Displaying One Session

Foyer mezzanine Thu, Dec 8, 2022
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15
Location
Foyer mezzanine
Chairs
  • Samra Turajlic (London, United Kingdom)
  • E.G. Elisabeth De Vries (Groningen, Netherlands)
  • Kevin J. Harrington (London, United Kingdom)
  • Lisa H. Butterfield (San Francisco, CA, United States of America)
Poster Display Poster Display session

4P - FDG PET derived Metabolic Tumor Volume (MTV) and its transcriptomic correlates as biomarker to predict efficacy of immune checkpoint inhibitors (ICB) alone or in combination with chemotherapy in advanced NSCLC: A multicentric study. (ID 105)

Presentation Number
4P
Lecture Time
12:30 - 12:30
Speakers
  • Filippo G. Dall'Olio (Bologna, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

ICBs are today the backbone of non-oncogene addicted NSCLC, both alone and in combination with chemotherapy (CT-ICB). To date, PD-L1 is the only predictive factor for ICBs benefit. Tumor burden is a new emerging additional biomarker to select pts who may derive a benefit from a CT combination compared to single agent ICBs. The biological correlates of tumor burden are largely unknown.

Methods

In this multicentric study, 18F FDG PET scans were performed ≤ 42 days from 1st line initiation (CT, ICBs or CT-ICB). Total MTV was calculated with a threshold of 42% of SUV max. Progression Free Survival (PFS) and Overall Survival (OS) were analyzed with Kaplan Meyer method and log rank test. On a subset of pts with PET performed within 30 days from biopsy, transcriptomic data on fresh frozen tissue were correlated with MTV of the biopsied lesion.

Results

488 pts were enrolled, 162 treated with ICBs, 232 with CT-ICB and 94 with CT. The 3 groups were comparable in terms of MTV, sex, stage and PS, pts in ICBs are older (69y vs 64 for CT-ICB and 62 for CT). MTV was negatively correlated with the outcome of ICI, with a median PFS of 31.8 months for 1st quartile (1Q), 13.8 for 2Q, 4.7 for the 3Q and 1.8 for the 4Q (p < 0.001). A weaker correlation was found for CT-ICB, with a median PFS of 14.0m 1Q, 7.1 for 2Q, 7.0 for 3Q and 5.5 for 4Q (any PD-L1 expression), p 0.037, No correlation was found for CT ( p 0.696). When comparing ICI to CT-ICB in patients with PD-L1 ≥ 50%, an advantage in PFS was seen for CT-ICB (10.7 vs 3.0m, p 0.039 ) for pts with MTV > median (100cc) as well as in 3 (73% vs 92%, p 0.043) and 6 months OS (66 vs 89%, p 0.033), but not in those with MTV < median. Transcriptome analysis was performed on 48 pts and showed a negative correlation of MTV with GSEA immune signatures, as well as with RNAseq immune infiltrate quantification (Cybersort, p 0.016).

Conclusions

MTV identifies NSCLC pts with a worse prognosis and an increased rate of early progression and death on ICB. In the subgroups of PD-L1≥50%, pts with high MTV may do better on CT-ICB compared to ICB. Transcriptomic analysis suggests that tumor microenvironment become increasingly immune suppressive with the increase in volume.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

G. Zalcman: Financial Interests, Personal, Invited Speaker: Bristol Myers Squibb, AstraZeneca, Pfizer, Boehringer Ingelheim; Non-Financial Interests, Personal, Invited Speaker: Roche-France, Bristol Myers Squibb, Takeda. J. Remon Masip: Financial Interests, Personal, Invited Speaker: Roche, Pfizer, MSD, Boehringer Ingelheim; Financial Interests, Personal, Advisory Board: AstraZeneca, BMS, Jansse, Takeda, Sanofi; Financial Interests, Personal, Expert Testimony: OSE Immunotherapeutics; Non-Financial Interests, Personal, Principal Investigator, PI of PECATI trial in Thymic malignancies endorsed by a grant by MSD: MSD; Non-Financial Interests, Personal, Other, Co-PI of APPLE trial (EORTC-1525): AstraZeneca. L. Hendriks: Financial Interests, Institutional, Advisory Board: Amgen, Boehringer Ingelheim, Lilly, Novartis, Pfizer, Takeda, BMS, Merck, Janssen, MSD; Financial Interests, Personal, Other, mentorship with key opinion leaders funded by AstraZeneca: AstraZeneca; Financial Interests, Institutional, Invited Speaker, for educational webinar: AstraZeneca, Lilly; Financial Interests, Institutional, Invited Speaker, educational webinar/interview: Bayer; Financial Interests, Institutional, Invited Speaker, educationals: MSD; Financial Interests, Personal, Invited Speaker, for webinars: Medtalks; Financial Interests, Institutional, Advisory Board, one time also personal: Roche; Financial Interests, Institutional, Other, performing interviews at conference: Roche; Financial Interests, Personal, Other, travel support: Roche; Financial Interests, Institutional, Other, podcast on brain metastases: Takeda; Financial Interests, Personal, Invited Speaker, payment for post ASCO round table discussion: VJOncology; Financial Interests, Personal, Invited Speaker, payment for post ASCO/ESMO/WCLC presentations, educational committee member: Benecke; Financial Interests, Institutional, Invited Speaker, payment for post ESMO/ASCO discussion: high5oncology; Financial Interests, Institutional, Other, educational webinar: Janssen; Financial Interests, Personal, Other, member of the committee that revised these guidelines: Dutch guidelines NSCLC, brain metastases and leptomeningeal metastases; Financial Interests, Institutional, Research Grant, for IIS: Roche, Boehringer Ingelheim, AstraZeneca, Takeda; Financial Interests, Institutional, Invited Speaker: AstraZeneca, GSK, Novartis, Merck Serono, Roche, Takeda, Blueprint Medicines, Mirati, AbbVie, MSD, Gilead; Financial Interests, Institutional, Other, drug support for IIS (contract in negotiation): Beigene; Non-Financial Interests, Personal, Other, Chair metastatic NSCLC for lung cancer group: EORTC; Non-Financial Interests, Personal, Other, secretary NVALT studies foundation: NVALT. C. Helissey: Financial Interests, Personal, Invited Speaker: Janssens, Roche, Astellas, AstraZeneca, Sanofi; Non-Financial Interests, Personal, Principal Investigator: Janssen, Sanofi, Roche, Astellas. I. Monnet: Other, Personal, Other, invitation to virtual ASCO 2021 and 2022: Pfizer; Other, Personal, Other, invitation to ESMO congress 2021 and 2022: Takeda. D. Planchard: Financial Interests, Personal, Advisory Board: AstraZeneca, BMS, Merck, Novartis, Pfizer, Roche, Samsung, Celgene, AbbVie, Daiichi Sankyo, Janssen; Financial Interests, Personal, Invited Speaker: AstraZeneca, Novartis, Pfizer, priME Oncology, Peer CME, Samsung, AbbVie, Janssen; Non-Financial Interests, Personal, Principal Investigator, Institutional financial interests: AstraZeneca, BMS, Merck, Novartis, Pfizer, Roche, Daiichi Sankyo, Sanofi-Aventis, Pierre Fabre; Non-Financial Interests, Personal, Principal Investigator: AbbVie, Sanofi, Janssen. F. Barlesi: Financial Interests, Personal, 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; Non-Financial Interests, Personal, Principal Investigator: AstraZeneca, BMS, Merck, Pierre Fabre, F. Hoffmann-La Roche Ltd. 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, Janssen, Onxeo, OSE Immunotherapeutics, Pfizer, 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 Poster Display session

