +/- UV1 vaccination as first line treatment in patients with inoperable advanced or metastatic non-small cell lung cancer (NSCLC)

Presentation Number
156TiP
Lecture Time
05:00 - 09:15
Speakers
  • Elin Marie Stensland (Drammen, Norway)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Programmed Death Ligand 1 (PD1) inhibitors are indicated as monotherapy in first line in patients with stage IIIB/C and IV NSCLC with PD-L1 expression of 50% or above and no EGFR mutation or ALK translocation. Still, under 50% of these patients respond to Immune Checkpoint Inhibitor (ICI) treatment, and there is a need to increase the fraction of patients benefiting from ICI treatment. UV1 is a therapeutic peptide-based cancer vaccine targeting human telomerase (hTERT). hTERT is essential for tumor growth, expressed at high levels in 85% of human tumors, but only sparsely expressed in normal tissues. UV1 induces the expansion of CD4 T cells that recognize specific sequences in the UV1 peptides, and essentially initiates an anti-tumor immune response. UV1 is combined with ICI based on a presumed synergistic activity between the two modalities, as ICI blocks inhibitory signals for a vaccine-induced T cell expansion and anti-tumor effector activity.

Trial design

The LUNGVAC-study (NCT05344209) is a randomized phase II, open-label, multicenter study evaluating efficacy and safety of anti-PD-1 treatment with or without UV1 vaccination in treatment-naïve patients with advanced or metastatic NSCLC, with PD-L1 ≥ 50%. At least one measurable lesion according to Recist 1.1, adequate organ function, ECOG performance status 0-2 and no other active cancer are main eligibility criteria. Stratification factors are squamous versus non-squamous, and ECOG 2 versus 0+1. Primary endpoint is progression free survival. To test the PFS null hypothesis with 80% power and a 1-sided alpha level of 0.10, a total of 97 PFS events are required. Based on data published for pembrolizumab monotherapy in KEYNOTE-024, to generate the required 97 PFS events, 138 patients will be randomized 1:1 to PD-1-inhibitor for a maximum of 2 years, with or without 8 injections with UV1 vaccine during the first 2 months. Inclusion time is estimated to be 18 months and patients will be followed thereafter for a minimum of 18 months. 20 patients are included as of September 2023.

Clinical trial identification

NCT05344209, EudraCT 2021-005729-25.

Legal entity responsible for the study

Drammen Hospital, Vestre Viken Health Trust.

Funding

Ultimovacs.

Disclosure

E.M. Stensland: Financial Interests, Personal, Advisory Board, AdBoard NSCLC, June 21., 2022: Sanofi. O.T. Brustugun: Financial Interests, Institutional, Advisory Board: MSD, Roche, Takeda, AstraZeneca, Novartis, BMS, Janssen; Financial Interests, Institutional, Funding: Amgen, Ultimovacs; Financial Interests, Institutional, Research Grant: AstraZeneca, Pfizer, Roche. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

157TiP - Krascendo-170 Lung: a phase Ib/II study of divarasib + pembrolizumab _ platinum-based chemotherapy and pemetrexed in untreated KRAS G12C+ advanced non-small cell lung cancer (NSCLC)

Presentation Number
157TiP
Lecture Time
09:15 - 13:30
Speakers
  • Ferdinandos Skoulidis (Houston, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The KRAS G12C mutation, present in ∼12% of NSCLC patients, drives oncogenic signalling and cancer formation and is associated with poor prognosis. The current first-line treatment for advanced KRAS G12C+ NSCLC is checkpoint inhibitor (CPI) ± chemotherapy (CT). Novel combinations using a more targeted, biomarker-directed approach are supported by pre-clinical evidence and may further improve outcomes. Divarasib is an oral KRAS G12C inhibitor with potent pre-clinical and clinical anti-tumour activity. We hypothesize that divarasib + CPI ± CT may improve outcomes for patients with KRAS G12C+ NSCLC.

Trial design

Krascendo-170 Lung (NCT05789082) is a phase Ib/II, open-label study evaluating the safety and activity of divarasib + pembrolizumab in patients with PD-L1 tumour cell expression ≥1% (Cohort A) and of divarasib + pembrolizumab with platinum-based CT and pemetrexed in patients with any PD-L1 tumour cell expression level (Cohort B). Patients must be ≥18 years old with untreated unresectable/metastatic non-squamous NSCLC (measurable per RECIST v1.1), a confirmed KRAS G12C mutation, and an Eastern Cooperative Oncology Group performance status 0/1. Each cohort will have two stages: divarasib combination dose finding and dose expansion, with two planned dose levels of divarasib (Table). Tumour assessments will be performed at baseline and every 6 weeks for 48 weeks, then every 9 weeks thereafter. Plasma samples will be taken at various timepoints before and after divarasib and pembrolizumab dosing to characterise pharmacokinetics. Patients will be treated until disease progression per RECIST v1.1 or unacceptable toxicity. The co-primary endpoints are adverse events and change from baseline in targeted safety parameters. Key secondary endpoints include objective response rate, progression-free survival and duration of response (all investigator assessed per RECIST v1.1).

Cohort PD-L1 tumour cell expression Dose finding* Dose expansion†
A ≥1% Divarasib + Pembrolizumab Divarasib + Pembrolizumab
B Any Divarasib + Pembrolizumab + CT + Pemetrexed Divarasib + Pembrolizumab + CT + Pemetrexed

*Participants will receive one of two doses of divarasib. †Dose expansion will depend on pre-specified safety parameters during the dose-finding stage; CT: 4 cycles of carboplatin (intravenously, every 3 weeks, AUC5) or cisplatin (intravenously, every 3 weeks, 75 mg/m2) per investigator’s choice. Drug administration schedule: Divarasib: orally once daily; Pemetrexed: 500 mg/m2 intravenously every 3 weeks; Pembrolizumab: 200 mg intravenously every 3 weeks.

Clinical trial identification

NCT05789082.

Editorial acknowledgement

Medical writing support for the development of this abstract, under the direction of the authors, was provided by Neave Baldwin, BSc, of Ashfield MedComms, an Inizio company.

Legal entity responsible for the study

F. Hoffmann-La Roche Ltd.

Funding

F. Hoffmann-La Roche Ltd.

Disclosure

F. Skoulidis: Financial Interests, Personal, Advisory Board: AstraZeneca, Amgen Inc, Revolution Medicines, Novartis, BridgeBio, BeiGene, BergenBio, Guardant Health, Tango Therapeutics, Calithera Bioscieces, Hookipa Pharma, Novocure, Merck &Co; Financial Interests, Personal, Invited Speaker: ESMO, Japanese Lung Cancer Society, Medscape LLC, Intellisphere LLC, VSPO McGill Universite de Montreal, RV Mais Promocao Events LTDS, MJH Life Sciences, IDEOlogy Health, MI&T, PER LLC, CURIO LLC, DAVA Oncology, American Association for Cancer Research, IASLC; Financial Interests, Personal and Institutional, Local PI: Amgen Inc, AstraZeneca, Revolution Medicines, Novartis, Merck &Co, Tango Therapeutics, Genentech/Roche ; Financial Interests, Institutional, Research Funding: Revolution Medicines, Mirati Therapeutics, Amgen Inc, Novartis, Merck & Co; Financial Interests, Personal, Speaker, Consultant, Advisor: AstraZeneca, Amgen Inc, Revolution Medicines, Novartis, BridgeBio, BeiGene, BergenBio, Guardant Health, Tango Therapeutics, Calithera Bioscieces, Hookipa Pharma, Novocure, Merch &Co; Financial Interests, Personal, Steering Committee Member: AstraZeneca; Financial Interests, Personal, Stocks or ownership: BioNTech SE, Moderna Inc; Financial Interests, Personal, Trial Chair: Pfizer . K. Cuppens: Financial Interests, Personal, Expert Testimony: AstraZeneca, Merck Sharp Dohme; Financial Interests, Personal, Advisory Board: AstraZeneca, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, F. Hoffmann-La Roche Ltd, Merck Sharp Dohme; Financial Interests, Personal, Invited Speaker: Bristol Myers Squibb, F. Hoffmann-La Roche Ltd, Merck Sharp Dohme, Pfizer. A. Sacher: Financial Interests, Institutional, Coordinating PI: Genentech-Roche, BMS, AstraZeneca; Financial Interests, Institutional, Local PI: Amgen, Iovance, CRISPR Therapeutics, Merck, Pfizer, GSK, Spectrum, Lilly. V. Velcheti: Financial Interests, Personal, Speaker, Consultant, Advisor: BMS, Merck, AstraZeneca, Regeneron. D.H. Lee: Financial Interests, Personal, Speaker, Consultant, Advisor: Abion, AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly, ChongKeunDang, Janssen, MSD, Novartis, Ono, Pfizer, Roche, ST Cube, AbbVie, Takeda, Blueprint Medicine, BC World Pharm, Yuhan; Non-Financial Interests, Personal, Steering Committee Member: Roche, Abion. M.T. Lin, T.M. Fernando: Financial Interests, Personal, Full or part-time Employment: Genentech, Inc.; Financial Interests, Personal, Stocks/Shares: F. Hoffmann-La Roche Ltd. S. Li, M.S. Mathisen: Financial Interests, Personal, Full or part-time Employment: Genentech, Inc. D. Bradley: Financial Interests, Personal, Full or part-time Employment: Roche Products Ltd. M. Zarak Crnkovic: Financial Interests, Personal, Full or part-time Employment: F. Hoffmann-La Roche Ltd.

Collapse
Poster Display (ID 34) Poster Display

159TiP - Two Phase 1 Studies Assessing the Safety and Efficacy of the Small Molecule Oral PD-L1 Inhibitor INCB099280 in Combination with Adagrasib (INCB 99280-204 [Study 204]) or Ipilimumab (INCB 99280-205 [Study 205]) in Adults with Advanced Solid Tumors

Presentation Number
159TiP
Lecture Time
13:30 - 17:45
Speakers
  • David Berz (Los Angeles, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

INCB099280, an oral programmed death ligand 1 (PD-L1) inhibitor, has shown acceptable safety and preliminary efficacy in advanced solid tumors in an ongoing phase 1 study (Prenen et al. SITC 2022). As combination treatment (tx) can enhance the antitumor activity of anti-PD-1 therapy, we plan to conduct 2 Phase 1, open label, multicenter studies to evaluate safety and efficacy of INCB099280 combination tx in adults with select solid tumors. Study 204 will evaluate INCB099280 with adagrasib, an inhibitor of KRASG12C, a common mutation in non-small cell lung cancer (NSCLC) and colorectal cancer (CRC). Study 205 will evaluate INCB099280 with ipilimumab (IPI), a CTLA4 inhibitor that has shown synergistic antitumor activity with anti PD-1 agents in clinical studies.

Trial design

Eligibility criteria and dosing are outlined in Table. Both studies will consist of Part 1 dose-escalation and Part 2 dose-expansion. Part 1 will follow a hybrid of modified toxicity probability interval type design and dose-toxicity model. In Part 2, for Study 204, patients (pts) in NSCLC and CRC tumor-specific cohorts will be randomized within each cohort to ≤3 selected recommended doses for expansion (RDEs); for Study 205, pts in renal cell (RCC) and hepatocellular carcinoma (HCC) cohorts will be randomized 1:1 within each cohort to 1 of 2 selected RDEs. Pts will be treated with INCB099280 for ≤2 years, followed by a 90-day safety follow-up period . Primary endpoints are dose-limiting toxicities, tx emergent adverse events (TEAEs), and TEAEs leading to dosing modifications. Secondary/exploratory endpoints will include INCB099280 pharmacokinetic parameters, objective response, disease control, and duration of response. Recruitment has started for both studies.

Eligibility criteria and dosing

Study INCB 99280-204 INCB 99280-205
Eligibility criteria Part 1 Previously treated KRASG12C-mutant advanced solid tumors (≤45 pts) Unresectable/metastatic cutaneous melanoma or HCC, advanced clear cell RCC, or centrally confirmed MSI-H/dMMR metastatic CRC (≤36 pts)
Part 2 Previously treated, KRASG12C inhibitor-naive KRASG12C-mutant advanced NSCLC/ CRC (≤80 pts) Immuno-/systemic tx-naive Unresectable/metastatic HCC or RCC (≤80 pts)
Combination drug regimen Adagrasib 400 mg BID Ipilimumab q3w for ≤4 doses, RCC & CRC: 1 mg/kg HCC & melanoma: 3 mg/kg

Clinical trial identification

NCT06039384, NCT05909995.

Editorial acknowledgement

Editorial assistance was provided by Emily Sun and Andrew Marson-Neep of Envision Pharma Group (Philadelphia PA, USA).

Legal entity responsible for the study

Incyte Corporation, Wilmington, DE.

Funding

Incyte Corporation, Wilmington, DE.

Disclosure

D. Berz: Financial Interests, Personal, Advisory Board: Biocept, Prelude Dx; Financial Interests, Personal, Speaker’s Bureau: Caris Life Sciences, Tempus, Natera, Biocept, Boehringer Ingelheim, Genentech, Novartis, AstraZeneca, Sun, and Oncocyte. D.J. Pinato: Financial Interests, Personal, Speaker, Consultant, Advisor: ViiV Healthcare, Bayer Healthcare, Roche, Mursla, MiNa Therapeutics, Eisai, H3B, AstraZeneca, DaVolterra, Exact Sciences, Ipsen, Avamune, and Lift Biosciences; Financial Interests, Institutional, Research Grant: Bristol Myers Squibb, GSK, and MSD. M. Barve: Financial Interests, Personal, Full or part-time Employment: Texas Oncology; Financial Interests, Personal, Stocks or ownership: Texas Oncology; Financial Interests, Personal, Research Funding: Mary Crowley Research Center. J. Pulini, J. Bowman: Financial Interests, Personal, Full or part-time Employment: Incyte Corporation; Financial Interests, Personal, Stocks or ownership: Incyte Corporation. M.L. Johnson: Financial Interests, Personal, Speaker, Consultant, Advisor: Genentech/Roche, AstraZeneca, Calithera Biosciences, Merck, Sanofi, Mirati Therapeutics, Ribon Therapeutics, AbbVie, GSK, Gritstone Bio, Janssen Oncology, Lilly, Amgen, Daiichi Sankyo, Eisai, Axelia Oncology, Black Diamond Therapeutics, CytomX; Financial Interests, Institutional, Research Funding: EMD Serono, Kadmon, Janssen, Mirati Therapeutics, Genmab, Pfizer, AstraZeneca, Stem CentRx, Novartis, Array BioPharma, Regeneron, Merck, Hengrui Pharmaceutical, Lycera, BeiGene, Tarveda Therapeutics, Loxo, AbbVie, Boehringer Ingelheim, Guardant Health, Da; Financial Interests, Personal, Other, Travel grant: AbbVie, AstraZeneca, Genentech, Incyte, Merck, Pfizer, Sanofi.

Collapse
Poster Display (ID 34) Poster Display

160TiP - Safety and Antitumor Activity of GEN1042 in Combination with Pembrolizumab _ Chemotherapy in Solid Tumors: Phase 2b Dose-Expansion Trial in Progress

Presentation Number
160TiP
Lecture Time
17:45 - 22:00
Speakers
  • Ignacio Melero (Pamplona, Spain)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

GEN1042 (DuoBody CD40x4-1BB) is a novel, bispecific, agonistic antibody that combines targeting and conditional activation of CD40 and 4-1BB on immune cells to enhance priming and (re)-activation of tumor-specific immunity. In the phase 1a dose-escalation part of this open-label, multicenter phase 1/2 trial (NCT04083599), single-agent GEN1042 showed biologic and early clinical activity with a manageable safety profile in patients with advanced solid tumors. Preclinical and preliminary clinical results suggest that as a bispecific antibody, GEN1042 can leverage conditional CD40 and 4-1BB agonism in combination with PD-(L)1 blockade ± chemotherapy (CTX) to improve the antitumor response. In the safety run-in and expansion cohorts, GEN1042 in combination with pembrolizumab (PEM) ± CTX was generally well tolerated (no dose-limiting toxicities) with encouraging early clinical activity, including complete responses, observed in patients with advanced/metastatic head and neck squamous cell carcinoma (HNSCC) receiving GEN1042 + PEM + CTX (Melero I, et al. ESMO IO 2022). Enrolment into the combination therapy expansion cohorts is continuing, with this abstract focused on the evaluation of GEN1042 + PEM + CTX in checkpoint inhibitor (CPI) treatment–naive patients with non-small cell lung cancer (NSCLC) and HNSCC.

Trial design

The expansion part includes parallel cohorts of patients with NSCLC and HNSCC. Eligible patients with NSCLC will receive GEN1042 + PEM ± CTX. Eligible patients with recurrent/metastatic HNSCC (PD-L1 combined positive score ≥1) will receive GEN1042 + PEM ± cisplatin or carboplatin + 5-fluorouracil. Primary endpoint (phase 2b) is objective response rate per RECIST v1.1. Secondary endpoints are duration of response, disease control rate, progression-free survival, overall survival, safety/tolerability, and pharmacokinetics. Combination therapy expansion cohorts are recruiting across several countries including Spain, US, and Denmark.

Clinical trial identification

NCT04083599; Release Date: September 10, 2019.

Editorial acknowledgement

Medical writing and editorial support were provided by Stephanie Phan, PharmD, from Peloton Advantage, an OPEN Health company, Parsippany, NJ, USA.

Legal entity responsible for the study

Genmab A/S.

Funding

Genmab A/S and BioNTech SE.

Disclosure

I. Melero: Financial Interests, Personal, Advisory Board: Agneus, Alligator Bioscience, AstraZeneca, BMS, BioLineRx, Boehringer Ingelheim, Boston Pharma, CRISPR Therapeutics, CatalYm GmbH, Crescendo Biologics, Curon BIopharmaceutical, EMD Serono, F-Star, Genentech, Hookipa Pharma, Genmab, Gossamer Bio, Highlight Therapeutics, HotSpot Therapeutics, Inc., ImmuneSensor Therapeutics, Inc., Janssen, MSD, Merus N.V., Monopteros Therapeutics, NOXXON Pharma AG, Numab, Phenomic Bio, Pieris Pharmaceuticals GmbH, Pierre Fabre, Roche, Sanofi, Third Rock Ventures, Senti Biosciences, Servier, Shattuck Labs; Financial Interests, Personal, Other, Consultant: Pharma Mar; Financial Interests, Institutional, Coordinating PI: AstraZeneca, Roche, BMS, Genmab, Alligator. E. Grande: Financial Interests, Personal, Invited Speaker: Adacap, AstraZeneca, Bristol Myers Squibb, Eisai, Eusa Pharma, IPSEN, Janssen, Lilly, Merck KGa, Pfizer, Roche, Dr. Reddy's; Financial Interests, Personal, Advisory Board: Astellas, Bayer, MSD, Novartis, Sanofi-Genzyme; Financial Interests, Institutional, Advisory Board: Caris Life Sciences, ONCODNA (Biosequence); Financial Interests, Institutional, Research Grant, Independent research grant: Astellas, AstraZeneca, Lexicon, MTEM/Threshold, Nanostring Technologies, Pfizer, Roche, Merck; Financial Interests, Institutional, Coordinating PI, Independent research grant: IPSEN; Non-Financial Interests, Personal, Other, AD BOARD member: ENETS; Non-Financial Interests, Personal, Member of Board of Directors: GETNE, GUARD Consortium, Grupo centro de Tumores Genitourinarios. M.L. Johnson: Financial Interests, Institutional, Other, Consulting: AbbVie, Amgen, Arcus Biosciences, Arrivent, Astellas, AstraZeneca, Axelia Oncology, Black Diamond, Calithera Biosciences, Daiichi Sankyo, EcoR1, Genentech/Roche, Genmab, Genocea Biosciences, GSK, Gritstone Oncology, Ideaya Biosciences, Immunocore, iTeos, Janssen, Jazz Pharmaceuticals, Merck, Mirati Therapeutics, Molecular Axiom, Novartis, Oncorus, Pyramid Biosciences, Regeneron Pharmaceuticals, Revolution Medicines, Sanofi-Aventis, SeaGen, Synthekine, Takeda Pharmaceuticals, Turning Point Therapeutics, VBL Therapeutics; Financial Interests, Institutional, Research Grant: AbbVie, Acerta, Adaptimmune, Amgen, Apexigen, Arcus Biosciences, Array BioPharma, Artios Pharma, AstraZeneca, Atreca, BeiGene, BerGenBio, BioAtla, Black Diamond, Boehringer Ingelheim, Bristol Myers Squibb, Calithera Biosciences, Carisma Therapeutics, Checkpoint Therapeutics, City of Hope National Medical Center, Corvus Pharmaceuticals, Curis, CytomX, Daiichi Sankyo, Dracen Pharmaceuticals, Dynavax, Lilly, Elicio Therapeutics, EMD Serono, EQRx, Erasca, Exelixis, Fate Therapeutics, Genentech/Roche, Genmab, Genocea Biosciences, GSK, Gritstone Oncology, Guardant Health, Harpoon, Helsinn Healthcare SA, Hengrui Therapeutics, Hutchinson MediPharma, IDEAYA Biosciences, IGM Biosciences, Immunitas Therapeutics, Immunocore, Incyte, Janssen, Jounce Therapeutics, Kadmon Pharmaceuticals, Kartos Therapeutics, Loxo Oncology, Lycera, Memorial Sloan-Kettering, Merck, Merus, Mirati Therapeutics, Mythic Therapeutics, NeoImmune Tech, Neovia Oncology, Novartis, Numab Therapeutics, Nuvalent, OncoMed Pharmaceuticals, Palleon Pharmaceuticals, Pfizer, PMV Pharmaceuticals, Rain Therapeutics, RasCal Therapeutics, Regeneron Pharmaceuticals, Relay Therapeutics, Revolution Medicines, Ribon Therapeutics, Rubius Therapeutics, Sanofi, Seven and Eight Biopharmaceuticals/Birdie Biopharmaceuticals, Shattuck Labs, Silicon Therapeutics, Stem CentRx, Syndax Pharmaceuticals, Takeda Pharmaceuticals, Tarveda, TCR2 Therapeutics, Tempest Therapeutics, Tizona Therapeutics, TMUNITY Therapeutics, Turning Point Therapeutics, University of Michigan, Vyriad, WindMIL Therapeutics, Y-mAbs Therapeutics. J.R. Bauman: Financial Interests, Personal, Advisory Board: Janssen, Blueprint Medicines, Merck, BeiGene, Turning Point, Pfizer, Mirati; Financial Interests, Personal, Speaker, Consultant, Advisor: Lilly. V. Moreno Garcia: Financial Interests, Personal, Advisory Board: BMS, Janssen, Roche, Basilea, Bayer, AstraZeneca; Financial Interests, Personal, Full or part-time Employment: START; Financial Interests, Institutional, Local PI, AbbVie, AceaBio, Adaptimmune, ADC Therapeutics, Aduro, Agenus, Amcure, Amgen, Astellas, AstraZeneca Bayer BeiGene BioInvent International AB, BMS, Boehringer, Boheringer, Boston, Celgene, Daiichi Sankyo, DEBIOPHARM ,Eisai, e-Terapeutics, Exelisis, Forma Therapeutics, Genmab, GSK, Harpoon, Hutchison, Immutep, Incyte, Inovio, Iovance, Janssen, Kyowa Kirin, Lilly, Loxo, MedSir, Menarini, Merck, Merus, Millennium, MSD, Nanobiotix, Nektar, Novartis, Odonate Therapeutics, Pfizer, Pharma Mar, PharmaMar, Principia, PsiOxus, Puma, Regeneron, Rigontec, Roche, Sanofi, Sierra Oncology, Synthon, Taiho, Takeda, Tesaro, Transgene, Turning Point Therapeutics, Upshersmith.: Multiple. D.R. Adkins: Financial Interests, Personal, Speaker, Consultant, Advisor: Merck, Cue Biopharma, Blueprint Medicine, Exelixis, Immunitas, Kura Oncology, Targimmune Therapeutics, TwoXAR, Vaccinex, Xilio Therapeutics, Boehringer Ingelheim, Eisai Europe, Coherus Biosciences, Gilead Sciences, Jazz Pharmaceuticals; Financial Interests, Personal, Principal Investigator: Hookipa, Pfizer, Eli Lilly, Merck, Celgene/BMS, Novartis, AstraZeneca, Blueprint Medicine, Kura Oncology, CUE Biopharma, Cofactor Genomics, Debiopharm, ISA Pharmaceuticals, Gilead Sciences, BeiGene, Roche, Vaccinex, Adlai Nortye, BioAtla, Calliditas, Epizyme, Natco, Immutep, Tizona, Genmab, Boehringer Ingelheim. M.J. De Miguel: Financial Interests, Personal, Speaker, Consultant, Advisor: Roche, MSD, Janssen. S. Gattoni-Celli: Financial Interests, Personal, Full or part-time Employment: BioNTech; Financial Interests, Personal, Stocks/Shares: BioNTech. I. Celik: Financial Interests, Personal, Full or part-time Employment: BioNTech; Financial Interests, Personal, Stocks/Shares: BioNTech. J. Steinberg, T.J. Ong: Financial Interests, Personal, Full or part-time Employment: Genmab; Financial Interests, Personal, Stocks/Shares: Genmab. E. Felip: Financial Interests, Personal, Advisory Board: AbbVie, Amgen, AstraZeneca, Bayer, BeiGene, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly, F. Hoffmann-La Roche, Gilead, GSK, Janssen, Merck Serono, Merck Sharp & Dohme, Novartis, Peptomyc, Regeneron, Sanofi, Takeda, Turning Point, Pfizer; Financial Interests, Personal, Invited Speaker: Amgen, Daiichi Sankyo, Genentech, Janssen, Medical Trends, Medscape, Merck Serono, PeerVoice, Pfizer, Sanofi, Takeda, Touch Oncology, AstraZeneca, Bristol Myers Squibb, Eli Lilly, F. Hoffmann-La Roche, Merck Sharp & Dohme; Financial Interests, Personal, Member of Board of Directors, Independent member: Grifols; Financial Interests, Institutional, Local PI, Clinical Trial: AstraZeneca AB, AbbVie, Amgen, Bayer Consumer Care AG, BeiGene, Boehringer Ingelheim GmbH, Bristol Myers Squibb International Corporation, Daiichi Sankyo Inc., Exelixis Inc., F. Hoffmann-La Roche Ltd., Genentech Inc., GSK Research and Development Limited, Janssen Cilag International NV, Merck Sharp & Dohme Corp, Merck KGAA, Mirati Therapeutics Inc, Novartis Pharmaceutica SA, Pfizer, Takeda Pharmaceuticals International; Non-Financial Interests, Personal, Leadership Role, President (2021-2023): SEOM (Sociedad Espanola de Oncologia Medica); Non-Financial Interests, Personal, Member, Member of Scientific Committee: ETOP (European Thoracic Oncology Platform); Non-Financial Interests, Personal, Member, Member of the Scientiffic Advisory Committee: CAC Hospital Universitari Parc Taulí. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

161TiP - A Phase 1/2, Open-label Study of an Anti-ILT2 (LILRB1) Antibody, SAR444881, Administered Alone and in Combination With Pembrolizumab, With or Without Chemotherapy, or Cetuximab in Patients With Advanced Solid Tumors

Presentation Number
161TiP
Lecture Time
22:00 - 02:15
Speakers
  • Ruth Perets (Haifa, Israel)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immunoglobulin-like transcript-2 (ILT2), also known as LILRB1, an inhibitory receptor expressed on various immune cells, binds to classical and nonclassical major histocompatibility complex class-I (MHC-I), with highest affinity to human leukocyte antigen-G (HLA-G). HLA-G is expressed on various solid tumors and the interaction of ILT2 with HLA-G was found to impair phagocytosis, cytotoxicity, and cytokine secretion of macrophages, natural killer (NK) cells, and T cells. SAR444881 (BND-22), a novel humanized immunoglobulin G4 monoclonal antibody (mAb), selectively binds to ILT2 and blocks its interaction with MHC-I on tumor cells and may restore immune cell functions. (Mandel I, et al. J Immunother Cancer. 2022) In preclinical studies, SAR444881 alone and in combination with an anti-programmed cell death protein-1 mAb demonstrated enhanced anti-tumor activities of macrophages, NK cells, and T cells. (Mandel I, et al. Cancer Res. 2020).

Trial design

This is a Phase 1/2, open-label, multicenter, dose escalation, optimization, and expansion study evaluating safety, tolerability, and anti-tumor activity of intravenous (IV) SAR444881 as monotherapy and in combination with pembrolizumab, with or without chemotherapy, or cetuximab in patients with advanced solid tumors (NCT04717375). Study has 2 parts: dose escalation (Part 1; n=156) and dose optimization/expansion (Part 2; n=300). Part 1 has three sub-parts - SAR444881 monotherapy (Part 1A; IV, every two weeks [Q2W]), SAR444881 + pembrolizumab (Part 1B; IV, Q3W), and SAR444881 + cetuximab (Part 1C; IV, Q2W). Primary objectives for Part 1 are safety and tolerability and determining the maximum tolerated dose or maximum administered dose of SAR444881 monotherapy or its combinations and for Part 2 are preliminary anti-tumor activity and identification of SAR444881 optimal dose with its combinations. Key secondary objectives include anti-tumor activity, pharmacokinetics, immunogenicity, safety, and tolerability. Study is accruing patients in the United States and Israel. Study opened on March 2021 and has enrolled 71 patients as of August 2023.

Editorial acknowledgement

Medical writing support was provided by Parag Betkar of Sanofi.

Legal entity responsible for the study

Sanofi.

Funding

Sanofi.

Disclosure

R. Perets: Financial Interests, Personal, Other, Consultant: Galmed Therapeutics, Gilboa Therapeutics, 1E Therapuetics; Financial Interests, Institutional, Local PI: Jannsen, MSD, BMS, Genentech, Amgen, AbbVie, Ammune; Financial Interests, Institutional, Coordinating PI: Biomica; Non-Financial Interests, Institutional, Product Samples, Antibody for research: AbbVie. S.M. Stemmer: Financial Interests, Personal, Research Grant: CAN-FITE, AstraZeneca, BiolineRx, BMS, Halozyme, Clovis Oncology, CTG Pharma, Exelixis, Geicam, Incyte, Lilly, Moderna, Teva Pharmaceuticals and Roche; Financial Interests, Personal, Stocks/Shares: CTG Pharma, DocBoxMD, TyrNovo, VYPE, Cytora and CAN-FITE. R. Geva: Financial Interests, Personal, Advisory Board: Medison, Roche, Janssen, MSD, and Pfizer; Financial Interests, Personal, Advisory Role: Eisai, AstraZeneca, Bayer, MSD, BOL Pharma, Ranium, JNJ, and Roche; Financial Interests, Personal, Other, travel and accommodation expenses: Takeda, Pyxis; Financial Interests, Personal, Other, equity interests: BOL Pharma and Pyxis Oncology. T. Golan: Financial Interests, Personal, Research Grant: AstraZeneca; Financial Interests, Personal, Other, Received honoraria or consultation fees: AbbVie, MSD Merck, Teva; Financial Interests, Personal, Other, Received receipt of speakers bureau: AbbVie. M. Fakih: Financial Interests, Personal, Research Grant: Amgen, AstraZeneca, Novartis; Financial Interests, Personal, Other, Personal Fees: Amgen, AstraZeneca, Novartis; Financial Interests, Personal, Advisory Role: Array, Bayer, GSK, Taiho, Incyte, and Pfizer. J. Cohen: Financial Interests, Personal, Other, Received honoraria: AstraZeneca, Roche, Medison Pharma; Financial Interests, Personal, Advisory Board, Travel and accommodation: Medison Pharma. Z. Jin: Financial Interests, Personal, Other, Consulting or Advisory Role: Novartis, QED Therapeutics, Lilly, GSK, Daichi Sankyo/AstraZeneca. P. Lorusso: Financial Interests, Personal, Advisory Board: AbbVie, Genmab, Genentech, CytomX, Takeda, Cybrexa, Agenus, IQVIA, TRIGR, Pfizer, ImmunoMet, Black Diamond, Astellas, GSK, QED Therapeutics, AstraZeneca, EMD Serono, Shattuck, Salarius, Silverback, MacroGenics, Kyowa Kirin, Kineta, Bayer, Zentalis, Molecular Templates, ABL Bio, STCube, Relay Therapeutics, Stemline, Compass BADX, BAKX Therapeutics, Scenic Biotech, Qualigen, Roivant Sciences, NeuroTrials, Seagen, imCheck, Mekanist, Mersana; Financial Interests, Personal, Other, Data Safety Monitoring Board: Agios, Five Prime, Halozyme; Financial Interests, Personal, Other, imCORE Alliance: Roche-Genentech; Financial Interests, Personal, Other, Consultant: Sotio, SK Life; Financial Interests, Personal, Other, Data Safety Monitoring Committee: Tyme; Financial Interests, Personal, Stocks/Shares: BAXK; Financial Interests, Institutional, Local PI: AbbVie, ADC Therapeutics, Boehringer Ingelheim, ALX Oncology, Astellas Pharma, Astex Pharmaceuticals, AstraZeneca, Bayer, Black Diamond, Calico Life Sciences, Corvus Pharmaceuticals, CytomX Therapeutics, Eisai Pharmaceuticals, Eli Lilly, EMD Sernono, Five Prime, FLX Bio, F-Star Delta Limited, Genentech, MedImmune, Genmab, Incyte, Linnaeus Therapeutics, Merck Sharp & Dohme, Moderna Therapeutics, NextCure, Pfizer, Takeda, Ribon Therapeutics, Sotio, Stemline Therapeutics, Tesaro, Jounce; Financial Interests, Personal, Advisory Board, Advisory Board & Consultant: I-Mab; Non-Financial Interests, Personal, Other, AACR Methods in Clinical Cancer Research Workshop - Co-Director: American Association for Cancer Research; Non-Financial Interests, Personal, Other, AACR Annual Report Committee - Member: American Association for Cancer Research; Non-Financial Interests, Personal, Other, Continuing Medical Education Committee - Member: American Association for Cancer Research; Non-Financial Interests, Personal, Other, Molecular Cancer Therapeutics Editorial Board - Member: American Association for Cancer Research; Non-Financial Interests, Personal, Other, ASCO Conquer Cancer Young Investigator Award Grand Selection Committee - Member: American Society of Clinical Oncology; Non-Financial Interests, Personal, Other, Chair - Phase 0 Task Force: American Association of Cancer Research; Non-Financial Interests, Personal, Other, AACI Clinical Research Innovation Steering Committee - Member: Association of American Cancer Institutes; Non-Financial Interests, Personal, Other, New Drugs in Oncology Seminar Planning Committee - Member: American Society of Clinical Oncology; Non-Financial Interests, Personal, Other, New Agents Committee: Translational Research Panel - Chair: Cancer Research Unite Kingdom; Non-Financial Interests, Personal, Other, Scientific Advisory Board - Member: Targeted Anti-Cancer Therapies; Non-Financial Interests, Personal, Member: American Association for Cancer Research, ASCO; Other, Personal, Other, Investigational Drug Steering Committee - Committee Member: National Cancer Institute; Other, Personal, Other, Phase I Special Emphasis Panel - Grant Reviewer/Discussion Leader: National Cancer Institute; Other, Personal, Other, NeXT Special Emphasis Panel - Grant Reviewer: National Cancer Institute/National Institute of Health; Other, Personal, Other, Board of Scientific Counselors, Clinical Sciences & Epidemiology: National Cancer Institute; Other, Personal, Other, Young Investigator Meeting, Cancer Therapy Evaluation Program - Professor: National Cancer Institute; Other, Personal, Other, Academic Advisory Board SPORE GI Malignancies - Case Western Reserve University: Case Western Reserve University; Other, Personal, Other, Scientific External Advisory Board - Member: University of California at San Diego; Other, Personal, Other, External Advisory Board - Member: University of Arizona; Other, Personal, Other, External Scientific Advisory Board - Member: University of New Mexico. N. Ashtamker, I. Friedman, M. Hakim: Financial Interests, Personal, Other, Employed by Biond Biologics: Biond Biologics; Financial Interests, Personal, Stocks/Shares: Biond Biologics. N. Crawford, R. Perez, M. Agarwal, G. Abbadessa, J. Lin, C. Deantonio: Financial Interests, Personal, Other, Employed by Sanofi: Sanofi; Financial Interests, Personal, Stocks/Shares: Sanofi. Financial Interests, Personal, Stocks/Shares: Sanofi. M. Wu: Financial Interests, Personal, Other, Employed by Sanofi: Sanofi; Financial Interests, Personal, Stocks or ownership: Sanofi. M. Borad: Financial Interests, Institutional, Other, Received grant to institution: Senhwa Pharmaceuticals, Adaptimmune, Agios Pharmaceuticals, Halozyme Pharmaceuticals, Five Prime Pharmaceuticals, Celgene Pharmaceuticals, EMD Merck Serono, Toray, Dicerna, Taiho Pharmaceuticals, Sun Biopharma, Isis Pharmaceuticals, Redhill Pharmaceutical; Financial Interests, Personal, Other, travel support: AstraZeneca. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

164P - Disentangling the Joint and Distinct Immunomodulation and Vulnerability Between KEAP1/NFE2L2 and SMARCA4 Alterations in Lung Adenocarcinoma

Presentation Number
164P
Lecture Time
02:15 - 02:15
Speakers
  • Anlin Li (Guangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

KEAP1 and SMARCA4 mutations are two extensively studied alterations in lung adenocarcinoma (LUAD). Recent studies have proposed that SMARCA4 mutations can mimic the transcriptional profile of KEAP1/NFE2L2 mutations, suggesting they might share mutual targets. However, SMARCA4 mutations have been shown to confer a poor prognosis and affect immunotherapy efficacy, independent of KEAP1 status. Thus, both mutations conceivably have non-overlapping effects on the tumor microenvironment, but their joint and distinct influences are unclear.

Methods

Using 585 LUAD patients, we conducted differential expressed gene (DEG) and enrichment analyses separately for KEAP1/NFE2L2-MUT and SMARCA4-MUT tumors vs their WT counterparts. Only pathogenic mutations were considered. To avoid confounding effects, tumors with SMARCA4 mutations were excluded when analyzing KEAP1/NFE2L2 mutations, and vice versa. Tumors with EGFR mutations were also excluded, as the two mutations were enriched in EGFR-WT tumors.