5P - Tumor microenvironment cellular crosstalk predicts response to adoptive TIL therapy in melanoma patients (ID 106)

Presentation Number
5P
Lecture Time
12:30 - 12:30
Speakers
  • Denarda Dangaj (Lausanne, Switzerland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TILs) has proven efficacious in metastatic melanoma (Mel). Nevertheless, long-term clinical efficacy remains unsatisfactory for the majority of patients. Therefore, understanding TILs and the orchestrated tumor-microenvironment (TME) states associated with clinical response is of paramount importance.

Methods

We report the translational analysis of a single-center phase I study to assess feasibility and efficacy of TIL-ACT in Mel patients (NCT03475134). We performed comprehensive translational studies on patients` tumor longitudinal samples including multispectral immuno-fluorescence (mIF) imaging, bulk RNA-sequencing and single-cell RNA-sequencing before and after TILs infusion (30 days).

Results

Thirteen patients successfully completed TIL-ACT therapy and were included in the analysis. Best overall response was 46.1% (6/13) at 3 months, including two patients with ongoing complete response 3 years after infusion. Responders (Rs) exhibited immunogenic tumor cell states with higher inferred CNVs and overexpressed DNA sensing/IFN and class I antigen presentation-related genes. Multiplex IF imaging revealed that Rs had increased densities of polyfunctional intra-epithelial (ie)CD8+ T cells marked by higher PD-1 expression at baseline, and these cells persisted 30 days after TILs infusion. Single-cell transcriptomic analyses confirmed higher cytotoxic and exhaustion CD8 T cell states at baseline. PDCD1 (PD-1), CXCL13, TNFRSF9 (CD137), GZMB, HAVCR2 (TIM3) and PRF1 genes were overexpressed in CD8+ TILs and associated to clinical responses. Cell-cell interaction predictions corroborated by spatial neighborhood analyses revealed that tumors of Rs were highly enriched for ie-and stromal T-cell networks involving activated myeloid and costimulatory B cells. Successful TIL-ACT therapy reprogrammed the myeloid compartment and further increased CD8 TIL-myeloid cell networks.

Conclusions

Our systematic target discovery study reveals CD8 T-cell network-based biomarkers that could improve patient selection and guide the design of ACT clinical trials.

Clinical trial identification

NCT03475134.

Legal entity responsible for the study

Centre Hospitalier Universitaire Vaudois (CHUV), Ludwig Institute for Cancer Research.

Funding

Centre Hospitalier Universitaire Vaudois, Lausanne Branch of the Ludwig Insitute for Cancer Research.

Disclosure

O.A. Michielin: Financial Interests, Institutional, Invited Speaker: BMS, Amgen, Pierre Fabre, Roche, BMS; Financial Interests, Institutional, Advisory Board: BMS, Amgen, Roche, Novartis, Pierre Fabre, MSD; Financial Interests, Personal, Other, Advisory Role: BMS, MSD, Roche, Novartis, Pierre Fabre, Amgen, GSL; Financial Interests, Institutional, Research Grant: BMS, MSD, Pierre Fabre, Amgen, Merck; Financial Interests, Institutional, Funding: BMS, MSD, Pierre Fabre, Amgen, MSD; Non-Financial Interests, Personal, Principal Investigator: BMS, MSD, Amgen, Roche, Pierre Fabre, Novartis. G. Coukos: Financial Interests, Personal, Invited Speaker: CHUV. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

6P - Response and survival according to the interferon-gamma (IFN-y) signature and tumor mutational burden (TMB) in the PRADO trial testing neoadjuvant ipilimumab and nivolumab in stage III melanoma (ID 107)

Presentation Number
6P
Lecture Time
12:30 - 12:30
Speakers
  • Irene Reijers (Amsterdam, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The PRADO trial (n=99) tested different surgical and adjuvant therapy strategies based on the pathologic response after neoadjuvant IPI 1mg/kg and NIVO 3mg/kg in stage III melanoma patients (pts). The pathologic response rate (pRR: ≤50% viable tumor) was 72%, including 61% major pathologic responses (MPR: ≤10% viable tumor). After a median follow-up of 28.1 months, the 2-year (2y) event-free survival (EFS) rate was 80%. Here, we report the response and EFS data of PRADO according to the IFN-γ signature and TMB.