Results

The KEAP1/NFE2L2 and SMARCA4 mutations shared upregulated DEGs from the aldo/keto reductase superfamily (AKR1C1, AKR1C2, AKR1C3, AKR1C4). These genes are activated by the KEAP1/NFE2L2 pathway and execute cytoprotective and stress responses. Meanwhile, a larger proportion of their DEGs were mutation-specific (Up/Down: KEAP1/NFE2L2, 77.2%/82.6%; SMARCA4, 87.6%/88.7%). The complement/coagulation cascades and proliferation signatures were activated in SMARCA4-MUT tumors but not in KEAP1/NFE2L2-MUT tumors. The depletion of antigen processing and presentation, leukocyte migration, and B cell-related functions were observed in KEAP1/NFE2L2-MUT tumors. Conversely, SMARCA4 mutations did not recapitulate these critical immune depletions.

Conclusions

The KEAP1/NFE2L2-based cytoprotective activity is a joint feature of the two mutations, suggesting that they might both benefit from cytoprotective inhibitors, such as a glutaminase inhibitor. KEAP1/NFE2L2-MUT tumors showed various immunological abnormalities, highlighting the need for identifying targets to reinvigorate the immune response. SMARCA4-MUT tumors may be vulnerable to complement-targeted therapy.

Legal entity responsible for the study

A. Li.

Funding

This study was supported by the National Natural Science Foundation of China (81972898 and 82172713), the Natural Science Foundation of Guangdong Province (2023B1515020008), and the Fundamental Research Funds for the Central Universities, Sun Yat-sen University (22ykqb15).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

165P - Immunosuppressive F13A1+ Mo/M_ in the tumor microenvironment as a hallmark for multiple primary lung cancers

Presentation Number
165P
Lecture Time
02:15 - 02:15
Speakers
  • Jiahao Qu (Shenzhen, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The increasing number of patients with multiple primary lung cancers (MPLCs) endows a rising challenge for clinical management. The tumor microenvironment (TME) has a critical role in MPLCs but its contribution to cancer onset remains unclear. Further explorations of TME via single cell transcriptomics would allow for discovery of diagnostic biomarkers and potential therapeutic targets for MPLCs, differing from solitary primary lung cancers (SPLCs).

Methods

We profiled 13 tumors and paired paracancerous samples from 5 MPLC and 3 SPLC patients using single-cell RNA sequencing (scRNA-seq). Seurat was used for dimensional reduction, cluster identification and cell type annotation. Myeloid clusters were analyzed for their marker gene expression and tissue distribution. Cell-cell communication was examined using CellChat. Results were validated in scRNA-seq datasets from independent external cohorts (GSE200972 and E-MTAB-6149), including 25 samples from 4 MPLCs and 3 SPLCs.

Results

We classified 79,944 Monocytes/Macrophages (Mo/Mϕ) into 5 clusters based on the expression of FABP4, F13A1, FCN1, LYVE1 and MKI67. Of note, the F13A1+ Mo/Mϕ subset is preferentially enriched in MPLC tumors rather than normal tissues or SPLC tumors. They display a transcriptional profile resembling M2-like Mϕs and SPP1-expressing Mϕs that are pro-tumor and associated with poor prognosis. They also express high levels of anti-inflammatory markers and low levels of pro-inflammatory genes. CellChat analysis revealed a strong interaction between F13A1+ Mo/Mϕ and NK/T cells in MPLCs, but not SPLCs, via SPP1:CD44 and HLA-E:NKG2A axes, both of which have a negative impact on anti-tumor immunity. These findings were further validated in independent datasets.

Conclusions

We identified a previously undescribed F13A1+ Mo/Mϕ subpopulation preferentially enriched in MPLCs. Our data suggest that they could serve as a diagnostic biomarker to distinguish MPLCs from normal tissues and SPLCs. Crosstalk between F13A1+ Mo/Mϕ and immune cells through specific ligand-receptor interactions may contribute to immune suppression during cancer development, which could be a specific druggable target for MPLCs.

Legal entity responsible for the study

Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center.

Funding

National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen+SZ2020ZD011; Medical Scientific Research Foundation of Guangdong Province, China; Sanming Project of Medicine in Shenzhen (No. SZSM201612097) and Shenzhen Key Medical Discipline Construction fund (No. SZXK075).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

166P - Three-dimensional (3D) Innervation of Mouse Lungs and Airways in a Lung Metastatic Tumor Model

Presentation Number
166P
Lecture Time
02:15 - 02:15
Speakers
  • Yan Zhou (Shanghai, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Schwann cells (SCs) are a type of peripheral nerve system(PNS) glial cell that support the viability of myelinated and unmyelinated peripheral nerve fibers. Our previous studies identified SCs in human lung cancer specimens and demonstrated that SCs could increase the metastasis of lung cancer by activating the PI3K/AKT/GSK-3b/Snail-7wist signaling pathway and enhancing M2 macrophage polarization via CCL2 secretion. However, in the lung cancer mouse model, complete identification of airway nerves (or other cellular/subcellular objects) has not been possible due to patchy distribution and micron-scale size.

Methods

In this study, we describe a method using tissue clearing to acquire the first complete image of three-dimensional (3D) innervation in lung cancer. Mouse airways were harvested from the Metastatic tumor model of C57BI/6 mice and separated from the heart. The tissues were dehydrated, rehydrated and completely transparented. The specimens were labeled for the pan-neuronal marker PGP9.5 with a mouse antibody. All images were acquired using a confocal microscope. 3D images of fluorescently casted airways were processed to make airway masks. To quantify visceral pleural nerve distribution in mice, nerves were modeled using computer tractography and measured in different lung cancer regions.

Results

We found that nerves are widely distributed in both lung and lung tissues. The shape of peripheral nerves is similar to blood vessels, showing reticular shape along the bronchi and bronchioles. However, the distribution of peripheral nerves in the lung has not changed due to lung cancer. Interestingly and critically, the fluorescence intensity of the nerve near the side of lung cancer is much stronger, suggesting that the phenotype of peripheral nerves may change after interaction with lung cancer cells. The nerve growth in lung cancer tissue is abundant and active.

Conclusions

We first complete the image of three-dimensional (3D) innervation in lung cancer. The interaction between peripheral nerves and lung cancer may affect the phenotype of peripheral nerves and thus regulate the malignant biological course of lung cancer. PNS may play an oncogenic role in lung tumor progression and may serve as a therapeutic target for further analysis.

Legal entity responsible for the study

Shanghai Chest Hospital.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

167P - Lurbinectedin, a DNA minor groove inhibitor launches a multimodal immune response through activation of the cytosolic DNA-Sensing cGAS-STING pathway.

Presentation Number
167P
Lecture Time
02:15 - 02:15
Speakers
  • Triparna Sen (New York, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Small cell lung cancer (SCLC) is the most aggressive subtype of lung cancer and is accompanied by frequent primary or acquired resistance to first-line treatment of chemo-immunotherapy resulting in very modest clinical benefit. Lurbinectedin is FDA-approved as a second-line treatment for SCLC.

Methods

In this study, we evaluated the effect of lurbinectedin on the immune microenvironment in SCLC in multiple immunocompetent mouse models and performed multi-color flow cytometry and bulk RNA sequencing.

Results

In multiple immunocompetent SCLC mouse models, lurbinectedin treatment remarkably enhanced the anti-tumor effect of PD-L1 blockade in tumors that show primary resistance to immunotherapy. Moreover, lurbinectedin caused significant tumor regression in mice that had developed acquired resistance to chemoimmunotherapy treatment demonstrating its efficacy as a superior second-line treatment. Lurbinectedin treatment caused micronuclei formation and activated the cGAS/STING pathway, induced the expression of Type I and II interferon pathways, and caused significant tumor infiltration of cytotoxic and memory/effector T-cells. Similarly, pro-inflammatory M1 type macrophages and dendritic cells were increased, while immunosuppressive M2 type macrophages and MDSC cells were dramatically decreased. Interestingly, lurbinectedin also led to significant induction of MHC class I in SCLC in vitro and in vivo models. Analysis of pre-and post-treatment clinical samples in patients with relapsed SCLCs validated the induction of MHC class I and interferon pathway genes. Finally, pre-and post-lurbinectedin treatment demonstrated a STING-mediated increase in damage-associated molecular patterns (DAMPs) associated with immunogenic cell death.

Conclusions

We provide the first mechanistic insight into the lurbinectedin-induced cGAS-STING activation and multimodal immune modulation in SCLC. Lurbinectedin is already approved as a second-line agent in SCLC. Hence, our findings highlight lurbinectedin as a potentially transformative therapy for SCLCs that have relapsed from chemoimmunotherapy treatment, paving the way for combination clinical trials with anti-PD-L1.

Legal entity responsible for the study

The author.

Funding

Has not received any funding.

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

158TiP - MK-3475A-D77 Study: Phase 3, Randomized, Open-Label Study of First-Line (1L) Subcutaneous (SC) Pembrolizumab Coformulated With Hyaluronidase vs Intravenous (IV) Pembrolizumab With Chemotherapy in Metastatic Squamous or Nonsquamous NSCLC

Presentation Number
158TiP
Lecture Time
02:15 - 02:15
Speakers
  • Mehmet Ali Nahit Sendur (Ankara, Turkey)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

168P - Effect of sequence treatment of chemotherapy plus radiotherapy activates innate immunity in SCLC

Presentation Number
168P
Lecture Time
02:15 - 02:15
Speakers
  • CATERINA DE ROSA (Napoli, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

In the past decade, radiotherapy (RT) has been widely practised for treating extensive-stage small cell lung cancer (ES-SCLC). Despite this, it has been discouraged in phase III trials of first line chemoimmunotherapy. Instead, robust rationales have emerged in favour of using RT as a booster to promote a sustained anti-tumour immune response. On basis of published data defining a subtype of SCLC patients who respond to chemotherapy (CT) classified as \"inflamed\" and expressing high innate immune genes such as the STimulator of INterferon pathway (STING) pathway and based on further preclinical data supporting STING levels as potential biomarkers of response to combined chemo- and immunotherapy, we hypothesised that innate immune activation may provide an opportunity to evaluate anti-tumour immune activity in vitro.

Methods

We explored the landscape of STING, Mitochondrial antiviral-signaling protein (MAVS), Gamma-interferon-inducible protein (IFI-16) and immune-related cytokines expression both at mRNA and protein expression levels among two SCLC cell lines, namely H82 and H524. We then tested the effect of in vivo treatment with CT and RT (4 Gy dose) on PBMC subpopulations derived from SCLC patients by LDH cytotoxicity assay.

Results

We performed sequential in vitro treatment with CT and RT (4 Gy) in selected SCLC cell lines and we investigated changes in STING pathway, as indicator of DNA damage induced by CT and RT. cGAS, STING and downstream protein p-TBK1 and p-IRF3 were upregulated by CT plus RT in both cell lines. Interestingly, these increments were accompanied by high levels of DNA damage, as suggested by increased levels of H2A.X, ATM, ATR, and DNA-PK. IFI-16 DNA sensor was also increased at both protein and mRNA levels. Moreover, inflammatory T cells recruiting the chemokines IL6, IFN-β, CXCL5, CXCL10, were also significantly increased by sequential treatment with CT and RT. The SCLC patients-derived immune cells activity patients were also significantly increased post-RT as measured by LDH cytotoxicity assay.

Conclusions

We demonstrated in cellular models of SCLC a positive modulation of innate immune pathways with CT and RT sequential treatment, suggesting a possible boost role of RT in SCLC patients treated with chemoimmunotherapy.

Legal entity responsible for the study

The authors.

Funding

AIRC.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

169P - High-dimensional analysis of tumor infiltrating immune cells reveals major differences in the tumor immune microenvironments of pleural mesothelioma and lung cancer

Presentation Number
169P
Lecture Time
02:15 - 02:15
Speakers
  • Angelica Rigutto (Fribourg, Switzerland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Pleural mesothelioma (PM) and non-small cell lung cancer (NSCLC) are aggressive thoracic malignancies with different cells of origin, pathogenesis and genetic features. Despite both tumors are treated with chemotherapy and immunotherapy, patients with PM and NSCLC exhibit varying responses. The aim of this study is to characterize the differences in tumor infiltrating immune cell populations in PM and NSCLC using high-dimensional flow cytometry analysis.

Methods

Fresh-frozen tumors derived from 42 PM and 30 NSCLC patients with available clinical data were analyzed. Single-cell suspensions were stained with a panel of 33 markers and samples were acquired using a Spectral Analyzer. An R-based pipeline for the in-depth characterization of the immune cells subsets was developed. Machine learning methods were used for unsupervised phenotypes discovery and differential abundant analysis was carried out to identify differences between samples. Correlation analysis with available clinical data was performed.

Results

By using high-dimensional single-cell analysis, 18 tissue-resident and invading immune cells subsets within the tumor immune microenvironment (TIME) of PM and NSCLC samples were identified. The comparison between the two tumor entities reveled significant differences in the tumor landscape, with increased infiltration of CD4+ and CD8+ T cell subsets in NSCLC compared to PM. While NSCLC tumors share similar tumor infiltrating immune cell subsets, PM tumors cluster in three distinct subgroups. Specifically, the subgroup with decreased infiltration of B cells and increased presence of tumor-associated macrophages is characterized by a significant worse overall survival (6 vs 18 vs 27 months, p=0.00012).

Conclusions

Our findings demonstrate significant differences in the composition of the TIME between PM and NSCLC, especially in T cell abundance. These disparities offer insights into the varying responses to immunotherapy. Moreover, we highlight the pivotal role of the TIME in predicting PM patient outcomes. These findings may be crucial to optimize immunotherapy for these malignancies, potentially leading to improve patients' outcomes.

Legal entity responsible for the study

The authors.

Funding

Stiftung für Angewandte Krebsforschung (SAKF).

Disclosure

I. Opitz: Financial Interests, Personal, Invited Speaker: Roche, AstraZeneca; Financial Interests, Personal, Advisory Board: AstraZeneca, MSD, BMS; Financial Interests, Personal, Other, Proctorship: Intuitive; Financial Interests, Institutional, Research Grant: Medtronic, Roche. A. Curioni-Fontecedro: Financial Interests, Personal, Advisory Board: AstraZeneca, Bristol Meyer Squibb, Boehringer Ingelheim, MSD, Novartis, Amgen, Roche, Takeda, Janssen; Non-Financial Interests, Personal, Leadership Role: Swiss Academy for Clinical Cancer Research (SAKK); Non-Financial Interests, Personal, Principal Investigator, of clinical trials: Roche; Non-Financial Interests, Personal, Principal Investigator, Clinical Trials: Takeda, MSD, Bristol Meyer Squibb, Amgen. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

170P - Targeting myeloid cells in non-small cell lung cancer and hepatocellular carcinoma: a window-of-opportunity trial of nivolumab with BMS-813160 (CCR2/5i) or BMS-986253 (anti-IL8)

Presentation Number
170P
Lecture Time
02:15 - 02:15
Speakers
  • Nicholas J. Venturini (New York, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Monocyte-derived macrophages (mo-macs) and polymorphonuclear leukocytes (PMNs) are abundant in non-small cell lung cancer (NSCLC) and hepatocellular carcinoma (HCC), limiting response to immune checkpoint blockade (ICB) by promoting an immunosuppressive tumor microenvironment (TME). Preclinical work shows that tumor-derived CCR2 ligands and IL8 play a key role in recruiting mo-macs and PMNs, respectively, to the TME. Disrupting these signaling pathways augments ICB in mouse models, but clinical benefit has yet to be observed.

Methods

This phase IIa trial assessed the efficacy of BMS-813160 (CCR2/5i) or BMS-986253 (anti-IL8) administered with nivolumab (NIVO) over 4 weeks prior to resection. NSCLC patients were treated with NIVO and CCR2/5i (arm A) or NIVO and anti-IL8 (arm B). HCC patients were treated with NIVO (arm C), NIVO and CCR2/5i (arm D), or NIVO and anti-IL8 (arm E). Primary endpoints were major pathologic response (≤10% viable tumor) for NSCLC and significant tumor necrosis (>70% necrosis) for HCC. Secondary endpoints were safety/tolerability, time to surgery (TTS), and radiographic response.

Results

36 patients were enrolled from March 2020-August 2023; 14 were treated with NIVO and CCR2/5i, 16 with NIVO and anti-IL8, and 6 with NIVO. CCR2/5i and anti-IL8 were safe/tolerated (dose-limiting toxicities or grade 3/4 treatment-related adverse events). 32 of 36 patients underwent resection (mean TTS of 34.8 days). 3 patients met the primary endpoints: 2 in arm B and 1 in arm D. In patients treated with CCR2/5i, serum concentration of CCR2/5 ligands increased, and number of circulating monocytes decreased, after treatment. In patients treated with anti-IL8, serum concentration of IL8 decreased after treatment; however, number of circulating PMNs was not affected. Tissue analysis with scRNA-seq and multiplex imaging is ongoing to elucidate the biologic effects of these agents.

Conclusions

Although CCR2/5i and anti-IL8 appear to be biologically active and exert effects on chemokine levels, they fail to significantly augment the role of ICB in the preoperative setting, contrary to preclinical evidence.

Clinical trial identification

NCT04123379.

Legal entity responsible for the study

The authors.

Funding

CIMAC-CIDC, PACT.

Disclosure

S. Gnjatic: Financial Interests, Personal and Institutional, Funding, Unrelated to Current Work: Regeneron Pharmaceuticals, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Genentech, EMD Serono, Pfizer, and Takeda. M. Merad: Financial Interests, Personal, Advisory Board: Compugen Inc., Myeloid Therapeutics Inc., Morphic Therapeutic Inc., Asher Bio Inc., Dren Bio Inc., Nirogy Inc., Oncoresponse Inc., Owkin Inc., Larkspur Inc., Innate Pharma Inc., DBV Inc., Pionyr Inc., OSE Inc., Genenta Inc.; Financial Interests, Personal, Stocks/Shares: Compugen Inc., Myeloid Therapeutics Inc., Morphic Therapeutic Inc., Asher Bio Inc., Dren Bio Inc., Nirogy Inc., Oncoresponse Inc., Owkin Inc., and Larkspur Inc.; Financial Interests, Personal and Institutional, Funding: Regeneron Inc. and Boehringer Ingelheim Inc. T.U. Marron: Financial Interests, Personal, Advisory Board: Rockefeller University, Regeneron Pharmaceuticals, AbbVie, Bristol Meyers Squibb, Boehringer Ingelheim, Atara, AstraZeneca, Genentech, Celldex, Chimeric, Glenmark, Simcere, Surface, G1 Therapeutics, NGMbio, DBV Technologies, Arcus, and Astellas; Financial Interests, Personal and Institutional, Funding: Regeneron, Bristol Myers Squibb, Merck, and Boehringer Ingelheim. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

162TiP - Phase I, open-label, dose-escalation trial investigating the safety and efficacy of oncolytic virus BI 1821736 in patients with advanced solid tumors

Presentation Number
162TiP
Lecture Time
02:15 - 06:30
Speakers
  • Anthony W. Tolcher (San Antonio, United States of America)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

171P - Immune landscape and CLEVER-1 expression in hepatoblastoma

Presentation Number
171P
Lecture Time
06:30 - 06:30
Speakers
  • Ville Väyrynen (Helsinki, Finland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Hepatoblastoma (HB) is the most common primary liver malignancy among infants and young children. HBs are regarded as embryonal tumors arising from hepatic progenitor cells. Current treatment modalities for HB produce unsatisfactory results (20-30% respond poorly to interventions). To decrease adverse effects and suboptimal outcomes in patient care, research in immuno-oncology is investigating safer and more effective treatment options. Finding new targets for HB immunotherapeutics requires improved knowledge of the tumor immune microenvironment (TIME). In this study, our objective is to elucidate the TIME of HBs and investigate known targets of novel treatment options such as CLEVER-1, inhibition of which triggers M2 macrophage conversion towards M1 type and provokes an antitumoral response in the immune system.

Methods

Whole-slide multiplex immunofluorescence staining (mIHC) was utilized to assess the immunological characteristics of HB patient specimens (n=25) and normal liver samples (n=2). The mIHC panel chosen included antibodies against all macrophages, M2 macrophages, M1 macrophages, T-cells, and all leukocytes, as well as stellate cells, and epithelial cells. Antibody validation was performed to assess the consistency of staining of the targeted proteins in HB tissue. Image analyses were carried out using machine learning-assisted software (QuPath) to calculate the proportions of each immune cell type and assess the expression of potentially targetable proteins. QuPath was used to train an artificial intelligence model (AIM) for each marker. After automatic cell detection, the AIMs were applied to selected stained specimens, and cells positive for different markers were uncovered. Subsequently, spatial analyses regarding the proximity of immune cells to the tumor were performed.

Results

CLEVER-1-positive M2 macrophages are detected in HB tissue. Furthermore, preliminary results indicate that the staining is adequate for detecting other immune cell types within the specimens. Moreover, the AIMs show promising accuracy in automatically classifying immune cells.

Conclusions

HB is a cancer type with CLEVER-1-positive M2 macrophages and is thus a possible candidate for novel immunotherapies.

Legal entity responsible for the study

The authors.

Funding

Helsinki University Central Hospital Research Grants, Päivikki and Sakari SohlbergFoundation, Sigrid JuséliusFoundation, and Aamu Pediatric Cancer Foundation.

Disclosure

K. Eloranta, J. Lohi, S. Jalkanen: Financial Interests, Personal, Stocks/Shares: Faron Pharmaceuticals. J. Lohi: Financial Interests, Personal, Stocks/Shares: Faron Pharmaceuticals. M. Hollmén: Currently employed by and own shares of Faron Pharmaceuticals. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

172P - PLCE1 stabilizes ENO1 to enhance glycolysis in esophageal squamous cell carcinoma (ESCC) and induces an immune-suppressive tumor microenvironment

Presentation Number
172P
Lecture Time
06:30 - 06:30
Speakers
  • Ju Yang (nanjing, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

PLCE1 has been identified as a susceptibility gene for ESCC. However, the precise involvement of PLCE1 in glycolysis and its impact on anti-tumor immunity remain elusive.

Methods

We conducted an analysis of DEG) in ESCC cell lines following the silencing of PLCE1 using Affymetrix GeneChip technology. We employed IP-MS to identify molecules interacting with PLCE1, which are also associated with glycolytic processes. The mechanisms were further probed through a combination of IP assays, in vivo tumor growth experiments, and ubiquitination assays. To study the role of PLCE1 in glycolysis and its impact on anti-tumor immunity, we established an ESCC-induced model in C57BL/6 mice with the PLCE1-/- genotype, utilizing the carcinogen 4NQO.

Results

Bioinformatics analysis revealed that DEGs were significantly enriched in cell metabolism, particularly the glycolysis pathway. Knockdown of PLCE1 led to the suppression of glycolysis in ESCC cell lines through the regulation of ENO1 expression, a key enzyme in glycolysis. This effect was observed both in vitro and in vivo. Additionally, we have elucidated a novel pathway in which PLCE1 interacted with CDK2 and ENO1 to enhance the phosphorylation and stability of ENO1. Phosphorylation of ENO1 effectively prevented its ubiquitination and proteasome-mediated degradation, which was orchestrated by FBXW7-a recognized E3 ubiquitin ligase. In human ESCC tissues, we observed an increase in the population of CD8+ T cells in close proximity to PLCE1+ENO1+ tumor cells. A heightened accumulation of CD8+ PD1+ T cells was noted around these PLCE1+ENO1+ tumor cells. In an ESCC-induced mice model, a more pronounced infiltration of both CD4+ T cells and CD8+ T cells was observed in the PLCE1-/- genotype. Notably, T cells within the PLCE1-/- genotype exhibited heightened cytokine production and lower PD1 expression, an effect that was further potentiated by the ENO1 inhibitor.

Conclusions

Our study demonstrates that PLCE1 has the capacity to interact with CDK2 and ENO1, thereby counteracting FBXW7-mediated ubiquitination of ENO1. This intricate mechanism leads to an augmentation of glycolysis in ESCC and fosters an immune-suppressive tumor microenvironment.

Legal entity responsible for the study

The authors.

Funding

Natural Science Foundation of China.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

173P - Depleting resident peritoneal macrophages is an effective treatment for peritoneal metastasized colorectal cancer

Presentation Number
173P
Lecture Time
06:30 - 06:30
Speakers
  • Job Saris (Amsterdam, Netherlands)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Peritoneal metastases (PM) have an extremely poor survival irrespective of the primary cancer type. The reason for poor survival is the lack of therapies and the poor response to systemic treatment. We hypothesize that the peritoneal immune system (PerIS) underlies therapy resistance. The aim of this study was to characterize the PerIS in healthy individuals and patients with PM-colorectal cancer (PM-CRC), to identify novel treatment targets and combat peritoneal carcinomatosis.

Methods

Cellular indexing of transcriptomes and epitopes sequencing (CITE-seq) and single cell RNA-sequencing (scRNA-seq) was used to define the PerIS in peritoneal flushes (PF) from healthy individuals (n=5) and PM-CRC patients (n=13) undergoing elective surgery. The PerIS was compared to peripheral blood mononuclear cells (PBMCs), primary colon tissue and liver tissue. In PM-CRC mouse models (i.p. injection mouse CRC cell line CT26), macrophages were depleted using i.p. anti-CSF1R, with or without anti-PD1 treatment, or vehicle control. Primary outcome was survival and secondary outcomes were the modified peritoneal carcinomatosis index (mPCI) and ascites score.

Results

The healthy PerIS is characterized by immunosuppressive resident C1QA+VSIG4+ macrophages. The PerIS contains significantly more macrophages (mean: 28.0%) compared to colon (mean: 3.2%) and liver (mean: 1.6%). In PM-CRC, these macrophages become even more immunosuppressive with higher gene expression of IL10 and VEGFA and reduced expression of antigen presenting molecules. Moreover, protein levels of cytokines like IL10 (p=0.0006) and VEGFA (p=0.0003) are increased in PF of PM-CRC patients compared to healthy individuals and correlate to the PCI. In PM-CRC, C1Q+SPP1+ infiltrating macrophages were amongst the most abundant immune cells and contributed to immunosuppression. Intriguingly, mice studies demonstrated that anti-CSF1R/anti-PD1 combination therapy effectively reduced mPCI, ascites score and improved survival compared to vehicle control (p=0.0001), anti-CSF1R (p=0.005) and anti-PD1 (p=0.039).

Conclusions

Peritoneal resident macrophages define the peritoneal immunosuppressive niche and are a promising therapeutic target for PM-CRC.

Legal entity responsible for the study

Amsterdam UMC.

Funding

NWO VENI, KWF YIG, TKI grant and Amsterdam UMC PhD scholarship.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

174P - Targeting SPHK1 in macrophages suppresses liver metastasis of colorectal cancer and decouples anti-tumor immunity from immunotherapy toxicity

Presentation Number
174P
Lecture Time
06:30 - 06:30
Speakers
  • Yizhi Zhan (Guangzhou, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Colorectal cancer (CRC) is the second leading cause of cancer deaths, and liver metastasis accounts for most fatalities in CRC patients. Developing more effective treatments for patients with metastatic CRC is an urgent unmet need. Sphingosine kinase 1 (SPHK1) is selectively expressed in tumor-associated macrophages (TAMs) and SPHK1+ TAMs are abundant in metastatic tumor microenvironment. Here, we aimed to reveal the role of SPHK1+ TAMs in regulating immunosuppression and develop novel therapeutic strategies to enhance the efficacy of immune checkpoint blockade (ICB) therapy and mitigate ICB-induced toxicity.

Methods

The expression of SPHK1 in TAMs was determined using laser scanning microscopy and single-cell sequencing databases. The inhibitory effect of SPHK1 blockade alone or combined with ICB on liver metastasis was assessed using an orthotopic mouse model or intrasplenic injection of tumor cells. Flow cytometry and mass cytometry were used to analyze the immune microenvironment in SPHK1-/- knockout mice. The effect of SPHK1 inhibitor PF543 on the ICB-induced toxicity was verified by DSS-induced colitis or humanized mouse model in a combination of ICB. RNA sequencing and western blot were used to further explore the molecular mechanism by which SPHK1 promoted inflammasome activation and IL-1β release.

Results

SPHK1 was selectively expressed by TAMs in CRC, and the abundance of SPHK1+ TAMs was associated with adverse clinical outcomes in CRC patients. In vivo, SPHK1 knockout and inhibition suppressed liver metastasis of CRC and enhanced the anti-tumor activity of ICB. Blocking SPHK1 reduced the infiltration of TAMs and exhausted T cells, and promoted cytotoxicity of CD8+ T cells. Mechanistically, SPHK1/S1P axis led to IL-1β secretion in response to AIM2 and NLRP3 inflammasome activation. SPHK1 inhibitor PF543 and anti-IL-1R alleviated ICB-induced toxicity, including colitis and liver damage. PF543 plus anti-PD-1 therapy could induce complete regression of liver metastasis and mitigate liver dysfunction when further combined with radiotherapy.

Conclusions

Targeting SPHK1 in macrophages could inhibit liver metastasis of CRC and decouple ICB anti-tumor immunity and toxicity.

Legal entity responsible for the study

The authors.

Funding

National Natural Science Foundation of China (No.82103595).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

175P - MicroRNA-548c: An Immune-Activator microRNA at the Tumor Microenvironment and Immune Milieu of Breast Cancer

Presentation Number
175P
Lecture Time
06:30 - 06:30
Speakers
  • Alyaa Dawoud (New Cairo, Egypt)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Cancer cells evade the human immune system by manipulating the tumor-immune microenvironment (TIME) via suppressing the immuno-activator and promoting the immune-suppressive mediators. Hence, targeting immuno-modulatory factors became an ingenious treatment option, particularly for cancer types that shows scarcity of treatment options such as triple negative breast cancer (TNBC). Novel immuno-modulatory targets such as Galectin (GAL) 3 and 9 have been found to be overexpressed in TNBC, repressing the immune surveillance. Similarly, TNBC tumors were found to prevent the activation of immune infiltrating lymphocytes via overexpressing immune checkpoint CD155 expression on its surface. MHC class I polypeptide–related sequence A (MICA) and MICB are stress-induced ligands essential for innate immunity activation at the TIME. However, their expression is also often suppressed in TNBC, leading to immune suppression and poor prognosis. Our research group has extensively worked on microRNAs to modulate multiple targets simultaneously. Hence, this work unravels the expression profile of the miR-548c in BC tissues, evaluate its unprecedented immunomodulatory role in TNBC via altering GAL3/9, CD155, and MICA/B expression.

Methods

BC female patients (n=20) were recruited. In-silico analysis was used to identify miRNAs that target immunomodulatory targets. MDA-MB-231 TNBC cells were cultured and transfected by oligonucleotides. Total RNA was extracted using Biazol, reverse transcribed and quantified using qRT-PCR.

Results

miRNA-548c is a down-regulated tumor suppressor miRNA in BC tissues compared to its normal counterparts. MiRNA-548c was validated to regulate the immunogenic profile in TNBC. Given the overexpression of the immuno-suppressive GAL3, GAL9, and CD155, miR-548c simultaneously constrained their expression in TNBC cells. Moreover, ectopic expression of miR-548c in MDA-MB-231 cells resulted in a marked upregulation of the expression of the immuno-activators MICA and MICB, enhancing the overall immunogenic profile of TNBC cells.

Conclusions

This study identifies miRNA-548c as a booster of the immune surveillance by concomitant adjustment for the immunogenic ligands expressed/produced by the TNBC cell.

Legal entity responsible for the study

The authors.

Funding

This work was supported by Swiss National Science Foundation (SNSF), grant IZSTZ0_198887.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

176P - Multiplex-immunoflourescence spatial patterns to predict triple-negative breast cancer molecular subtypes in the IMMUcan study

Presentation Number
176P
Lecture Time
06:30 - 06:30
Speakers
  • Andrea Joaquin Garcia (Brussels, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

IMMUcan (SPECTA NCT02834884) is a European initiative to profile the tumor microenvironment (TME) for a better understanding of immune-tumor interactions. Here, we explored the association between distinct molecular phenotypes and spatial TME patterns in the prospective neoadjuvant IMMUcan TNBC cohort.

Methods

From a preliminary cohort of 132 patients, matched baseline RNA-seq and multiplex immune fluorescence (mIF) data were available for 66 cases. The mIF panel included CD8, PD1, PD-L1, granzyme B (GB), Ki67 and CK markers. Spatial TME patterns were defined by a graph-based approach detecting densely populated regions of tumor cells and their immune neighbors. TNBC molecular subtypes were derived from RNA-seq as described by Bareche et al. Area Under the Curve (AUC) was used to evaluate the accuracy of spatial patterns to predict TNBC subtypes.

Results

A total of eight distinct clusters were identified across the 66 samples, each exhibiting a specific spatial distribution of mIF markers. Two of the clusters showed high performance in predicting immunomodulatory phenotype (AUC: 0.72, 0.71, respectively). These clusters presented elevated densities of CD8+, CD8+/GB+, and CD8+/Ki67+ cells, consistent with CD8+ effector T cells. In addition, a cluster characterized by tumor cells correlated with the luminal androgen-receptor phenotype (AUC: 0.91). The basal-like phenotype was represented by a cluster exhibiting high levels of Ki67+ tumor cells (AUC: 0.61). A distinct cluster displaying an intermediate proportion of Ki67+ tumor cells was observed as well, representing the mesenchymal subtype (AUC: 0.69).

Conclusions

These preliminary analyses revealed the presence of informative spatial patterns populating mIF data, linked to the distribution of immune/tumor markers within the TME of TNBC. Of note, these spatial patterns were associated with distinct RNA-seq TNBC subtypes. These findings suggest the predictive power of mIF markers as a potential surrogate to discern TNBC heterogeneity. Consequently, these observations, if confirmed by further validations, could facilitate the implementation of treatment strategies tailored to the TNBC molecular subtypes.

Legal entity responsible for the study

EORTC.

Funding

IMI2 JU grant agreement 821558, supported by EU’s Horizon 2020 and EFPIA.

Disclosure

M. Morfouace: Financial Interests, Personal, Full or part-time Employment: Merck. H.S. Hong: Other, Personal, Full or part-time Employment: Merck. M. Cesaroni: Financial Interests, Personal, Full or part-time Employment: Sanofi. C. Sotiriou: Financial Interests, Institutional, Advisory Board: Astellas, Vertex, Seattle Genetics, Amgen, INC, Merck & Co; Financial Interests, Personal, Advisory Board: Cepheid, Puma; Financial Interests, Personal, Invited Speaker: Eisai, Prime oncology, Teva; Financial Interests, Institutional, Other, Travel: Roche; Financial Interests, Institutional, Other, Internal speaker: Genentech; Financial Interests, Personal, Other, Regional speaker: Pfizer; Financial Interests, Institutional, Invited Speaker: Exact Sciences. L. Buisseret: Financial Interests, Institutional, Advisory Board: Domain Therapeutics; Financial Interests, Institutional, Other, Steering Committee: iTEOS Therapeutics; Financial Interests, Personal, Other, writing of clinical cases: Mirrors of Medicine; Financial Interests, Institutional, Other, Travel grant: Gilead; Financial Interests, Institutional, Research Grant, Research Grant for an investigator initiated trial: Astra Zenaca; Non-Financial Interests, Personal, Principal Investigator: iTeos Therapeutics; Non-Financial Interests, Personal, Member: EORTC, BSMO. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

177P - The Immune-microenvironment Confers Chemoresistance in Breast cancer through activation of VEGFR2/STAT3/BIRC5 signaling

Presentation Number
177P
Lecture Time
06:30 - 06:30
Speakers
  • Bhawna Deswal (Noida, India)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Chemotherapy with Taxanes is often the preferred clinical treatment for breast cancer patients. Unfortunately, these agents become increasingly ineffective during treatment. Pro-tumoral M2 tumor-associated macrophages (TAMs) are the most abundant immune cells in the breast tumor microenvironment. In a first-in-human study (NCT00358163, NCT00731861), Vatalanib (PTK787/ZK) was well tolerated alone or in combination with paclitaxel. Here, we explored the pre-clinical efficacy of combination of Vatalanib and paclitaxel in TAM-induced chemoresistance in breast cancer.

Methods

We reported translational analysis of expression of BIRC5 and its correlation with Overall Survival (OS) in breast cancer patients. Western blotting, immunofluorescence imaging, and Real-time PCR analysis were used to determine survivin (BIRC5), pVEGFR2 and STAT3 in vitro. 4T1 cells and RAW 264.7 cells were injected in BALB/c mice to generate syngeneic chemoresistant breast tumor model in vivo.

Results

We measured survivin (BIRC5) upregulation in breast tumor (n=288) as compared to normal breast tissue (n=211) using TCGA datasets. Further, retrospective study confirmed that an increase in BIRC5 expression decreases Overall Survival (OS) in breast cancer. Using the invitro co-culture system, we validated that the macrophage secreted VEGF-A contributes to chemoresistance against paclitaxel. Mechanistically, VEGFA, by binding to VEGFR2, activated the transcription factor STAT3 leading to increased expression of anti-apoptotic protein survivin in breast cancer cells. Therapeutically, the TAM-induced STAT3/survivin signaling could be abrogated by Vatalanib thereby reversing the chemoresistance. The data shows that the combination of paclitaxel with Vatalanib exhibit reduced M2-TAMs, indicating that the combination effectively altered tumor microenvironment in the in vivo 4T1 breast tumor model.

Conclusions

Together, our study revealed the role of TAMs in conferring paclitaxel resistance in breast cancer. Combinatorial VEGFR2 blockade and paclitaxel chemotherapy decreased TAM population and showed better prognostic outcomes in breast cancer.

Legal entity responsible for the study

B. Deswal, S. Kapoor.

Funding

Department of Biotechnology, Government of India; Department of Science and Technology, Government of India.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

178P - Dynamics of breast cancer T cell repertoire during neoadjuvant chemotherapy / immunotherapy.

Presentation Number
178P
Lecture Time
06:30 - 06:30
Speakers
  • Charlotte Birchall (Woluwe-Saint-Lambert, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The immunogenicity of breast cancer (BC) determines the efficacy of treatment with immune checkpoint inhibitors (ICI), but it remains difficult to assess.