Methods

TMB and the IFN-γ gene expression signature (GES) were examined in baseline tumor biopsies by whole exome sequencing (n=76) and mRNA sequencing (n=80). Association with pRR, MPR or EFS was examined by logistic/Cox regression analyses. Cutoffs between high (H) and low (L) were calculated using ROC curves.

Results

The table shows the association between IFN-γ GES and TMB with pRR, MPR, and EFS. IFN-γ GES and logTMB were not correlated (R = 0.065; p = 0.579). Pts with a high IFN-γ GES had a higher pRR and MPR rate than pts with a low IFN-γ GES (87% vs 50%, p=0.001 and 77% vs 35%, p<0.001, respectively), and also a higher 2y EFS rate (87% vs 68%, log-rank p=0.015). Pts with a high TMB were more likely to achieve a pathologic response (pRR 83% vs 58%, p=0.024) and MPR (80% vs 38%, p<0.001), but did not have a lower risk of relapse (2y EFS 77% vs 76%, log-rank p=0.531). When combined, the pRR for pts with IFN-γ H/TMB H was 90%, IFN-γ H/TMB L 79%, IFN-γ L/TMB H 67% and IFN-γ L/TMB L 42%. For MPR rates this was 85%, 63%, 67% and 19%, respectively.

Univariable analysis
OR / HR 95% CI p-value
pRR
IFN-γ score (continuous) 1.089 (1.027-1.156) 0.004
IFN-γ high vs low 6.800 (2.208-20.944) 0.001
logTMB (continuous) 1.311 (0.988-1.741) 0.061
TMB high vs low 3.654 (1.183-11.286) 0.024
MPR
IFN-γ score (continuous) 1.092 (1.031-1.157) 0.003
IFN-γ high vs low 6.190 (2.304-16.634) <0.001
logTMB (continuous) 1.376 (1.037-1.826) 0.027
TMB high vs low 6.588 (2.240-19.374) 0.001
EFS
IFN-γ score (continuous) 0.965 (0.922-1.011) 0.130
IFN-γ high vs low 0.308 (0.112-0.846) 0.022
logTMB (continuous) 0.935 (0.726-1.205) 0.604
TMB high vs low 0.747 (0.298-1.872) 0.533

Conclusions

Similar to findings in our previous trials, baseline IFN-γ GES and TMB were independent biomarkers for pathologic response and MPR, and the IFN-γ GES was associated with EFS. However, TMB was not associated with risk of relapse, possibly owing to the different response-driven surgical and adjuvant therapy strategies in PRADO.

Clinical trial identification

NCT02977052.

Legal entity responsible for the study

Netherlands Cancer Institute.

Funding

BMS.

Disclosure

R. Saw: Financial Interests, Personal, Invited Speaker: Bristol Myers Squib, Novartis; Financial Interests, Personal, Advisory Board: Novartis, MSD, Qbiotics; Financial Interests, Personal, Other, On Faculty, support of University of Sydney salary: Melanoma Institute Australia. E. Kapiteijn: Financial Interests, Institutional, Advisory Board: BMS, Novartis, Pierre Fabre, Merck, Delcath, Bayer; Financial Interests, Institutional, Invited Speaker: BMS. A.A.M. Van der Veldt: Financial Interests, Institutional, Advisory Board: BMS, MSD, Merck, Roche, Eisai, Pfizer, Sanofi, Novartis, Pierre Fabre, Ipsen. K. Suijkerbuijk: Financial Interests, Institutional, Advisory Board: Novartis, BMS, AbbVie, Pierre Fabre, MSD; Financial Interests, Institutional, Invited Speaker: Roche; Financial Interests, Institutional, Research Grant: Novartis, TigaTx. G. Hospers: Financial Interests, Institutional, Advisory Board: Amgen, Roche, MSD, BMS, Novartis, Pierre Fabre, Pfizer; Financial Interests, Institutional, Research Grant: BMS, Seerave. W. Van Houdt: Financial Interests, Institutional, Invited Speaker: Amgen, BMS; Financial Interests, Institutional, Advisory Board: Belpharma; Financial Interests, Institutional, Expert Testimony: Sanofi, MSD; Financial Interests, Personal, Other, travel grant: Novartis. R. Scolyer: Financial Interests, Institutional, Advisory Board: F. Hoffmann-La Roche, Evaxion, Provectus Biopharmaceuticals Australia, QBiotics, Novartis, MSD, NeraCare, Amgen, BMS, Myriad Genetics, GlaxoSmithKline. A.M. Menzies: Financial Interests, Personal, Advisory Board, advisory board: BMS, MSD, Novartis, Roche, Pierre-Fabre, QBiotics. A.C.J. van Akkooi: Financial Interests, Institutional, Advisory Board: Amgen, Bristol Myers Squibb, Novartis, MSD - Merck, Merck-Pfizer, Pierre Fabre, Sanofi, Sirius Medical, 4SC, Provectus; Financial Interests, Institutional, Research Grant, NIVEC study: Amgen; Financial Interests, Institutional, Research Grant: Merck-Pfizer. G.V. Long: Financial Interests, Personal, Other, Consultant Advisor: Agenus Inc, Amgen Inc, Array Biopharma Inc., Boehringer Ingelheim International GmbH, Bristol Myers Squibb, Evaxion Biotech A/S, Hexal AG (Sandoz Company), Merck Sharpe & Dohme (Australia) Pty Limited, Novartis Pharma AG, OncoSec Medical Australia, Pierre Fabre, Provectus Australia, Qbiotics Group Limited, Regeneron Pharmaceuticals Inc; Financial Interests, Personal, Advisory Board, Consultant Advisor: Highlight Therapeutics S.L. C.U. Blank: Financial Interests, Institutional, Advisory Board: BMS, MSD, Roche, Novartis, GSK, AZ, Pfizer, Lilly, GenMab, Pierre Fabre; Financial Interests, Personal, Expert Testimony: Third Rock Ventures; Financial Interests, Personal, Stocks/Shares: Immagene; Financial Interests, Personal, Stocks/Shares, intention to develop IFN signature algorithm: NewCo, no name yet; Financial Interests, Institutional, Invited Speaker: BMS, Novartis, NanoString, 4SC; Other, Personal, Other, pending patent: WO 2021/177822 A1. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