Methods

We analyzed the T cell repertoire (TCR) in 7 patients from B-IMMUNE trial exploring a neoadjuvant treatment with paclitaxel (week 1-12) and epirubicin / cyclophosphamide combined with durvalumab (week 13-24) on locally advanced luminal B HER2(-) or TN BC. Paired blood / tumor samples were collected before treatment, at week 12 and 24. The TCR repertoire was assessed by NGS targeted to TCRβ genes using an absolute quantitative bias-controlled approach. Paired tumor and blood TCR repertoire were compared in 5000 TCRβ sequenced molecules randomly selected. A tumor-enriched T-cell clonotype (TE-TC) was defined as a clonotype at least 50x more abundant in tumor than in blood (Fisher's test = p < 10-6). The spatial localization of TE-TCs in tumors was determined using a TCR-targeted custom adaptation of the 10x visium spatial transcriptomics method.

Results

TE-TC could be explained by an anti-tumor T-cell response and be a sign of tumor immunogenicity. They were tested before treatment in 7/7 patients and monitored at weeks 12 and 24 in 5/7. Overall, 101 TE-TCs were identified: 34 before treatment, 26 at week 12 and 41 at week 24 (respectively 2 to 9, 1 to 10 and 2 to 20 per patient). Of the 34 TE-TCs before treatment, 10 (29%) remained enriched in the tumor at week 12 and/or W24. Among the 41 TE-TCs detected at week 24, 31 (75%) were not detected before. The tumor spatial distribution of TE-TCs prior to treatment was assessed by spatial transcriptomic (50 μm resolution) in 4/7 patients. TE-TCs were detected in 3/4 patients in a minority of spatial spots (0.3 to 3%). For one patient, the spatial analysis was also assessed after 12 weeks of treatment. TE-TCs were detected in 10% of spatial spots with a predominant TE-TC that emerged during treatment and appears to be associated with Granzyme B and PD-1 expression in spatial clusters.

Conclusions

Before treatment, variable quantities of T cell clonotypes are highly enriched in locally advanced BC compared to blood, and new ones are enriched during neoadjuvant/ICI treatment. The influence of these clonotypes on tumor response needs to be studied.

Clinical trial identification

NCT03356860.

Legal entity responsible for the study

GHdC.

Funding

Seqalis.

Disclosure

J. Carrasco: Non-Financial Interests, Personal, Advisory Role: Seqalis. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

179P - Integrating multiplex immunofluorescence with gene expression data in the IMMUcan HER2-positive breast cancer cohort

Presentation Number
179P
Lecture Time
06:30 - 06:30
Speakers
  • Mattia Rediti (Brussels, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

IMMUcan (SPECTA NCT02834884) is an ongoing European effort to profile the tumor microenvironment (TME) in different cancer types. HER2-positive breast cancer (HER2+ BC) is heterogeneous, and TME characteristics impact treatment response. Here, we explored the correlation between multiplex immunofluorescence (mIF) and gene expression data in the IMMUcan prospective neoadjuvant early-stage HER2+ BC cohort.

Methods

A cohort of 149 patients was identified for preliminary analyses. mIF data were obtained from pretreatment tumor biopsies using 3 panels of marker antibodies [cytokeratin, CD15, CD163, CD11c, CD20, CD3, PD1, PD-L1, Ki67, granzyme B (GB), CD8, CD4, CD56, FOXP3] in 49 patients, evaluating stroma and tumor regions. Paired RNA sequencing (RNAseq) data were available for 44/49 patients. Spearman correlations (significant for P <0.05) were computed between mIF marker/cell phenotype densities (cells/mm2) and selected genes/gene expression signatures (GESs).

Results

Correlations between immune GESs and mIF markers/phenotypes ranged between 0.3 and 0.71. Of note, the gene CD274 and PD-L1 mIF stromal density presented high correlation (0.7), while the B cell stromal density correlated with a tertiary lymphoid structure GES (0.6). GESs describing proliferation were correlated with the densities of Ki67+ tumor cells (0.57 to 0.66), as well as with PD-L1 and several cell types (e.g., PD1+/Ki67+ tumor cells, CD8+/GB+/PD-L1+ cells, CD163-expressing macrophages; range 0.3 to 0.58). Expression levels of a PTEN loss GES was positively correlated with PD-L1 densities in both tumor and stroma (0.66 to 0.67), while GESs depicting glucose and lipid metabolism correlated (0.31 to 0.48) with CD163-expressing macrophages densities. Finally, negative correlations were observed between stroma-related signatures and densities of immune cells/markers (particularly CD8+ cells) as well as of proliferative tumor cells (-0.3 to -0.62).

Conclusions

In these preliminary analyses in the IMMUcan study, correlations between mIF and RNAseq data unveil biological processes potentially responsible for different immune infiltration patterns in HER2+ BC. Additional analyses are ongoing and further validation is warranted.

Clinical trial identification

NCT02834884; First posted: July 15, 2016.

Legal entity responsible for the study

EORTC.

Funding

IMI2 JU grant agreement 821558, supported by EU’s Horizon 2020 and EFPIA.

Disclosure

M. Morfouace: Financial Interests, Personal, Full or part-time Employment: Merck KGaA. L. Greillier: Financial Interests, Personal, Advisory Board: AbbVie, AstraZeneca, BMS, MSD, Novartis, Sanofi, Takeda, Roche; Financial Interests, Personal, Invited Speaker: Lilly, Pfizer; Financial Interests, Institutional, Local PI: AstraZeneca, AbbVie, BMS, MSD, Novartis, Takeda, Pfizer, PharmaMar; Financial Interests, Institutional, Coordinating PI: Sanofi; Financial Interests, Personal, Local PI: Roche. H.S. Hong: Financial Interests, Personal, Full or part-time Employment: Merck KGaA. M. Cesaroni: Financial Interests, Personal, Full or part-time Employment: Sanofi. C. Sotiriou: Financial Interests, Institutional, Advisory Board: Astellas, Vertex, Seattle Genetics, Amgen, INC, Merck & Co; Financial Interests, Personal, Advisory Board: Cepheid, Puma; Financial Interests, Personal, Invited Speaker: Eisai, Prime oncology, Teva; Financial Interests, Institutional, Other, Travel: Roche; Financial Interests, Institutional, Other, Internal speaker: Genentech; Financial Interests, Personal, Other, Regional speaker: Pfizer; Financial Interests, Institutional, Invited Speaker: Exact Sciences. L. Buisseret: Financial Interests, Institutional, Advisory Board: Domain Therapeutics; Financial Interests, Institutional, Other, Steering Committee: iTEOS Therapeutics; Financial Interests, Personal, Other, writing of clinical cases: Mirrors of Medicine; Financial Interests, Institutional, Other, Travel grant: Gilead; Financial Interests, Institutional, Research Grant, Research Grant for an investigator initiated trial: AstraZenaca; Non-Financial Interests, Personal, Principal Investigator: iTeos Therapeutics; Non-Financial Interests, Personal, Member: EORTC, BSMO. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

180P - Immunogenomic profiling of the randomized NeoPembrOv clinical trial reveals the VEGFR2 angiogenic axis as a promising actionable target to overcome immunoresistance of high-grade ovarian carcinomas

Presentation Number
180P
Lecture Time
06:30 - 06:30
Speakers
  • Olivia Le Saux (Lyon, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immune checkpoint inhibitors (ICI) have shown limited efficacy in high-grade serous carcinomas (HGSC). The NeoPembrOv randomized phase II trial investigated the combination of Pembrolizumab (P) with NeoAdjuvant ChemoTherapy (NACT), providing an opportunity to identify resistance mechanisms.

Methods

Patients (n=91) were randomized 2:1 to NACT+P or NACT. Paired samples were analyzed using RNA-seq (n=53) and multiplexed immunofluorescence staining (n=64) for CD8, PD-1, ActCasp3, panCK, CD31, and VEGFR2. Immune and endothelial signatures were assessed using MCP-counter and quanTIseq algorithms. Cell densities and nearest neighbors were determined based on InForm software output. We investigated the impact of NACT±P on the tumor microenvironment and identified parameters associated with survival. As no transcriptomic dataset of HGSC patients under ICI was available, we used an external validation cohort of 102 head and neck cancer patients.

Results

Analysis of DEG revealed a significant enrichment of T cell activation/differentiation pathways in the NACT+P arm only after treatment. We showed a significant increase in CD8+PD-1+ T cells density in tumor islets in the NACT+P arm compared to NACT (p=0.015). This increase observed in 15% of cases (n=6/41) was associated with improved overall survival (OS) (LR test, p=0.01).The median distance between apoptotic tumor and the nearest CD8+PD-1+ cell significantly decreased after NACT+P. Univariate cox models revealed a negative association between the MCP-counter endothelial cell signature and survival (HR=1.97, 95% CI [1.10-3.49], p=0.02) in the NACT+P arm, with a significant interaction with treatment arm (p=0.046). In situ analysis confirmed the negative predictive impact of high expression of VEGFR2 on CD31+ endothelial cells in patients receiving NACT+P, significantly affecting OS (p=0.005). KDR expression (VEGFR2) was validated as a predictive biomarker in an external cohort for OS (p=0.044).

Conclusions

Only a subset of HGSC patients can benefit from ICI. Targeting VEGFR2+ endothelial cells may overcome resistance. Clinical trials using VEGFR2 inhibitors are warranted.

Clinical trial identification

NCT03275506.

Legal entity responsible for the study

Arcagy-Gineco.

Funding

ARC Foundation.

Disclosure

O. Le Saux: Financial Interests, Personal, Advisory Board: Novartis, MSD, GSK; Financial Interests, Personal, Invited Speaker: Lilly, AstraZeneca, Clovis; Financial Interests, Institutional, Trial Chair: Novartis, Hospira-Pfizer foundation, Astellas. E. Coquan: Financial Interests, Personal, Advisory Board: Sanofi, MSD, BMS, Ipsen, AstraZeneca. I.L. Ray-Coquard: Financial Interests, Personal, Advisory Board: Roche, GSK, AstraZeneca, Mersana, Deciphera, Amgen, Oxnea, Merck Sereno, Agenus, Novartis, Macrogenics, Clovis, EQRX, Adaptimmune, Eisai, SUTRO, BMS, Adaptimmune, Daiichi Sankyo; Financial Interests, Institutional, Other, COLIBRI translational research: BMS; Financial Interests, Institutional, Advisory Board, translational research NEOPREMBROV trial: MSD; Non-Financial Interests, Personal, Principal Investigator: PAOLA1; Non-Financial Interests, Personal, Other, President: Gineco. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

181P - Tertiary lymphoid structures and B cells determine clinically relevant T cell phenotypes in ovarian cancer

Presentation Number
181P
Lecture Time
06:30 - 06:30
Speakers
  • Jitka Palich Fucikova (Prague, Czech Republic)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Intratumoral tertiary lymphoid structures (TLSs) have been associated with improved outcome in a variety of cancer patient cohorts, reflecting the key role of TLSs in the initiation of tumor-targeting immunity. Although, the clinical and biological relevance of TLSs, B cells, plasma cells and humoral adaptive immunity for patients with EOC have been previously reported, the precise immune contexture of developing TLSs, their impact the phenotypic profile of intratumoral T cells, and their influence on sensitivity to immunotherapy remain to be investigated in detail.

Methods

We harnessed spatial transcriptomics, multiplex immunofluorescence microscopy, and flow cytometry to characterize TLSs in patients with high-grade serous ovarian carcinoma (HGSOC), non-small cell lung carcinoma (NSCLC) and experimental mouse models.

Results

Here, we demonstrate that high-grade serous ovarian carcinoma (HGSOC) contains distinct immune aggregates with varying degrees of organization and maturation. Specifically, mature TLSs (mTLS) as forming only in 16% of HGSOCs with relatively elevated tumor mutational burden (TMB) were associated with an increased intratumoral density of CD8+ effector T (TEFF) cells and TIM3+PD1+, hence poorly immune checkpoint inhibitor (ICI)-sensitive, CD8+ T cells. Converesely, CD8+ T cells from immunologically hot tumors like non-small cell lung carcinoma (NSCLC) were enriched in ICI-responsive TCF1+ PD1+ T cells. Spatial B-cell profiling identified patterns of in situ maturation and differentiation that were associated with mTLSs. Moreover, B-cell depletion promoted signs of a dysfunctional CD8+ T cell compartment among tumor-infiltrating lymphocytes from freshly isolated HGSOC and NSCLC biopsies.

Conclusions

Taken together, our data demonstrate that – at odds with NSCLC – HGSOC is associated with a low density of follicular helper T cells and thus develops a limited number of mTLS that are insufficient to preserve a ICI-sensitive TCF1+PD1+ CD8+ T cell phenotype. These findings point to key quantitative and qualitative differences between mTLSs in ICI-responsive vs ICI-irresponsive neoplasms that may guide the development of alternative immunotherapies for patients with HGSOC.

Legal entity responsible for the study

The authors.

Funding

Sotio Biotech.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

182P - Exploring Cross-Compartmental Tumor Cell Plasticity and Immunogenicity in Serous Ovarian Cancer

Presentation Number
182P
Lecture Time
06:30 - 06:30
Speakers
  • Louisa S. Hell (Hamburg, Germany)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Ovarian cancer (OvCa) is one of the most prevalent cancers affecting women, with 5-year survival rates below 40%. Thus, it becomes imperative to deepen our comprehension of disease heterogeneity and progression. Within this context, the extraction of tumor cell aggregates from the solid tumor mass and the ascitic fluid offers a unique opportunity for a direct comparison of the two compartments. The identification of immune targets could provide guidance for immunotherapeutic strategies in the management of recurrent OvCa.

Methods

As part of debulking surgery, samples of solid tumor tissue and ascites were obtained from a cohort of 16 patients diagnosed with serous OvCa. Cell aggregates (ø 40-100μm) were isolated and cryo-preserved. Afterwards, specimens were subjected to a multi-parametric flow cytometric analysis, encompassing markers related to tumor (stem) characteristics and immunological features. Samples were scored by expression of plasticity markers (CD10, CD24, CD90, CD133) and immune markers (PD-L1, PD-L2, PVR, PVRL2, CD47, Calreticulin, CD39, CD73, HLA-A/B/C, HLA-DR/DP/DQ) were compared between groups of varying plasticities.

Results

Overall, ligands for co-inhibitory T-cell-receptors PD-L1 and PVR, as well as phagocytosis-associated molecules CD47 and Calreticulin were more highly expressed by tumor cells isolated from ascites, rather than solid tumors. Furthermore, EpCAM+ tumor cell subsets displaying ≥1 plasticity marker showed significantly higher levels of the investigated immunological targets. These findings were confirmed in a set of matched samples (n=4).

Conclusions

Our data indicates elevated levels of plasticity in cell aggregates derived from solid tumors. Immunogenicity markers, however, were more abundantly expressed by ascites-derived tumor cells. Given the frequent association of ascites with the dissemination and recurrence of OvCa, individuals with advanced-stage disease may benefit most from personalized immunotherapeutic interventions. Functional testing using patient-specific co-cultures of ascites-derived cell aggregates and immune cells could offer a path toward pre-therapeutic identification of patients, likely to respond to immunotherapeutic treatments.

Legal entity responsible for the study

The authors.

Funding

2cureX GmbH and BMBF funding.

Disclosure

L.S. Hell, T. Sturmheit, J. Kupper: Financial Interests, Personal, Full or part-time Employment: 2cureX GmbH. W.M. Fiedler: Non-Financial Interests, Personal, Other: AbbVie; Financial Interests, Personal, Research Grant: Amgen, Pfizer. B. Schmalfeldt: Financial Interests, Personal, Advisory Board: AstraZeneca, Roche, MSD, Eisai, GSK, Olympus; Financial Interests, Personal, Research Funding: Roche, MSD, GSK, AstraZeneca. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

183P - Multi-omics Investigation Reveals Cancer-Associated Fibroblast-Secreted FGF7 as an Ovarian Cancer Progression Promoter through HIF-1_/EMT Modulation

Presentation Number
183P
Lecture Time
06:30 - 06:30
Speakers
  • Songwei Feng (Nanjing, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Ovarian cancer (OC) is a highly aggressive malignancy with limited treatment options. Recent studies have focused on understanding the role of cancer-associated fibroblasts (CAFs) in OC progression.

Methods

Machine learning algorithms were utilized to analyze large-scale bulk transcriptomic datasets and identify FGF7 as a potential oncogenic factor. Expression levels of FGF7 were compared between CAFs, OC tissues, normal fibroblasts (NFs), and non-cancerous tissues. Various experimental techniques, including single-cell transcriptome analysis and in vitro experiments, were employed to investigate the interaction between FGF7 and OC cells, as well as the downstream signaling pathways involved.

Results

FGF7 expression was significantly elevated in CAFs and OC tissues compared to NFs and non-cancerous tissues, respectively. Higher FGF7 levels were associated with advanced tumor stage, vascular invasion, and poor prognosis. Experimental results demonstrated that CAFs-derived FGF7 enhanced OC cell proliferation, migration, and invasion. Mechanistic investigations revealed that FGF7 inhibited the degradation of hypoxia-inducible factor 1 alpha (HIF-1α) under normoxia, leading to the activation of EMT-related transcription factors and down-regulation of epithelial markers.

Conclusions

This study suggests that targeting the FGF7/HIF-1α/EMT axis may provide therapeutic opportunities for intervening in OC progression. Inhibition of FGF7 or HIF-1α signaling may be potential strategies to consider in future therapeutic interventions for OC.

Legal entity responsible for the study

The author.

Funding

National Natural Science Foundation of China.

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

184P - Elevated baseline circulating IL-8 is associated with increased expression of the IMmotion myeloid gene signature (GS) in metastatic clear cell renal cell carcinoma (mRCC) patients (pts) treated with nivolumab (nivo) within the NIVOREN GETUG-AFU 26 study.

Presentation Number
184P
Lecture Time
06:30 - 06:30
Speakers
  • LUCIA CARRIL AJURIA (Villejuif, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immunotherapy is standard of care in mRCC but tools for patients’ selection are lacking. The NIVOREN GETUG-AFU 26 study of nivolumab in mRCC included a translational program with assessment of circulating cytokines. We found IL-8 to be associated with overall survival (Carril-Ajuria et al. ASCO. 2022). Here, we aimed to evaluate the association between IL-8 and the mRCC immune landscape, including both circulating and tissue-based assessments.

Methods

Out of 720 pts treated with nivo within the NIVOREN study, 353 with available cytokine quantification were included for analysis. Immunohistochemistry expression of VEGF and CD163 at the invasive margin (IM) and at the tumor core (TC) was assessed. Previously published tissue-based GS were analyzed, including IMmotion myeloid, IMmotion Teff, and JAVELIN 101 immuno. Circulating inflammatory markers included neutrophils, NLR (neutrophil lymphocyte ratio), LIPI (Lung Immune Prognostic Index, based on derived NLR>3 and LDH>upper limit of normal). The association of baseline levels of IL-8 with baseline levels of circulating inflammatory markers, and with VEGF/CD163 expression and tissue-based GS expression were analyzed.

Results

Baseline characteristics were similar to the overall trial population. Higher levels of baseline circulating IL-8 (cut-off:17.9 pg/ml) were associated with increased baseline circulating neutrophiles (p=0.046), and NLR (p=0.008) as well as with LIPI (p=0.005). Among 353 pts, only 64 had available information on GS expression. Baseline IL-8 significantly correlated with increased IMmotion myeloid expression (p=0.041). No significant correlations between baseline circulating IL-8 and the other two GS, VEGF or CD163 expression were observed.

Conclusions

In our study, elevated baseline circulating IL-8 was not only significantly associated with higher levels of other circulating inflammatory markers, but also with increased expression of the myeloid inflammation gene signature at tumor level in mRCC pts.

Clinical trial identification

NCT03013335.

Legal entity responsible for the study

Unicancer.

Funding

Institut National du Cancer and Bristol Myers Squibb.

Disclosure

Y. Vano: Financial Interests, Personal, Invited Speaker: Ipsen, BMS, MSD, Pfizer, Merck; Financial Interests, Personal, Other, Congress and travel funding: Ipsen, MSD, Pfizer; Financial Interests, Institutional, Funding, Funding support for the CABIR study: Ipsen; Financial Interests, Institutional, Funding, Funding support for the BIONIKK study: Bristol Myers Squibb; Financial Interests, Personal, Steering Committee Member, Member of the scientific committee of the WITNESS study: Bristol Myers Squibb. B. Beuselinck: Financial Interests, Institutional, Advisory Board, Advisory board: BMS, MSD, AstraZeneca, Ipsen; Financial Interests, Institutional, Advisory Board, Testimony: BMS, MSD; Financial Interests, Institutional, Invited Speaker, Conference: Merck, Pfizer, Ipsen; Financial Interests, Institutional, Research Grant, Research grant for transational research on kidney cancer: BMS. L. Albiges: Financial Interests, Institutional, Other, Consulting: Astellas, BMS, Eisai, Ipsen, Janssen, MSD, Novartis, Pfizer, Roche, Merck; Financial Interests, Personal, Other, Honoraria: Novartis; Non-Financial Interests, Personal, Principal Investigator, Clinical trial steering committee: Pfizer, BMS, AVEO, AstraZeneca, MSD; Non-Financial Interests, Personal, Principal Investigator: Ipsen; Non-Financial Interests, Personal, Other, Clinical trial steering committee: Roche, Exelixis; Non-Financial Interests, Personal, Member: ASCO; Non-Financial Interests, Personal, Other, Medical Steering Committee: Kidney Cancer Association; Non-Financial Interests, Personal, Other, Member of the Renal Cell Carcinoma Guidelines Panel: European Association of Urology (EAU). All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

185P - The Immunosuppressive Landscape of Leukemia Inhibitory Factor (LIF) in Clear Cell Renal Cell Carcinoma

Presentation Number
185P
Lecture Time
06:30 - 06:30
Speakers
  • Yazan Al Zu’bi (Irbid, Jordan)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

LIF is an interleukin 6 class cytokine that plays a crucial role in the development of many solid tumors. Herein, we identify the elusive role of LIF in clear cell renal cell carcinoma (ccRCC) and its impact on tumor immune microenvironment.

Methods

The human cancer genome atlas (TCGA) was utilized to obtain ccRCC clinicopathological and expression data. The prognostic utility of LIF expression in ccRCC was evaluated using univariate and multivariate Cox logistic regression analysis. Co-expressed genes were explored using LinkedOmics and their biological roles were identified using gene set enrichment analysis (GSEA) through KEGG pathways terminology. The immune microenvironment was evaluated using Tumor IMmune Estimation Resource (TIMER 2.0). Correlations between LIF expression and immune-related genes were obtained using the Gene Expression Profiling Interactive Analysis (GEPIA).

Results

High LIF expression is associated with shortened overall survival (HR: 1.828, 95% CI: 1.358-2.460, P < .0001) in TCGA ccRCC cohort. Multivariate Cox logistic regression analysis identified LIF expression as an independent prognostic variable (HR: 0.48, 95% CI: 0.303-0.759, P = .002). GSEA showed enrichment in IL-17, TNF, JAK-STAT and NF-kB signaling pathways besides cytokine-cytokine receptor interaction, transcriptional misregulation in cancer, complement and coagulation cascades. LIF expression is positively corelated with immunosuppressive cellular elements including Tregs (ρ = 0.14, Q = 0.012), M2 macrophages (ρ = 0.25, Q < .0001), and myeloidderived suppressor cells (ρ = 0.18, Q value < .001). In addition, LIF corelated significantly with Tregs-related genes including FOXP3 (ρ = 0.28, P < .0001), CCR8 (ρ = 0.23, P < .0001), and TGF-β (ρ = 0.38, P < .0001). M2 macrophages-related genes were also significantly corelated with LIF expression as in CD163 (ρ = 0.30, P < .0001), VSIG4 (ρ = 0.27, P < .0001), and MS4A4A (ρ = 0.24, P < .0001). Immune check points genes were also influenced by LIF expression including CTLA4 (ρ = 0.25, P < .0001) and LAG3 (ρ = 0.21, P < .0001).

Conclusions

LIF upregulation corelates with poor prognosis in ccRCC and induces an immune suppressed microenvironment.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

186P - Post-anti-PD1 tumor characterization of HPV-negative R/M SCCHN: an EORTC IMMUcan sub-project

Presentation Number
186P
Lecture Time
06:30 - 06:30
Speakers
  • Athénaïs Van Der Elst (Brussels, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Characterization of the tumor molecular landscape and immune micro-environment (TIME) of recurrent and/or metastatic (R/M) squamous cell carcinoma of the head and neck (SCCHN) patients (pts) that progress after platinum therapy and PD-1 inhibitors will guide the rational development of new therapeutic approaches.

Methods

EORTC-1559 is a biomarker-driven study including pts with R/M SCCHN. Tumor biopsies are undergoing WES, RNA-seq, multiplex immunofluorescence (mIF) and Imaging Mass Cytometry (IMC) within the IMMUcan consortium.

Results

132 HPV-negative R/M SCCHN in disease progression were biopsied, including 112 who progressed on/after anti-PD1 treatment (anti-PD1-exposed) and 20 anti-PD1 naive pts. Pathogenic mutations in KEAP1 and focal amplifications in MDM2, LRP5 and deletions in APLNR were more frequent in the anti-PD1-exposed pts compared to anti-PD1 naive pts. RNA-seq analyses on 108 pts were mined to accommodate possible confounding factors based on regression models using feature selection. Compared to anti-PD1 naïve tumors, anti-PD1 exposed tumors were enriched in cytoskeleton, cell adhesion and DNA repair gene modules. Those genes were enriched in specific loci characterized by high GC content and specific transcriptional factor motifs like DNMT1. Among the anti-PD1 exposed pts, those treated with anti-PD1 as last treatment before tumor biopsy, were enriched in interferon response and immune activation gene sets, and had higher expression of LAG3. Finally, IMC was analyzed in 91 pts. Pts with anti-PD1 as last treatment had a trend for higher CD4, CD8, Treg and PDL1 + macrophages global densities. The local densities of 13 immune cell types within the radius of tumor cells were compared to their global density. Cluster analysis identified 3 groups: group 1 was infiltrated by neutrophils, group 2 by diverse immune cells excluding neutrophils, and group 3 was excluded for all immune cell types.

Conclusions

We report the global landscape of genetic, phenotypic and TIME changes associated with the selective pressure of PD1 inhibition of R/M SCCHN. The implications and validation in another prospective cohort of IMMUcan are underway.

Clinical trial identification

NCT03088059, EORTC-HNCG-1559.

Legal entity responsible for the study

EORTC.

Funding

IMI2 JU grant agreement 821558, supported by EU’s Horizon 2020 and EFPIA.

Disclosure

D. Herrero Saboya: Financial Interests, Personal, Full or part-time Employment: Servier Research & Develpment. M. Morfouace: Financial Interests, Personal, Full or part-time Employment: Merck Healthcare. R. Galot: Other, Personal, Other, Travel expenses: Merck. H.S. Hong: Financial Interests, Personal, Full or part-time Employment: Merck Healthcare. C. Lefebvre: Financial Interests, Personal, Full or part-time Employment: Servier Research & Development. C. Even: Financial Interests, Personal, Advisory Board: BMS, MSD, Innate Pharma, Merck Serono; Financial Interests, Institutional, Advisory Board: F Star Therapeutics, Novartis, Elevar; Financial Interests, Institutional, Local PI: BMS, AstraZeneca, ISA pharmaceutics, MSD, Debiopharma, Ayala, Gilead; Financial Interests, Institutional, Coordinating PI: BMS, Novartis, Sanofi. C. Le Tourneau: Financial Interests, Personal, Advisory Board: BMS, MSD, Merck Serono, Nanobiotix, Roche, Rakuten, Seattle Genetics, GSK, Celgene, ALX Oncology, Exscientia. A. Daste: Financial Interests, Personal, Advisory Board: Merck, BMS, MSD. P. Saintigny: Financial Interests, Personal, Funding: HTG Molecular Diagnostics, Inivata, ArcherDx, Bristol Myer Squibb, Roche Molecular Diagnostics; Financial Interests, Personal, Research Funding: Roche; Financial Interests, Personal, Advisory Board: AstraZeneca, Novartis, BMSFoundation, Illumina; Financial Interests, Institutional, Advisory Board: Omicure; Financial Interests, Personal, Other, Travel, Accommodations, Expenses: Illumina, Bristol Myer Squibb, AstraZeneca, Roche. J. Machiels: Financial Interests, Institutional, Advisory Board: Novartis, MSD, Pfizer, Roche, Debio, AstraZeneca, Innate, Nanobiotix, Bayer, Merck Serono, Boehringer Ingelheim, BMS, Pfizer, Cue Pharma, Incyte, Janssen, Johnson & Johnson, ALX Oncology, F-star, Nektar, F-star, Seagen, Astellas, Genmab; Financial Interests, Institutional, Other, Travel expense: Gilead, MSD, Sanofi; Financial Interests, Institutional, Steering Committee Member: AstraZeneca, MSD; Financial Interests, Institutional, Coordinating PI: MSD, iTeos, eTheRNA; Financial Interests, Institutional, Local PI: Pfizer, Ceylad, MSD, Novartis, KURA, Roche, Lilly, Boehringer, Sanofi-Aventis, Incyte, Bayer, Merck - Serono, Janssen, Johnson & Johnson, Amgen, AbbVie, GSK; Non-Financial Interests, Personal, Leadership Role, Chair: EORTC head and neck group. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

187P - Local and systemic anti-tumor response during tumor development in an immune privileged site: the case of uveal melanoma

Presentation Number
187P
Lecture Time
06:30 - 06:30
Speakers
  • Francesca Lucibello (Paris, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Uveal melanoma (UM), an uncommon but aggressive eye cancer, exhibits genetic diversity, classifying into M3 (poor prognosis) and D3 (better outcome) subsets. Despite low mutation rates, metastatic UM patients have detectable CD4 and CD8 T cells, suggesting an immune response. We recently identified public neo-antigens generated by SF3B1 mutations in D3 tumors, triggering CD8+ T cell responses. In M3 tumors with BAP1 inactivation, immune infiltration increases, but CD8 T cell specificity remains unclear.

Methods

We collected blood and tumor samples from uveal melanoma patients at Institut Curie. Specific peptide-loaded HLA-A2 tetramers were labeled and used for staining blood and tumor cells. Single-cell RNA-Seq experiments were performed on TILs.

Results

In our analysis of 24 enucleated patient samples, we observed an enrichment of CD8+ T cells over CD4+ T cells within the tumor microenvironment. A notable fraction of CD8+ T cells exhibited exhaustion markers like PD-1 and CD39, suggesting the presence of an Ag-specific immune response. Among these exhausted CD8+ T cells, 1-10% were specific for HLA-A2: Melan-A. By using scRNA-seq and VDJ TCR-seq we explored T cells response within 3 primary HLA-A2+ M3 tumors. Our analysis identified nine distinct clusters, notably Cluster 0, which displayed upregulation of genes such as PDCD1 (PD-1), ENTPD1 (CD39), HAVCR2 (TIM3), LAG3, GZMB, and GZMK. This gene expression profile suggests chronic activation due to exposure to tumor antigens. Melan-A tetramer-positive cells predominantly localized to Cluster C0, and the largest clonotype was associated with this cluster. We also assessed systemic recirculation by sequencing TCRs in PBMCs, revealing varying recirculation levels among different T-cell clusters. While chronically activated (C0) T cells displayed limited recirculation, T cells with a memory phenotype (C2), cytotoxic functions (C4), and effector T-cell responses (C6) were more present in the peripheral blood.

Conclusions

These findings challenge the notion of the eye as an immune-privileged site, as we consistently observed immune responses within UM tumors. The mechanism by which tumor antigens prime naïve T cells in this site remains mysterious.

Legal entity responsible for the study

F. Lucibello.

Funding

Ligue Contre le Cancer.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

188P - Expression of the co-stimulatory checkpoint protein OX40L (TNFSF4) in the melanoma micro-environment

Presentation Number
188P
Lecture Time
06:30 - 06:30
Speakers
  • Raya Leibowitz-Amit (Be'er-Yaakov, Israel)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immune checkpoint inhibitors revolutionized treatment of both locally advanced and metastatic melanoma patients, yet some patients fail to achieve long-lasting benefit or progress on treatment, highlighting the need for new immune-modulatory agents. The co-stimulatory ligand OX40L and its receptor OX40 (also named TNFSF4/ TNFRSF4) were previously shown to elicit anti-tumor immune responses in pre-clinical cancer models. This led to several clinical trials of OX40-agonism as therapeutic interventions in cancer, which showed limited efficacy thus far. We previously analyzed RNA-sequencing data of OX40L expression in melanoma and found that low mRNA expression was associated with worse prognosis and with worse outcome following anti-PD1 treatment. These findings encouraged further studies to substantiate the role of OX40L in the melanoma microenvironment.

Methods

Multiplex immunofluorescent microscopy was used to evaluate the expression of OX40L/OX40 in FFPE surgical specimens of primary/metastatic melanoma tumors. Previously established cell-specific markers were used in parallel to identify OX40L- expressing cell phenotypes. Quantification was performed with GEN5 PRIME software.

Results

OX40L+ cells were detected in 15/16 tumors tested, in variable abundances, which correlated with the abundance of OX40+ cells and with co-localization events of OX40L+/OX40+ cells, suggesting functional OX40 ligand-receptor interactions. OX40L expression was frequently detected on Macrophages/Dentritic-cells, less on CD4+ or CD8+ T cells and rarely on melanoma cells. Unexpectedly, we now identified high prevalence of OX40L expression on Foxp3+ regulatory T cells (Treg), in addition to OX40, which was previously shown to antagonize their suppressive function.

Conclusions

Here we show that OX40L is abundantly expressed, together with OX40, on Treg within the melanoma micro-environment. These novel findings may suggest a role for OX40L in modulating Treg suppressive activity. If further proven, our observations may be important in prognostication and prediction of response in melanoma. Furthermore, they may point to new IO combinations for further research and have therapeutic implications towards ‘heating’ up tumors.

Legal entity responsible for the study

The authors.

Funding

Israeli Cancer Association (ICA).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

189P - The impact of immune microenvironment subopopulations on soft tissue sarcomas

Presentation Number
189P
Lecture Time
06:30 - 06:30
Speakers
  • Shokhrukhbek Khujaev (Tashkent, Uzbekistan)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Malignant tumors are sophisticated, organized ecosystems in addition to collections of cancer cells. It has long been established that the formation, recurrence, and metastasis of cancers are all tightly correlated with their immune components. However, the impact of the tumor microenvironment in soft tissue sarcoma (STS) remains mainly unclear. Studying how the immunological microenvironment affects the clinical presentation and prognosis of soft tissue sarcomas is the objective.

Methods

Retrospective research was conducted on 168 soft tissue sarcoma patients who received care at the Republican specialized Scientific and practical Medical Center of Oncology and Radiology of Uzbekistan. Through immunohistochemistry analyses of CD3, CD4, CD8, CD20, and CD68, lymphocyte subpopulations that infiltrate tumours and their subpopulations were investigated.

Results

In the immunological microenvironment, CD 68+ macrophages and CD 3+ T cells were the most prevalent cell types. There was a substantial positive correlation between CD 68 expression and the probability of local recurrence (p = 0.014) in a multivariate analysis employing the Fine & Gray risk regression model with death as a competing event, independent of age, resection margins, and the presence of B cells. Furthermore, B cell abundance was significantly lower (p = 0.013) and macrophage abundance was much higher in patients older than middle age. Regarding histological subtypes, undifferentiated pleomorphic sarcoma and myxofibrosarcoma were more frequently found to have these cells, which had a high total number of tumor-infiltrating lymphocytes in general and macrophages in particular.

Conclusions

The significant number of CD 68 macrophages in soft tissue sarcomas and their detrimental effect on the prognosis for local recurrence are both confirmed by this investigation.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

190P - Immune-related roles of B7H3 in glioblastoma

Presentation Number
190P
Lecture Time
06:30 - 06:30
Speakers
  • Arnaud Simonet (Liège, Belgium)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Glioblastoma is the most frequent and aggressive primitive brain cancer, associated with dreadful prognosis. This project focuses on a protein called B7H3, member of the B7 « immune checkpoint » protein family. Two isoforms of B7H3 protein (i.e. 2Ig and 4Ig) are produced by alternative splicing and expressed at the cell membrane. Of note, the ectodomain of B7H3 exists as a soluble protein. B7H3 expression in several cancers is associated with poorer prognosis and increased aggressiveness, however its precise role in immune cell modulation in the context of glioblastoma remains elusive.

Methods

Using a murine glioblastoma cell line (CT2A), we allografted immunocompetent mice with CT2A glioma cells overexpressing (OE) the 2Ig isoform of B7H3 (vs cells with control vector). We also engineered a human cell line (U87 OE 2Ig B7H3 vs control vector) to decipher the role of the B7H3 protein on immune cell modulation. Finally patient-derived glioblastoma stem-like cells (GSCs) were used for additional in vitro experiments.

Results

We observed a slight increase in survival in CT2A OE-B7H3 mice, which surprisingly indicated a tumor suppressive effect. We are currently reproducing the experiment using another murine cell line (GL261). Conversely, in vitro coculture experiments revealed no impact of B7H3 on T cell activation or apoptosis, but preliminary results suggest that B7H3 lowers T cell proliferation in vitro. We showed that patient-derived GSCs release soluble B7H3 in the extracellular space and is especially found on extracellular vesicles (EVs).

Conclusions

These results suggest that B7H3 may exert diverse functions in different aspects of glioblastoma immunogenicity. We are exploring the roles of B7H3 isoforms as well as the role it could play on glioblastoma cell-derived EVs, to understand its impact on the immune system.

Legal entity responsible for the study

The authors.

Funding

FNRS, Télévie, Fondation Léon Frédéricq.