7P - PrimeCUTR: Identifying clinically relevant neoantigens in the untranslated regions of cancer genomes. (ID 108)

Presentation Number
7P
Lecture Time
12:30 - 12:30
Speakers
  • Christopher C. Sng (London, United Kingdom)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Tumour-specific antigens are one of the principal targets in immune checkpoint inhibitor (CPI) treatment and fundamental to the development of personalised immunotherapy. The search for immunogenic neoantigens has primarily focused on mutations in protein-coding regions of genes. In this study, we investigate how novel open-reading frames (neoORFs) in the 5’ and 3’ untranslated region (UTR) of genes (generated by premature start-gain and stop-loss mutations respectively) contribute to the immune landscape of cancer.

Methods

We analysed two pan-cancer CPI-treated patient cohorts from the Hartwig Medical Foundation (HMF, n = 384), and Genomics England (GEL, n = 364). The frequency and length of UTR neoORFs for each patient tumour were predicted from whole genome sequencing data using PrimeCUTR, an R-based bioinformatics tool we developed. CPI treatment response was determined based on RECIST 1.1 criteria.

Results

Start-gain neoORFs occurred in 65.4–75.5% (GEL–HMF) of patients, while stop-loss neoORFs were found in 18.6–30.5%. 53.2–65.6% had start-gain neoORFs spanning 20 amino acids or more, with an overall median length of 28 amino acids. Among prevalent cancer groups, lung and head and neck cancer had the longest start-gain neoORFs. CPI response in HMF was associated with longer total length of start-gain (but not frameshift or stop-loss) neoORFs (P = 0.005). This association was also seen subsetting for low tumour mutational burden (TMB) (P = 0.014). In GEL, survival benefit was specifically seen in melanoma (n = 155), where patients with start-gain neoORFs longer than 28 amino acids (n = 60) had greater OS compared to those without (HR 0.59, 95% CI 0.36–0.95, P = 0.03).

Conclusions

UTR neoORFs occur frequently in cancer, yielding a promising source of neoantigens. We designed a computational pipeline for identifying these neoORFs, which will be made available upon peer-review. We show that 5’ UTR start-gain neoORFs were associated with clinical response to CPI, even in the low TMB setting which is typically linked to reduced CPI response. These findings warrant further study to validate UTR neoORFs as a biomarker and target for personalised immunotherapy.

Legal entity responsible for the study

The authors.

Funding

National Institute for Health and Care Research.

Disclosure

K.R. Litchfield: Financial Interests, Personal, Invited Speaker: Roche Tissue Diagnostics; Financial Interests, Personal, Other, Consulting work: Monopteros Therapeutics; 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 Poster Display session

8P - Phenotypic characterization of infused tumor-infiltrating lymphocytes (TIL) correlates with response to adoptive cellular therapy (ACT) in patients with metastatic melanoma (MM) (ID 109)

Presentation Number
8P
Lecture Time
12:30 - 12:30
Speakers
  • Joachim Stoltenborg Granhøj (Herlev, Denmark)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

ACT with TIL is personalized immunotherapy with promising results in MM. Recently, the positive outcome of a multicenter (National Center for Cancer Immune Therapy [Herlev, DK] and the Netherlands Cancer Institute [Amsterdam, NL]) phase 3 trial evaluating TIL in MM patients (NCT02278887) was recently announced. In this trial, the objective response (OR) rate for TIL-treated patients was 49% with 20% obtaining a complete response according to RECIST 1.1. However, there are currently no validated biomarkers that correlate with response to TIL. Identifying cellular subsets associated with response to treatment could in the future help guide clinicians in treating MM patients with TIL.

Methods

In this study, we analyzed the cryopreserved samples of the TIL infusion product from all patients with unresectable stage IIIC-IV MM treated with TIL in the phase 3 trial using multiparametric flow cytometry. The phenotypic characteristics of the TIL infusion product and OR to therapy according to RECIST 1.1 were investigated. The Mann-Whitney U test was used to test unpaired observations for statistical significance.

Results

In total, 80 patients received TIL, with 39 responders (complete + partial response) and 39 non-responders (stable + progressive disease). For 2 patients, the response was unevaluable. A higher number of TILs infused (Table) was significantly associated with response to therapy (p=0.013). Several lymphocyte subsets, including a higher number of CD8+ (p=0.004), CD8+CD28+ (p<0.001), CD8+BTLA+ (p=0.004), and CD8+CD39+ cells (p=0.007) favored response to therapy.

Absolute number of cells infused (x109)

Subset Responders (n=39) Non-responders (n=39) p-value
Total TIL 44.3 (38.4 - 51.2) 36.0 (29.6 - 43.5) 0.013
CD8+ 33.0 (22.9 - 36.8) 22.5 (15.8 - 26.1) 0.004
CD8+CD28+ 12.6 (9.5 - 15.6) 5.6 (4.0 - 9.1) <0.001
CD8+BTLA+ 6.8 (4.3 - 11.9) 3.0 (2.2 - 4.6) 0.004
CD8+CD39+ 29.3 (18.9 - 35.7) 20.8 (12.5 - 24.7) 0.007

Median (95 % confidence interval)

Conclusions

In this study, we present an explorative immune characterization of the melanoma TIL infusion product and identify several cellular subsets that correlate with response to TIL therapy.

Legal entity responsible for the study

National Center for Cancer Immune Therapy (Herlev, DK) and the Netherlands Cancer Institute (Amsterdam, NL).