Disclosure

G. Jerusalem: Financial Interests, Personal, Advisory Board: Novartis, Roche, Pfizer, BMS, Lilly, AstraZeneca, Daiichi Sankyo; Financial Interests, Personal, Invited Speaker: Novartis, Pfizer, BMS, Lilly, AstraZeneca, Seagen; Financial Interests, Personal, Steering Committee Member: Novartis, AZ/Daiichi Sankyo, BMS; Financial Interests, Institutional, Local PI: Novartis, Roche, Syneos Health, Iqvia, TRIO, Bayer, MSD, IBCSG, PRA, Eli Lilly, PPD, Theradex, ABCSG, Modra, AstraZeneca, Odonate Therapeutics, BMS, ICON, Boeringer, Quintiles, Pfizer, Lilly, Daiichi Sankyo, Seagen, MedImmune; Non-Financial Interests, Personal, Member: ASCO, BSMO; Other, Personal, Other, Medical writer support: MedImmune, Novartis, Roche, Lilly, Amgen, BMS, AstraZeneca, Merck, Pfizer; Other, Personal, Other, Support for attending meetings and/or travel: Novartis, Lilly, BMS, Roche, Amgen, AstraZeneca, Pfizer. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

191P - Senolytic treatment remodels glioblastoma microenvironment

Presentation Number
191P
Lecture Time
06:30 - 06:30
Speakers
  • Alexa Saliou (Paris, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Glioblastoma (GBM), isocitrate dehydrogenase wild-type (IDH-WT), is the most aggressive and invasive form of glioma in adults. This primary brain tumor remains refractory to standard protocols, resulting in a very limited patient survival. GBM is a cold tumor, characterized by a low infiltrative lymphocyte pool and the enhanced expression of immunosuppressive molecules which may explain the failure of immunotherapy. We recently showed that partial removal of malignant senescent cells improved the survival of GBM-bearing mice and remodels the tumor microenvironment (TME) towards a less anti-inflammatory phenotype. Further, we observed an upregulation of genes associated with interferon alpha and gamma, pathways which play key roles in immunity activation and anti-tumor response1.

Methods

To better understand how senescent cells shape the TME and to test whether senolytic drug therapy may be a beneficial adjuvant therapy for patients with GBM, we applied a genetic senolytic (p16-3MR + GCV) to an immunocompetent mesenchymal GBM mouse model in combination or not with immune checkpoint inhibitors (ICI). We analyzed the TME using transcriptomic approaches (bulk RNA sequencing) and the overall survival of mice bearing GBM after treatments.

Results

First, we observed that malignant cells were enriched in genes associated with the antigen-presenting machinery in GBM depleted for senescent cells (p16-3MR+GCV, n=9) compared with controls (WT+GCV, n=5). Second, we found that a combination of senolytic and a cocktail of ICI significantly increases the overall survival of mice bearing GBM compared with mice treated with senolytic and control isotypes.

Conclusions

All together our findings suggest a remodeling of the GBM microenvironment upon senescent cells’ removal, facilitating response to ICI. Further work is needed to understand underlying mechanisms of ICI response in a GBM-context and identify the immune cell types responsible for this survival benefit. 1 Salam, Saliou et al. Cellular senescence in malignant cells promotes tumor progression in mouse and patient Glioblastoma. Nat. Commun. 14, 1–21 (2023).

Legal entity responsible for the study

The authors.

Funding

Ligue contre le Cancer, Fondation ARC, Siric Curamus.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

192P - Analysis of Tumor-Associated Macrophages and Tumor-infiltrating Lymphocytes within the Tumor Microenvironment of Primary Tumors and Matched Brain Metastases

Presentation Number
192P
Lecture Time
06:30 - 06:30
Speakers
  • Markus Kleinberger (Vienna, Austria)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Response rates of immune checkpoint inhibitors (ICI) differ between primary tumors and brain metastases (BM). Here, we aimed to analyze infiltration patterns of inflammatory cells within the tumor microenvironment (TME) of primary tumors and matched BM tissue samples.

Methods

We retrospectively identified patients who received resection of primary tumor and BM between 01/1990 and 10/2022. Immunohistochemical FFPE tissue staining, density quantification of tumor-associated macrophages (TAMs; CD68+, CD163+) and tumor-infiltrating lymphocytes (TILs; CD3+, CD8+, CD45Ro+, FoxP3+) was performed. Images were processed with QuPath software, heterogeneity of generated heatmaps was measured by Shannon entropy.

Results

74 patients with matched samples were analyzed: 58/74 (78%) non-small cell lung cancer (NSCLC) , 8/74 (11%) breast cancer (BC), 8/74 (11%) renal cell carcinomas (RCC) (61.3% males, 38.7% females; median age 57 years). Median OS after primary diagnosis was 176.4 weeks (range 9.4-829.1). Median time to BM was 58.2 weeks (range -24.9 -520.6). CD163+ density was significantly higher in BM, whereas CD8+ was significantly lower in BM over all entities (p<.05) compared to primary tumors. CD3+ density was lower in BM of NSCLC and BC (p<.001, p=.008, respectively). CD68+ showed higher density in BC BM (p=.016) and FoxP3+ was higher in NSCLC primary tumors (p<.001; all Wilcoxon rank sum test). Cell density and entropy of FoxP3+ correlated in matched samples (p=.002, ρ=.37 and p<.001, ρ=.65), while CD3+ (p=.002, ρ=.37) and CD8+ (p<.001, ρ=.47) did only in BM. In primary NSCLC, FoxP3+ and CD163+ showed lower entropy indices, therefore more homogeneous distribution compared to matched BM (p=.002, p=.004, respectively). Multivariable Cox regression model demonstrated significant associations of CD3+ density (p=.006) and CD45Ro+ entropy (p=.001) in primary tumor with time to BM diagnosis.

Conclusions

Cell densities of CD163+-TAMs within the TME of BM are significantly higher compared to primary tumors, whereas CD8+ and CD3+ TILs are less present in BM. Differences in the inflammatory cell infiltration pattern could explain different ICI responses between primary tumor and BM.

Legal entity responsible for the study

The authors.

Funding

Austrian Federal Ministry for Digital and Economic Affairs, National Foundation for Research, Technology and Development, Christian Doppler Research Association.

Disclosure

M. Preusser: Financial Interests, Personal, Advisory Board: Bayer, Bristol Myers Squibb, Novartis, Gerson Lehrman, CMC Contrast, GSK, Mundipharma, Roche, BMJ Journals, MedMedia, AstraZeneca, AbbVie, Lilly, Medahead, Daiichi Sankyo, Sanofi, Gan & Lee Pharmaceuticals, Merck Sharp & Dohme, Tocagen, Adastra; Financial Interests, Institutional, Research Grant, Clinical Trials and research: Boehringer Ingelheim, Bristol Meyers Squibb, Roche, Daiichi Sankyo, Merck Sharp & Dohme, Novocure, GSK, AbbVie; Non-Financial Interests, Personal, Leadership Role, Brain Tumor Group Chair (current): EORTC; Financial Interests, Institutional, Coordinating PI: PharmaMar; Non-Financial Interests, Personal, Leadership Role, EANO Past-President (current): EANO; Non-Financial Interests, Personal, Other, Member Multi-Site Guideline Advisory Group: ASCO. J.N. Kather: Financial Interests, Personal, Invited Speaker, Talk in 2023: Fresenius, BMS; Financial Interests, Personal, Invited Speaker, Talk in 2022: Eisai, MSD; Financial Interests, Personal, Advisory Board, Scientific Advisory Board since 2022: Owkin, Panakeia, London, UK; Financial Interests, Personal, Advisory Board, Scientific Advisory Board since 2023: DoMore Diagnostics; Financial Interests, Personal, Invited Speaker, Talk in June 2023: Bayer; Financial Interests, Personal, Invited Speaker, Talk in Sept 2023: Roche Diagnostics International Ltd; Financial Interests, Personal, Advisory Board, consultation: Repare Therapeutics USA; Financial Interests, Personal, Invited Speaker, Talk in Oct 2023: Pfizer; Financial Interests, Personal, Stocks/Shares, Shares and part-time activities in a company that provides artificial intelligence services.: StratifAI GmbH. A.S. Berghoff: Other, Personal, Other: Daiichi Sankyo, Roche, Bristol Meyers Squibb, Merck, Daiichi Sankyo, Amgen, AbbVie. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

193P - Engagement of sialylated glycans with Siglec receptors on suppressive myeloid cells inhibit anti-cancer immunity via CCL2

Presentation Number
193P
Lecture Time
06:30 - 06:30
Speakers
  • Ronja Wieboldt (Basel, Switzerland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Overexpression of sialic acids on glycans, called hypersialylation is a common alteration found in cancer. Hypersialylation can, for example, enhance immune evasion via interaction with sialic acid-binding immunoglobulin-like lectin (Siglec) receptors on tumor-infiltrating immune cells. Here, we tested the role of sialic acid on myeloid-derived suppressor cells (MDSCs) and their interaction with Siglec receptors.

Methods

Siglec expression of murine and human MDSCs in healthy conditions and tumor setting was assessed by flow cytometry and MALDI-MS analyses. Functional analysis of Siglec-E knockout on MDSCs in mice was evaluated using Siglec-ExLysMCre mice and suppressive capacity was tested in vitro. Results were confirmed in the human setting using an in vitro assay to generate MDSC-like cells including RNA-Sequencing and a MDSC suppression assay with cancer-derived MDSCs.

Results

We found that MDSCs derived from the blood of lung cancer patients and tumor-bearing mice strongly express inhibitory Siglec receptors. In murine cancer models of emergency myelopoiesis, Siglec-E knockout on myeloid cells resulted in prolonged survival and increased infiltration of activated T cells. Targeting suppressive myeloid cells by blocking Siglec receptors or desialylation led to strong reduction of their suppressive potential. We further identified CCL2 as mediator involved in T cell suppression upon interaction of sialoglycans and Siglec receptors on MDSCs.

Conclusions

Our results provide mechanistic insights how sialylated glycans inhibit anti-cancer immunity by facilitating CCL2 expression.

Legal entity responsible for the study

The authors.

Funding

The authors.

Disclosure

H. Läubli: Financial Interests, Institutional, Advisory Board: Bristol Myers Squibb; Financial Interests, Personal, Advisory Board: Palleon Pharmaceuticals, GlycoEra; Financial Interests, Institutional, Invited Speaker: Novartis. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

194P - Achieving Reproducible Maturation Staging of Tertiary Lymphoid Structures: from Imaging Mass Cytometry Data to Pathology Applications

Presentation Number
194P
Lecture Time
06:30 - 06:30
Speakers
  • Marion Le Rochais (Brest, France)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

In cases of persistent inflammation, the migration of immune cells follow the organogenesis of secondary lymphoid organs (SLO) within the organs, leading to the formation of tertiary lymphoid structures (TLS). These structures have become important as diagnostic and prognostic markers in chronic diseases and cancers. However, there is ongoing debate regarding how to assess the maturation of TLS in tissue samples, both in terms of determining the stages themselves and achieving agreement among pathologists. Based on the composition and organization of TLS revealed through imaging mass cytometry (IMC), we aimed to propose a reproducible classification system consisting of three maturation stages for TLS, which can be applied in pathology practice.

Methods

We used formalin-fixed and paraffin-embedded gastric samples (45 tissues with 134 TLS) and colonic samples (50 tissues with 159 TLS), representing inflammatory, cancerous, and control conditions. These samples were subjected to IMC analysis using a panel of 39 markers. Additionally, we sought correlations between markers that reflect the maturation of TLS (composition and architecture). The two IMC-markers with the strongest correlation with TLS maturation were selected to transfer the TLS maturation staging from IMC to dual-staining immunohistochemistry (IHC). This transfer was done to investigate the level of agreement among pathologists when classifying TLS using a set of 60 tissue IHC images.

Results

Using IMC, we were able to characterize the maturation of TLS and propose three distinct stages: Early TLS, Primary Follicle-Like TLS, and Secondary Follicle-Like TLS, identified as IHC CD21-CD23-, CD21+CD23-, and CD21+CD23+, respectively. The analysis of the 60 dual CD21-CD23 IHC images by three pathologists showed a high level of inter-pathologist agreement in classifying TLS into the three maturation stages (Cohen’s Kappa values: > 0.8).

Conclusions

With a strong correlation to cellular and architectural changes during TLS maturation, the dual CD21-CD23 IHC marker allows for the staging of TLS in tissue samples with a high level of inter-pathologist agreement, which holds potential for prognostic and predictive purposes.

Legal entity responsible for the study

M. Le Rochais.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

195P - IMMUcan - Toward a better understanding of the tumor microenvironment to inform precision oncology approaches.

Presentation Number
195P
Lecture Time
06:30 - 06:30
Speakers
  • Marie Morfouace (Darmstadt, Germany)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immune checkpoint inhibitors (ICI) have rapidly changed the treatment landscape in oncology. However, the majority of cancer patients do not benefit from immunotherapies (primary resistance) or present with only short-term benefit (secondary resistance).

Methods

IMMUcan is a public-private European consortium funded by the Innovative Medicines Initiative, set-up in 2019, to elucidate the relationship between host and cancer cells within the tumor microenvironment (TME) and the impact of current standard of care treatments in up to 3,000 cancer patients. By integrating molecular (whole exome sequencing and total RNA sequencing) and cellular data (multiplex immunofluorescence (IF) and imaging mass cytometry (IMC)) of patient tumors with longitudinal clinical information, the tumor-host interaction and the response to therapies can be dissected. This represents a unique multi-modal dataset facilitating the discovery of novel biomarkers and resistance mechanisms. Thus, IMMUcan arguably represents one of Europe's largest TME-profiling efforts to date.

Results

More than 100 investigators from 17 European countries are prospectively recruiting patients with non-small cell lung cancer (NSCLC), head and neck squamous cell carcinoma (HNSCC), triple negative breast cancer (TNBC), renal cell carcinoma and colorectal cancer, using the EORTC SPECTA platform (NCT02834884). To date, more than half of the targeted patients have been recruited, including almost 400 HNSCC and NSCLC as well as more than 600 breast cancer patients.

Conclusions

The first analysis on these cohorts will be presented during the ESMO-IO conference highlighting the novel biomarkers identified in TNBC (early and metastatic settings), the resistance mechanisms of HNSCC patients treated with immunotherapies, as well as the integration of molecular and cellular modalities to gain an understanding of advanced NSCLC disease.

Clinical trial identification

SPECTA - NCT02834884.

Legal entity responsible for the study

European Organisation for Research and Treatment of Cancer.

Funding

Funding: IMI2 JU grant agreement 821558, supported by EU’s Horizon 2020 and EFPIA.

Disclosure

M. Morfouace: Financial Interests, Personal, Member, Employee: Merck Healthcare. L. Buisseret: Financial Interests, Institutional, Advisory Board: Domain Therapeutics; Financial Interests, Institutional, Other, Steering Committee: iTEOS Therapeutics; Financial Interests, Personal, Other, writing of clinical cases: Mirrors of Medicine; Financial Interests, Institutional, Other, Travel grant: Gilead; Financial Interests, Institutional, Research Grant, Research Grant for an investigator initiated trial: Astra Zenaca; Non-Financial Interests, Personal, Principal Investigator: iTeos Therapeutics; Non-Financial Interests, Personal, Member: EORTC, BSMO. B. Besse: Financial Interests, Institutional, Funding: 4D Pharma, AbbVie, Amgen, Aptitude Health, AstraZeneca, BeiGene, Blueprint Medicines, Boehringer Ingelheim, Celgene, Cergentis, Cristal Therapeutics, Daiichi Sankyo, Eli Lilly, GSK, Pfizer, Janssen, Onxeo, OSE Immunotherapeutics, Roche-Genentech, Sanofi, Takeda, Tolero Pharmaceuticals; Financial Interests, Institutional, Research Grant: Genzyme Corporation, Chugai pharmaceutical, Eisai, Inivata, Ipsen, Turning Point Therapeutics. J. Machiels: Financial Interests, Institutional, Advisory Board: Novartis, MSD, Pfizer, Roche, Debio, AstraZeneca, Innate, Nanobiotix, Bayer, Merck Serono, Boehringer Ingelheim, BMS, Pfizer, Cue Pharma, Incyte, Janssen, Johnson & Johnson, ALX Oncology, F-star, Nektar, F-star, Seagen, Astellas, Genmab; Financial Interests, Institutional, Other, Travel expense: Gilead, MSD, Sanofi; Financial Interests, Institutional, Steering Committee Member: AstraZeneca, MSD; Financial Interests, Institutional, Coordinating PI: MSD, iTeos, eTheRNA; Financial Interests, Institutional, Local PI: Pfizer, Ceylad, MSD, Novartis, KURA, Roche, Lilly, Boehringer, Sanofi-Aventis, Incyte, Bayer, Merck Serono, Janssen, Johnson & Johnson, Amgen, AbbVie, GSK; Non-Financial Interests, Personal, Leadership Role, Chair: EORTC head and neck group. C. Sotiriou: Financial Interests, Institutional, Advisory Board: Astellas, Vertex, Seattle Genetics, Amgen, INC, Merck & Co; Financial Interests, Personal, Advisory Board: Cepheid, Puma; Financial Interests, Personal, Invited Speaker: Eisai, Prime oncology, Teva; Financial Interests, Institutional, Other, Travel: Roche; Financial Interests, Institutional, Other, Internal speaker: Genentech; Financial Interests, Personal, Other, Regional speaker: Pfizer; Financial Interests, Institutional, Invited Speaker: Exact Sciences. D. Schaer: Financial Interests, Personal, Member, Employee: Bayer; Financial Interests, Personal, Stocks/Shares: Eli Lilly. H.S. Hong: Financial Interests, Personal, Member, Employee: Merck Healthcare. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

196P - Local glycan engineering induces systemic antitumor immune reactions via antigen cross-presentation

Presentation Number
196P
Lecture Time
06:30 - 06:30
Speakers
  • Natalia Rodrigues Mantuano (Basel, Switzerland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

New therapies that aim to remodel the tumor glycosylation and reactivate the immune system are being explored. In particular, approaches that target the sialoglycan-Siglec as glyco-immune checkpoint.

Methods

An adeno-associated virus was constructed to express influenza A sialidase (AAVsia). Efficacy of sialidase production in transduced cells and activity were tested in vitro and mouse tumor models after intratumoral injection. Combination therapy with checkpoint inhibitors (PD-1 and CTLA-4) and FUCA-1 (human fucosidase) were performed. In addition, efficacy of immune reactivation against tumors and cancer control was tested in syngeneic mouse models. Immune reactions were further characterized by flow cytometry and single cell RNA sequencing. To access the impact of desialylation in cross-presentation, bone marrow derived dendritic cells were pulsed with cancer cells and co-cultured with OT-I cells with sialidase. Proliferation and activation were evaluated by flow cytometry.

Results

Upon AAV-sia treatment, cancer cells express sialidase on the cell surface and are able to cleave sialic acid in the tumor microenvironment in mouse models. We are further able to show an inhibitory effect on tumor growth and survival in syngeneic tumor models responsive and unresponsive to PD-1 blockade. A relevant synergism combining AAV-sia and anti-PD-1 treatment was observed. Combination with human fucosidase (FUCA-1), sialidase and PD-1 was able to increase survival and the rate of complete cure up to 36%.Mechanistic studies have demonstrated an increased activation of T cells, a polarization of myeloid cells towards an anti-tumor phenotype and an increase in conventional dendritic cell infiltration. In addition, scRNAseq data shows an up-regulation of M1-like genes. Despite local injection and desialylation, we observed also a growth inhibition on distant tumor sites and an increase in tumor-specific T cells suggesting a systemic immune activation. We observed that cross- presentation is increased upon sialidase treatment.

Conclusions

Taken together, AAV-sia removes the immune-suppressant carbohydrate sialic acid from the tumor microenvironment and cancer cells rendering them more vulnerable for destruction by immune cells.

Legal entity responsible for the study

The authors.

Funding

SNF and KLBB.