Funding

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

Disclosure

T. Holz Borch: Financial Interests, Personal, Other, Honorary lectoures: Bristol Meyer Squibb. M. Donia: Financial Interests, Personal, Invited Speaker, Teaching: Novartis, Roche; Financial Interests, Personal, Other, Advisor: Achilles Therapeutics; Non-Financial Interests, Personal, Other, Sub-investigator of clinical trial with connected translational research: Bristol Myers Squibb; Financial Interests, Personal, Proprietary Information, Proprietary Data Access: Bristol Myers Squibb; Other, Institutional, Other, Chariman 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 Squibb, Achilles Therapeutics, Ipsen, Merck Sharpe & Dohme, Merck Serono, Pfizer, Molecular Partners, Novartis, Roche, Sanofi, Third Rock Venture, Iovance Biotherapeutics; 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, Stocks/Shares: Neogene Therapeutics; Financial Interests, Institutional, Research Grant: Bristol Myers Squibb, BioNTech US, Merck Sharpe & Dohme, Amgen, Novartis, Asher Bio; Non-Financial Interests, Personal, Member: ASCO, AACR, SITC; Other, Personal, Other, Editorial Board ESMO Open: ESMO; Other, Personal, Other, Editor-in-Chief IOTECH: 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, 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, Roche, TILT Biotherapeutics, Lytix Biopharma, Novartis. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

9P - Early circulating tumor DNA (ctDNA) kinetics and gene expression analysis to predict treatment outcomes with anti-PD-1 therapy (ID 110)

Presentation Number
9P
Lecture Time
12:30 - 12:30
Speakers
  • Enrique Sanz Garcia (Toronto, ON, Canada)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

ctDNA kinetics in the first 6 weeks and immune-related gene expression signatures can predict outcomes in patients (pts) treated with anti-PD-1 therapy. The earliest predictive timepoint in ctDNA kinetics for benefit to this therapy is unknown.

Methods

Pts with recurrent/metastatic head and neck squamous cell carcinoma treated with pembrolizumab or nivolumab monotherapy were prospectively enrolled. Plasma was collected at baseline (B), 2, 3, 8, 15, 22 and 29 days after first dose. Whole exome sequencing (WES) and RNA-seq were performed in archival tumor. ctDNA was analyzed using a bespoke 16 gene panel based on matched WES (SignateraTM). Differential Gene Expression Analysis was performed with DESeq2. Gene Set Enrichment Analysis included different transcription factors (TF), molecular/immune pathways and PREDICT-IO signature (PMID 36055464).

Results

A total of 104 plasma samples from 15 pts were collected. ctDNA was detected in 90/104 (87%). Clinical Benefit (CB) rate, defined as complete (CR), partial response (PR) or stable disease (SD) > 6 months by RECIST 1.1, was 47% (1 CR, 3 PR, 3 SD). There were no differences in ctDNA quantity at B according to local/distant disease, primary site or PD-L1 status. A decrease in ctDNA at day 8 from B was associated with CB (100 vs 14%, p<0.01) and with longer progression-free survival (PFS) adjusted by Holm-correction for multiple comparisons (HR: 22.9 95%CI 2.5-211.3, p=0.02) compared to a stability/increase. This association was also seen at day 15, 22 and 29 but not at day 2 or 3. A steady decline in ctDNA beyond day 8 (↓ΔctDNA) was observed in 4 pts (3 PR, 1 SD). Immune-related pathways and TF were significantly enriched in CR/PR and ↓ΔctDNA. Oncogenic pathways and TF were enriched in pts with increase/stability beyond day 8 (↑ΔctDNA). PREDICT-IO High was associated with longer PFS (HR: 7.7 95%CI 1.5-38, p=0.005). All PREDICT-IO Low pts were ↑ΔctDNA (N=7). Within PREDICT-IO High pts, ↓ΔctDNA showed a trend to longer PFS than ↑ΔctDNA (HR: 6.9 95%CI 0.95-63, p=0.051).

Conclusions

A decrease in ctDNA by day 8 of anti-PD1 therapy is associated with CB and longer PFS. ctDNA kinetics can improve predictive value of PREDICT-IO High signature. Validations in larger pan-cancer cohorts are needed.

Clinical trial identification

NCT04606940.

Legal entity responsible for the study

Princess Margaret Cancer Centre.

Funding

Natera.

Disclosure

G. Laliotis: Financial Interests, Personal, Full or part-time Employment: Natera. A. Spreafico: Financial Interests, Personal, Advisory Board: Merck, Bristol Myers Squibb, Oncorus, Jansen, Medison and Immunocore; Financial Interests, Institutional, Principal Investigator, Clinical trial: Novartis; Financial Interests, Institutional, Principal Investigator: Bristol Myers Squibb, Symphogen, AstraZeneca/MedImmune, Merck, Bayer, Surface Oncology, Northern Biologics, Janssen Oncology/Johnson and Johnson, Roche, Regeneron, Alkermes, Array Biopharma, GSK, Oncorus, Treadwell, Amgen. A.R. Hansen: Financial Interests, Personal, Advisory Board: GSK, Merck, Eisai; Financial Interests, Institutional, Principal Investigator: GSK, Merck, Pfizer, MedImmune/Genetech, Roche, Janssen, BMS, AstraZeneca, Astellas, Boehringer, Bayer. S. Willingham: Financial Interests, Personal, Full or part-time Employment: Natera. M. Liu: Financial Interests, Personal, Full or part-time Employment: Natera. T. Pugh: Financial Interests, Personal, Advisory Board: Illumina, Merck, Chrysalis Biomedical Advisors, Canadian Pension Plan Investment Board; Financial Interests, Institutional, Research Grant: Roche/Genentech. S. Bratman: Financial Interests, Personal, Leadership Role: Adela; Financial Interests, Personal, Stocks/Shares: Adela; Financial Interests, Personal, Royalties: Roche. L.L. Siu: Financial Interests, Personal, Advisory Board: Merck, AstraZeneca, Roche, Oncorus, Seattle Genetics, Voronoi, Arvinas, Tessa, Navire, Relay Therapeutics, Janpix, Amgen, Marengo, InterRNA, Medicenna, Hoopika, Coherus; Financial Interests, Personal, Other, Spouse is co-founder: Treadwell Therapeutics; Financial Interests, Personal, Stocks/Shares, Spouse has stock ownership: Agios; Financial Interests, Institutional, Invited Speaker: Novartis, Bristol Myers Squibb, Pfizer, Boerhinger-Ingelheim, GlaxoSmithKline, Roche/Genentech, AstraZeneca, Merck, Astellas, Bayer, Amgen, Symphogen, Intensity Therapeutics, Shattucks, EMD Serono; Non-Financial Interests, Personal, Advisory Role: ICR. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

10P - Deep Learning-based prediction of patientÕs TLS status from HE images in pan-cancer cohort (ID 111)

Presentation Number
10P
Lecture Time
12:30 - 12:30
Speakers
  • Lucile Vanhersecke (Bordeaux, France)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Tertiary lymphoid structures (TLS) are ectopic lymphoid formations which are composed predominantly of B cells, T cells and dendritic cells. The presence in the tumor compartment of mature TLS - characterized by mature follicles containing germinal centers - has been strongly associated with improved survival upon cancer immunotherapies for patients with solid tumors. For this reason, TLS could be used as a predictive factor biomarker to identify the patients more likely to benefit from immune checkpoint inhibitors. However, the pathological assessment of the TLS status remains time-consuming and usually requires additional analysis including CD3/CD20 staining. This study aims to show that artificial intelligence techniques applied to digital pathology could offer a fast and cheap mass patient screening solution to assess TLS from Hematoxylin/Eosin (HE) digital pathology slides used in clinical workflow.

Methods

We analyzed a pan-cancer cohort of 289 HE-stained Whole Slide Images (WSI) from Institut Bergonié (43% NSCLC, 12% sarcoma, 45% other solid tumors) on which TLS were manually contoured by expert pathologists. A Deep Learning (DL) model was trained on WSI to predict TLS status at the patient level (presence or absence of TLS). Models were evaluated using five-fold cross validation. The best performing architecture included two main components: a predicted score of TLS presence on small areas (112x112 micrometers) of the WSI followed by an aggregation at the patient level. To assess the transferability of the model, a validation cohort (PEMBROSARC) of 236 sarcoma WSI was used.

Results

The best performing model achieved a ROC AUC score of 0.917 (std of 0.036) in five-fold cross validation on the pan-cancer cohort (specificity of 68% for a sensitivity of 90%). This model transferred very well on the PEMBROSARC study, the first clinical trial implementing TLS status as an inclusion criteria (Italiano et al Nature Medicine 2022) with a ROC AUC score of 0.89 (specificity of 64% for a sensitivity of 90%).

Conclusions

Our study demonstrates the predictive power of DL applied to predict the patient's TLS status from HE images. This model could be implemented in pathology labs as an efficient pre-screening tool.

Legal entity responsible for the study

Institut Bergonie.

Funding

Owkin, Inc.

Disclosure

L. Gillet, J. Le Douget, B. Schmauch, C. Maussion: Financial Interests, Institutional, Full or part-time Employment: Owkin, Inc. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

11P - Association of VISTA expressing CD66b positive neutrophils, with response and survival benefit from pembrolizumab in advanced malignant pleural mesothelioma (ID 112)

Presentation Number
11P
Lecture Time
12:30 - 12:30
Speakers
  • Krisztian Homicsko (Lausanne, Switzerland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

V-domain Ig suppressor of T cell activation, VISTA, is a type I transmembrane protein that functions as an immune checkpoint. VISTA can be expressed by a wide range of cell types. Malignant pleural mesothelioma (MPM) is a tumor type with the highest levels of VISTA expression. However, it remains unclear if VISTA could be a predictive or prognostic marker in MPM following PD-1 blockade.

Methods

The phase III PROMISE MESO study randomized 144 patients to either chemotherapy (vinorelbine or gemcitabine) or pembrolizumab. We analyzed 62 patients from the pembrolizumab arm and 57 patients from the chemotherapy arm with available tumour tissue. We performed multiplex IHC for the following markers/cell types: calretinin of WT1, CD8, CD14, CD66b, CD11c, VISTA, and DAPI.

Results

Most of the VISTA expression originated from the mesothelioma cells. Bioinformatic analysis of MPM RNAseq data showed enrichment for response to INFa and INFg with VISTA overexpression. Responses to pembro or chemo were not influenced by global VISTA expression. However, VISTA expression on CD66b-positive neutrophils negatively correlated with responses. The VISTA+/CD66b+ median value was 0.56 (IQR: 0.0-3.5), and the rounded median value of 1.0/mm2 was used as threshold. Patients with VISTA+/CD66b+ high had an ORR of 5.6% whereas VISTA+/CD66b+ low tumors had 36.8%. Neutrophil infiltration alone did not correlate with responses. The difference in ORR also translated to prolonged PFS in patients with low VISTA+/CD66b+ cell counts (interaction p=0.0051), but no difference in overall survival was detected (interaction p=0.35). Neutrophils were not predictive of PFS or OS. In public single-cell expression data of lung cancer patients, VISTA expression indeed was higher in a subset of neutrophils, specifically in tN1 and tN3 subtypes.

Conclusions

Overall, our data show that VISTA expressing neutrophils correlate with outcomes. Our data underscore the complex biology of VISTA and suggest that specific targeting of VISTA on neutrophils might be a viable therapeutic option in a subset of MPM patients.

Clinical trial identification

NCT02991482. ETOP 9-15 PROMISE-meso trial.

Legal entity responsible for the study

ETOP IBCSG Partners Foundation.