Disclosure

N. Rodrigues Mantuano: Financial Interests, Personal and Institutional, Member of Board of Directors: Glycocalyx. H. Läubli: Financial Interests, Personal and Institutional, Member of Board of Directors: Glycocalyx. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

197P - Computational pathology pipeline enables quantification of intratumor heterogeneity and tumor-infiltrating lymphocyte score

Presentation Number
197P
Lecture Time
06:30 - 06:30
Speakers
  • Daniel G. Tiezzi (Ribeirao Preto, Brazil)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Tumor mutation burden, intratumor heterogeneity and the cancer related immune infiltrate are associated with response to target and immune therapies in several cancer types. The aim of this study is to establish a computational pathology (CPath) pipeline for investigating useful histopathological features in hematoxylin and eosin (H&E) whole slide images (WSIs). The pipeline allows for the segmentation and classification of nuclei. As an application, a machine learning approach is used to quantify intratumor heterogeneity (ITH) and tumor-infiltrating lymphocyte (TIL) scores.

Methods

We randomly selected 178 invasive ductal carcinomas WSIs from the TCGA database to process. Image annotation and nuclei detection were performed on QuPath by a pathologist using StarDist and used to train a SVM-based nuclei type classifier. The classifier was trained to distinguish between tumor, lymphocyte, and stroma. A total of 113.211 nuclei images were extracted using OpenSlide and used to train an autoencoder for dimensionality reduction. The variability of the WSIs was computed by applying a statistical measure of dispersion to the feature vectors. The TIL score was computed as the average minimum distance between a tumor cell and its nearest lymphocyte neighbor. We used the mutation burden, the ITH, PAM50 classification, nuclear and histological grades data from [https://doi.org/10.1038/nm.3984] to validate our ITH score. The percentage of infiltrating lymphocytes inferred by the quanTIseq pipeline based on RNAseq data was used to validate the SVM model.

Results

The nuclei classifier achieved 88% accuracy on the test set and the 96%/92% sensitivity to tumor/lymphocyte on a validation dataset. The mean squared error of the autoencoder was 0.0004 in the test set. The ITH score was associated with the clonal number of a tumor (x2 = 8.8, p= 0.03). Finally, we observed a positive correlation between the TIL/tumor ratio inferred by the CPath pipeline and the total number of T lymphocytes inferred by quanTIseq (Spearman= 0. 34, p < 0.0001).

Conclusions

We explored a CPath pipeline for detecting and classifying nuclei. The pipeline was used to compute ITH and TIL scores which correlated with validation data. Thus, validating our pipeline.

Legal entity responsible for the study

D.G. Tiezzi.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

198P - Polarization of tumor-associated macrophages enhanced by 2-HP-_-cyclodextrin modified PLGA nanoparticles

Presentation Number
198P
Lecture Time
06:30 - 06:30
Speakers
  • HAO H. YUAN (GUI YANG, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immunotherapy for cancer is closely related to the tumor immunosuppressive microenvironment (TIME). Resiquimod (a mixed TLR7/8 agonist also known as R848) has been shown to mediate promising immunostimulatory activity in a variety of preclinical models. Poly(D, L-lactic-co-glycolic acid) (PLGA) is one of the most commonly used biodegradable polymer.

Methods

The nanoparticles loaded with R848 (CD@R848@NP) were prepared by improved emulsion-solvent evaporation method, and the drug loading rate and entrapment efficiency were calculated. Its physical and chemical properties and drug release stability were measured, and its polarization effect on macrophages was evaluated in vitro. After labeling with IR780 fluorescent dye, the localization of the nanoparticles in vivo and in vitro was analyzed. CD@R848@NP was used to treat MC38 colon cancer mice by tail vein injection combined with intraperitoneal injection of TNFR2 antibody.

Results

(1) The prepared CD@R848@NP nanoparticles have strong stability, no obvious cytotoxicity, sustained release effect and increased release under acidic conditions. The load content (LC) and entrapment efficiency (EE) of R848 were (3.11 ±0.61)% and (65.36 ±3)%. (2) In vitro experiments showed that HP-β-CD modified nanoparticles could be better absorbed by macrophages, and in vivo localization experiments confirmed that they could be better located in tumor tissues. (3) CD@R848@NP combined with Anti-TNFR2 could eliminate MC38 tumor in mice, and there was no significant change in body weight. Hue results showed that the nanoparticles had no obvious toxicity to all organs. After 24 hours of administration, the secretion of IL-12 factor in blood of mice was significantly higher than that of other groups. The results of tumor histochemistry showed that the secretion of TNF α and IL-6 factors in CD@R848@NP was significantly higher than that in other groups.

Conclusions

PLGA nanoparticles encapsulated with R848 can effectively deliver drugs to tumor tissue and induce tumor-associated macrophages to polarize into M1 macrophages. On the other hand, TNFR2 antibodies can inhibit the activation of Treg cells, reduce immunosuppression in tumor microenvironment and eliminate tumor.

Legal entity responsible for the study

The authors.

Funding

The National Natural Science Foundation of China (82060308), Guizhou Immunotherapy Research Talent Base (RCJD2018-11).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

199P - Scalable multiplexed image analysis across cancer types as part of the IMMUcan consortium

Presentation Number
199P
Lecture Time
06:30 - 06:30
Speakers
  • Nils Eling (Zurich, Switzerland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Highly multiplexed tissue imaging (HMTI) technologies have enabled the in-depth characterization of the tumor immune microenvironment (TIME) and how it relates to patient prognosis and treatment success. To analyze the TIME across breast, head and neck, colorectal, lung, and kidney cancer, the Integrated iMMUnoprofiling of large adaptive CANcer patient cohorts (IMMUcan) consortium performs broad molecular and cellular profiling of more than 2500 cancer patients, associated with longitudinal clinical data. As part of IMMUcan, multiplexed immunofluorescence (mIF) imaging detects major cell phenotypes across whole tissue slides while imaging mass cytometry (IMC) provides a zoomed in view on local cell phenotype interactions.

Methods

We developed two computational pipelines to process and analyze mIF and IMC data. The analysis of mIF data is supported by IFQuant, a web-based tool for image analysis that facilitates user-guided cell phenotyping. IMC data is analyzed using a workflow encompassing image analysis and machine learning-based cell phenotyping. Various computational approaches have been developed for the spatial analysis of HMTI data. First, the tysserand package allows the construction of spatial networks from which the mosna package computes cell phenotype assortativity scores. Second, the Spacelet and Cellohood models were developed to analyze higher-order interactions between cell phenotypes in tumor tissues.

Results

Based on a selected set of ten samples from five cancer indications, we validate that all major cell phenotypes as detected by the two computational pipelines show high correlation between mIF and IMC. The computed assortativity measures can be used for the definition of cellular niches and patient stratification. The Spacelet and Cellohood models allow the association of gradients of immune infiltration patterns and cellular neighborhoods to clinical data.

Conclusions

We have developed scalable computational tools for the analysis of mIF and IMC data within the IMMUcan consortium. These form a crucial foundation for the robust extraction of molecular features across five cancer indications facilitating patient stratification and clinical association studies.

Legal entity responsible for the study

EORTC.

Funding

IMI2 JU grant agreement 821558, supported by EU’s Horizon 2020 and EFPIA.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

200P - A novel multi-organ on chip model for metastatic tumor biology understanding

Presentation Number
200P
Lecture Time
06:30 - 06:30
Speakers
  • Elisabetta Palama (Genova, Italy)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Understanding the complex biology of metastatic tumors is critical for advancing cancer biology research and improving therapeutic interventions. To address this challenge, we have developed a novel multi-organ-on-chip (Multi-OOC) platform designed to unravel the intricacies of breast to bone (B2B) metastatic tumor growth. This innovative platform integrates organ-specific tissue models fluidically interconnected to mimic the dynamic interactions between primary tumor and distant organ during metastasis. By replicating the physiological microenvironments of various target organs, our model provides a unique opportunity to investigate the entire metastatic cascade, from tumor cell dissemination, circulating tumor cells (CTCs) survival under flow and colonization. In order to support real-time monitoring of cellular infiltration and response to therapeutic agents in a multi-organ context optically transparent OOC device has been developed, compatible with the optical microscope observation.

Methods

A breast cancer cell laden hydrogel has been developed by using MDA-MB-231 cells with and without endothelial cells (HUVEC), forming the capillary network. A computational fluid dynamic simulation was done to set up the proper capillary velocity and induced shear stresses. In a second chamber, a bone tissue model was developed as metastasis target: different ratio of hydroxyapatite (HA) has been included in a polymeric matrix to introduce a bone like mineral phase. Tumor cell infiltration and CTC survival rate have been monitored using different fluid-dynamic conditions and HA content.

Results

A multicompartmental OOC has been developed and successfully validated. The 3D breast cancer model displayed long term (2 months) survival in vitro in dynamic conditions, and a cells cytoskeleton reorganization was highlighted. The CTCs survival was shown correlated to the shear stresses induced by the fluid flow into the Multi-OOC. Different levels of CTCs infiltration in the mineralized matrix hosted in the metastatic OOC chamber were observed.

Conclusions

This platform holds great promise for accelerating the development of targeted therapies and personalized treatment strategies, ultimately advancing our understanding of metastatic cancer biology.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

M. Aiello, S. Scaglione: Financial Interests, Personal, Member of Board of Directors: React4life. All other authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

201P - Role of aerobic exercise training on the mitochondrial metabolism and effector function of CD8+ tumor-infiltrating lymphocytes

Presentation Number
201P
Lecture Time
06:30 - 06:30
Speakers
  • Janaina D. Vieira (Sao Paulo, Brazil)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Immunotherapy provided a new treatment alternative for cancer, but many patients still do not respond due to tumor-induced immunological dysfunction. Aerobic exercise training (AET) has emerged as a strategy to reduce cancer mortality and improve immune response, however the mechanisms explaining AET benefits immune system on cancer remain unclear. Thus, our aim was to evaluate whether AET could positively modify the tumor microenvironment, favoring infiltration and cytotoxicity of “antitumor” lymphocytes by regulating their mitochondrial content and function.

Methods

Balb/c male mice were submitted to 4 weeks of AET (60% of maximal capacity, 5 days/week, 1 h/day) before inoculation (s.c.,1x106 CT26 colon carcinoma cells). CT26 TR continued AET after inoculation and all groups were euthanized 9 days post tumor cells inoculation for further analysis. Tumor volume was measured daily. Experimental groups were divided into control (healthy sedentary mice), CT26 SED (sedentary tumor-bearing mice) and CT26 TR (trained tumor-bearing mice). Tumor-infiltrated T lymphocytes and mitochondrial density were measured by flow cytometry. Tumor hypoxia was measured by Hypoxyprobe Kit. Mitochondrial morphology was evaluated by electron microscopy and ATP production by an ATP Determination Kit. Statistical analysis: Anova One-way, Duncan post hoc, p<0.05. Ethical approval: CEUA EEFE-USP 2017/02.

Results

CT26 TR mice showed attenuated tumor progression when compared with CT26 SED group, with significant smaller tumor volume and mass. AET significantly reduced tumor hypoxic area compared to CT26 SED group. AET significantly increased the total amount of activated and effector memory CD8+ T cells, and effector memory CD4+ T cells in CT26 TR when compared to CT26 SED in the tumor. Additionally, increased function of T CD8+ cells in CT26 TR was followed by a higher mitochondrial number/cell and function (ATP production) in this cells compared with CT26 SED group.

Conclusions

AET increases tumor-infiltrated cytotoxic/effector CD8+ T cells and improves their mitochondrial content and function. These data suggest that AET can attenuate tumor growth by modulation of tumor-infiltrated lymphocytes number and profile.

Legal entity responsible for the study

The authors.

Funding

São Paulo Research Foundation (FAPESP, 11800-4/2021, 2015/22814-5, and 2017/13133-0).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

202P - Discovery of immunological cellular neighborhoods from protein markers in spatial tumor data

Presentation Number
202P
Lecture Time
06:30 - 06:30
Speakers
  • Marcin Mozejko (Warsaw, Poland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Spatial imaging of single cells and their protein markers in multiple tumor tissues offers detailed insights into the mechanisms of tumor-microenvironment interactions. The identification and characterization of cellular neighborhoods are vital to elucidating these mechanisms. However, existing approaches for cellular neighborhood analyses resort to predefined cell types or coarse, neighborhood-wide cell marker aggregations, failing to preserve the marker information at the single-cell level resolution.

Methods

We developed Cellohood - the first permutation-invariant, transformer-based autoencoder designed to model cellular neighborhoods explicitly. Cellohood compresses information about each cell and its marker expression for a given neighborhood, providing representations of the cellular neighborhoods. Based on these representations, we derived novel cellular neighborhood prototypes. Cell types, protein markers, and spatial arrangement patterns describe each identified prototype. Consequently, patients can be described by the abundance of cellular neighborhood prototypes and their mutual arrangements.

Results

We showcased the performance of Cellohood by applying it to the Imaging Mass Cytometry data from a non-small cell lung cancer (NSCLC) patient cohort (N=192) from the IMMUcan (Integrated iMMUnoprofiling of large adaptive CANcer patient cohorts) consortium. Prototypes discovered by Cellohood, including tumor neighborhoods characterized by i) low B2M expression, ii) IDO1+ tumor cells, iii) plasma cell enrichment, and iv) T-reg infiltration, defined patient clustering that surpassed the TNM cancer staging in terms of survival prediction. Analysis of macrophage-enriched neighborhoods indicated that CD206+ macrophages are a key differentiating factor between cancer histologies.

Conclusions

Thanks to transformer-based generative modeling, Cellohood is the first model to utilize complete cell marker information during training without resorting to coarse, neighborhood-wide approximation. Results on the NSCLC cohort from the IMMUcan consortium demonstrated that Cellohood enables marker-driven discovery of tumor-microenvironment interactions and their clinical implications.

Legal entity responsible for the study

IMMUcan (Integrated iMMUnoprofiling of large adaptive CANcer patient cohorts).

Funding

IMI2 JU grant agreement 821558, supported by EU’s Horizon 2020 and EFPIA.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

203P - Identification of High Confidence Candidate Markers for Macrophage Infiltrating Tumor Microenvironment through Single Cell Genomic Atlases

Presentation Number
203P
Lecture Time
06:30 - 06:30
Speakers
  • Constance Ciaudo Beyer (Zurich, Switzerland)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

The interaction between the immune system and the tumor microenvironment significantly impacts cancer progression and treatment response. Macrophages, with their diverse functional states, play a crucial role in this interplay. Identifying accurate markers for macrophage infiltration is vital for understanding their roles and therapeutic potential.

Methods

Immunai addresses this challenge by developing single cell atlases. Employing state-of-the-art single cell RNA sequencing (scRNA-seq) technologies, Immunai generates comprehensive and high-resolution transcriptomics data from diverse tumor samples, encompassing a wide range of cancer types. The establishment of scRNA-seq data atlases and references enables the identification of distinct macrophage populations within the tumor microenvironment, as well as their heterogeneity and functional states. Utilizing advanced computational algorithms and analytical pipelines, Immunai systematically analyses scRNA-seq datasets to uncover specific molecular signatures associated with macrophage infiltration.

Results

In the presented work, Immunai's approach enabled the discovery of novel macrophage-specific markers that can serve as valuable tools for both basic research and clinical applications. The identified markers offer insights into the dynamic nature of macrophage populations within the tumor microenvironment and provide potential avenues for the development of immunotherapies targeting tumor-associated macrophages. Moreover, these markers hold promise as diagnostic and prognostic indicators, aiding in patient stratification and personalized treatment strategies.

Conclusions

Immunai's approach provides a significant contribution to immuno-oncology and novel therapeutic approaches.

Legal entity responsible for the study

Immunai.

Funding

Immunai.

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

204P - Effect of STAT6 Inhibition as a Novel Strategy for Promoting the Induction of Potent and Stable Regulatory T Cells for Use in Colitis Therapy

Presentation Number
204P
Lecture Time
06:30 - 06:30
Speakers
  • SONIA LEON-CABRERA (Ciudad de Mexico, Mexico)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00

Abstract

Background

Colorectal cancer (CRC) ranks among the leading global malignancies, characterized by high mortality and incidence rates. Furthermore, inflammatory bowel disease has been implicated in the development of CRC associated with colitis (CAC). STAT6 is notably hyperactivated in various tumors, including CRC. In experimental CAC models, STAT6 KO mice have demonstrated decreased tumor development, reduced disease aggressiveness, and a reduced local and systemic inflammatory response compared to WT mice. This phenomenon has been attributed to an increased presence of Tregs during the early stages of CAC development. As such, we seek to investigate the differentiation and functional characteristics of induced Tregs (iTregs) generated in the presence of the specific STAT6 inhibitor, AS1517499, and evaluate its therapeutic potential in an in vivo CAC model.

Methods

Naive CD4 T cells were isolated from Foxp3eGFP mice and cultured under Treg-polarizing conditions with or without AS1517499 for five days. Subsequently, cells were expanded with IL-2 supplementation over a total of 15 days, followed by staining with anti-CD4 and anti-CD25 antibodies and analysis via FACS. Foxp3+ cells were isolated and the extent of differentiation and suppressive capability of these cells were assessed. Next, 2.0 × 105 cells were injected into WT mice using an AOM/DSS regimen. Over ten weeks, we monitored changes in body weight, disease activity index, colon length, and tumor burden. Additionally, we evaluated the severity of colon inflammation and markers of carcinogenesis.

Results

iTregs induced under STAT6 inhibition exhibited prolonged expression of Foxp3 and CD25, even during inflammatory conditions. The transfer of Tregs/AS1517499 into WT mice in the AOM/DSS model resulted in a remarkable reduction in inflammation, decreased expression of inflammatory cytokines, and a reduction in epithelial cell proliferation.

Conclusions

iTregs developed under STAT6 inhibition demonstrate enhanced stability when compared to their littermate counterparts. The transfer of Tregs/AS1517499 during inflammatory states leads to an improvement in the disease status of CAC in the in vivo model.

Legal entity responsible for the study

The authors.

Funding

Consejo Nacional De Humanidades Ciencia Y Tecnologia De Mexico.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display (ID 34) Poster Display

125P - Efficacy and safety of the therapeutic DNA vaccine VB10.16 in combination with atezolizumab in advanced HPV16-positive cervical cancer: Updated analysis from the VB-C-02 trial

Presentation Number
125P
Lecture Time
06:30 - 06:30
Speakers
  • Kristina Lindemann (Oslo, Norway)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

87P - Real-world analysis of atezolizumab in ES-SCLC patients and those with brain metastases: An improved survival outcome

Presentation Number
87P
Lecture Time
06:30 - 06:30
Speakers
  • Minglei Zhuo (Beijing, China)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

2P - Beyond PD-L1: unraveling the enigma of immunotherapy response in PD-L1 negative NSCLC patients through quantification of PD-1/PD-L1 engagement in the tumour microenvironment.

Presentation Number
2P
Lecture Time
06:30 - 06:30
Speakers
  • James Miles (Bilbao, Spain)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00
Poster Display (ID 34) Poster Display

8P - Exploration of Potential Biomarkers Correlated with Efficacy of Ociperlimab (OCI) plus Tislelizumab (TIS) in 1L PD-L1+ Non-Small Cell Lung Cancer (NSCLC)

Presentation Number
8P
Lecture Time
06:30 - 06:30
Speakers
  • Se Hyun Kim (Seongnam, Korea, Democratic People's Republic of)
Session Name
Poster Display (ID 34)
Room
Foyer mezzanine
Date
Thu, 07.12.2023
Time
12:00 - 13:00