Funding

ETOP IBCSG Partners Foundation, and by Merck Sharpe & Dohme.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

12P - Multi-omics correlation with clinical outcome in metastatic melanoma patients treated with ipilimumab plus guadecitabine: the NIBIT-M4 study (ID 113)

Presentation Number
12P
Lecture Time
12:30 - 12:30
Speakers
  • Anna M. Di Giacomo (Siena, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

DNA hypomethylating agents (DHA) combined with immune checkpoint inhibitors (ICI) are a promising strategy to improve the immunomodulatory and antitumor activity of ICI-based treatment. Providing initial support to this notion, guadecitabine plus ipilimumab in the phase Ib NIBIT-M4 trial, proved to be feasible, safe, and tolerable, with clinical and biological activity in metastatic melanoma patients (pts) (Di Giacomo AM, CCR 2019).

Methods

Pts enrolled in the NIBIT-M4 trial were assessed for long-term clinical outcomes. Molecular profiles of serial tumor biopsies performed at week 0, 4, and 12, including RNA, Whole Exome and Genome-wide methylation sequencing, were analyzed in an integrative manner and correlated with clinical data; pts were classified as Responder (R) or Non-responder (NR) based on disease control.

Results

Among the 19 pts enrolled (minimum follow-up 45 months), the 5-y OS rate was 29%; median duration of response was 20.6 months (95% CI,12.4-28.8). Analysis of longitudinal biopsies from 14 pts showed that primary/acquired resistance is underpinned by multi-level molecular mechanisms. Indeed, NR pts had higher mutation frequency in NRAS and harbored somatic alterations in genes involved in the Epithelial to Mesenchymal Transition (EMT) and chromatin organization pathways compared to R pts. Moreover, NR pts had a heterogeneous expression phenotype, characterized by activated patterns of proliferation/differentiation/EMT and by deactivation of INF-ɣ/HLA-II/immune-related genes. Conversely, R pts displayed dynamic patterns of activated immune signatures and favorable tumor-infiltrating microenvironment with T/NK cell clonal activation progressively increasing with therapy. We also developed a DNA-RNA immune stratification, simultaneously taking into account both the composition of the tumor microenvironment and the presence of reactive T-cells measured as the ratio between observed and predicted tumor neoantigens.

Conclusions

Our comprehensive longitudinal multi-omics analyses of tumors from the NIBIT-M4 study identify a landscape of biomarkers predictive of response that may guide patient stratification and selection in ICI plus DHA therapies.

Clinical trial identification

EudraCT 2015-001329-17, NCT02608437.

Legal entity responsible for the study

NIBIT Foundation Onlus.

Funding

Fondazione AIRC under 5 per Mille 2018 – ID 21073 program (principal investigator M. Maio).

Disclosure

A.M. Di Giacomo: Financial Interests, Personal, Advisory Board: Bristol Myers Squibb, MSD, Pierre Fabre, Novartis; Financial Interests, Personal, Invited Speaker: Sanofi. M. Maio: Financial Interests, Personal, Advisory Board: MSD, BMS, AstraZeneca, Incyte, Amgen, Pierre Fabre; Financial Interests, Personal, Invited Speaker: Roche, Eli Lilly; Financial Interests, Personal, Ownership Interest: Epigen Therapeutics, Theravance. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

13P - Tumor infiltrating CD8/CD103/TIM-3-expressing lymphocytes in Epithelial Ovarian Cancer co-express CXCL13 and associate with improved survival (ID 114)

Presentation Number
13P
Lecture Time
12:30 - 12:30
Speakers
  • Vrouyr Bilemjian (Groningen, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Reactivation of tumor-infiltrating T lymphocytes (TILs) with immune checkpoint inhibitors or co-stimulators has proven to be an effective anti-cancer strategy for a broad range of malignancies. However, epithelial ovarian cancer (EOC) remains largely refractory to current T cell-targeting immunotherapeutics. Therefore, identification of novel immune checkpoint targets and biomarkers with prognostic value for EOC is warranted. We here identified a TIM-3/CXCL13-positive tissue-resident memory (CD8/CD103-positive) T cell (Trm) population in EOC. Analysis of a cohort of ∼175 patients with high-grade serous EOC revealed TIM-3-positive Trm were significantly associated with improved patient survival.

Methods

- Single cell mRNA sequencing data analysis. - Immunohistochemical staining, image acquisition and analysis. Tumor Infiltrated Lymphocyte flow cytometric analysis.

Results

A cohort of EOC core samples was evaluated for the presence of tumor infiltrating CD8/CD103/TIM-3triple-positive T cells and subsequently correlated with patient survival. Interestingly, increased tumor infiltration of CD8/CD103/TIM-3triple-positive cells associated with improved patient survival in EOC, suggesting that CD8/CD103/TIM-3triple-positive TILs can serve as a prognostic marker for EOC. In line with this finding, a single-cell tumor immune transcriptomic dataset revealed upregulation of TIM-3, CXCL13 and CD103 within the terminally exhausted CD8-positive T cell fraction. Confirmatory flowcytometric evaluation of CXCL13 expression on isolated EOC TILs revealed that CXCL13 was predominantly found within the CD8/CD103/TIM-3triple-positive fraction compared to it’s single- and double-positive counterparts.

Conclusions

TIM-3 itself or as surrogate marker for prognostically favorable CXCL13-positive CD8-positive TILs may have prognostic value. As CXCL13-positive CD8-positive T cells have been strongly linked to patient response to anti-PD1 immune checkpoint blockade, combinatorial TIM-3 and PD-1 blockade therapy may be of interest for the (re)activation of anti-cancer immunity in EOC.

Legal entity responsible for the study

The author.

Funding

European Union H2020 Programme.

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster Display Poster Display session

14P - Deep learning-based quantification of immune infiltrate for predicting response to pembrolizumab from pre-treatment biopsies of metastatic non-small cell lung cancer: a study on the PEMBRO-RT Phase 2 trial (ID 115)

Presentation Number
14P
Lecture Time
12:30 - 12:30
Speakers
  • Joey Spronck (Nijmegen, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immunotherapy has become the standard of care for metastatic non-small cell lung cancer (mNSCLC) without a targetable driver alteration, yet we still lack insight into which patients (pts) will benefit from such treatments. To that end, we investigated characteristics of the immune infiltrate in the tumor microenvironment in relation to immunotherapy response. We report the results of an automated deep learning approach applied to digital H&E whole slide images (WSIs) of pre-treatment biopsies from the PEMBRO-RT clinical trial.

Methods

61 quality-checked H&E WSIs were processed with 3 deep learning algorithms. We extracted a tissue mask using an existing method (Bándi et al., 2019), and detected tumor and immune cells using HoVerNet (Graham et al., 2019). Tumor clusters were identified by combining the output of HoVerNet and tumor segmentation from an nnUnet (Isensee et al., 2021) model that we trained on external NSCLC images. From the output of this pipeline, we extracted immune infiltrate-based density metrics, calculated over all tissue (allINF), stroma within 500um from the tumor border (sINF), tumor region (tINF), and the combination of stroma and tumor (t+sINF). All metrics were used in ROC analysis after dichotomizing pts as responders and non-responders (response was defined as complete or partial response at any time point or stable disease for ≥12 weeks according to RECIST 1.1 measurement). Differences in metric distributions between the two groups were tested with a two-sided Welch t-test. Kaplan-Meier (KM) analysis was performed on progression-free survival (5-year follow-up).

Results

Our automated analysis reported denser immune infiltrates in responders, although not statistically significant (0.05<p≤0.2). All immune infiltrate metrics showed some predictive value with AUCs > 0.63, where tINF reported an AUC of 0.70. KM analysis showed p=0.07 if pts were stratified based on the median tINF, and p=0.02 if stratified based on the optimal operating point of its ROC curve.

Conclusions

Deep learning models that analyze the immune infiltrate density on H&E WSIs can identify mNSCLC responders to pembrolizumab.

Clinical trial identification

NCT02492568.

Legal entity responsible for the study

Netherlands Cancer Institute.

Funding

Dutch Research Council (NWO).

Disclosure

M. van den Heuvel: Research funding paid to their institution by AstraZeneca, Bristol Myers Squibb, Janssen, Merck, Novartis, Stichting Treatmeds, Merck Sharp & Dohme, Pamgene, Pfizer, Roche and Roche Diagnostics; Advisory Board fees paid to their institution by Bristol Myers Squibb, Pfizer, Merck, Merck Sharp & Dohme, AstraZeneca, Novartis and Roche-Genentech, all in the 36 months prior to manuscript submission; Member of the Dutch Association of Physicians in Chest Medicine and Tuberculosis (NVALT) and the Dutch Society for Medical Oncology (NVMO). W. Theelen: Financial Interests, Institutional, Research Grant: AstraZeneca, MSD, Sanofi. F. Ciompi: Financial Interests, Personal, Stocks/Shares: Aiosyn BV; Financial Interests, Personal, Advisory Board: TRIBVN Healthcare. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

15P - Prediction for pCR after Neoadjuvant Immunotherapy Combined with Chemotherapy Using Single-Cell RNA Sequencing in Patients with Locally Advanced Esophageal Squamous Cell Carcinoma (ESCC) : A Single-Arm Phase II Clinical Trial (ID 116)

Presentation Number
15P
Lecture Time
12:30 - 12:30
Speakers
  • Guangyu Yao (Shanghai, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

There is a lack of effective predictor for pCR of neoadjuvant immunotherapy in esophageal squamous cell carcinoma (ESCC) patients. High-throughput single-cell RNA sequencing (scRNAseq) can define the tumor microenvironment (TME), tumor heterogeneity and immune microenvironment. In this study, we sought to explore the immune pathways related to treatment response of immunotherapy by scRNAseq.

Methods

This was a single-arm phase II clinical trial, planned to enroll 20 locally advanced ESCC patients (cT2-4N0-1M0). Patients received neoadjuvant chemotherapy (paclitaxel-albumin + carboplatin) combined with tislelizumab and followed by minimaly invasive esophagectomy (MIE). We conducted a scRNAseq using fresh tumor tissues taken by endoscopy before neoadjuvant therapy. Histological outcome was divided into pCR (no residual tumor cells in tumor and lymph nodes), MPR (<10% residual tumor – excludes pCR) and non-MPR (>10% residual tumor).

Results

17 patients were enrolled since March,2022. 7 of 17 patients finished neoadjuvant therapy and MIE surgery. Among these 7 patients, 3 achieved pCR, 1 got MPR, and 3 were non-MPR, with a total of 42808 single cells sequenced. Compared to non-MPR, there were significantly increase in T lymphocytes (CD8+, T helper cell, Regulatory T cell) and mature dendritic cells, and significantly decrease in B cells and neutrophils in pCR patients. Morever, pCR patients showed greatly increase on the expression of PD-L1(CD274) in mature dendritic cells.

Conclusions

This study identified novel immunological predictor in baseline tissue for ESCC neoadjuvant immunotherapy. The enrichment of T lymphocytes (CD8+, T helper cell, Regulatory T cell) and mature dendritic cells together with the increase expression of PD-L1(CD274) in mature dendritic cells suggest a promising role of predicting completed pathological response (pCR) induced by neoadjuvant chemotherapy and immunotherapy in locally advanced ESCC 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 Poster Display session

17P - Using ex vivo organ culture technology to evaluate a T-Cell Bispecific (TCB) CEACAM5 Therapy (ID 118)

Presentation Number
17P
Lecture Time
12:30 - 12:30
Speakers
  • Adi Zundelevich (Rehovot, Israel)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Accurately evaluating immuno-oncology therapeutics in pre-clinical models presents a significant technological challenge in translational oncology. Both human organoid models and humanized mouse models fail to recapitulate the complexity of the cancer tumor microenvironment found in the patient. Multiple stromal components such as immune cells, fibroblasts, blood vessels, and even bacteria have been shown to affect tumor response to treatment, suggesting the need for their inclusion and faithful reconstruction in pharmacological development.

Methods

To overcome these challenges, we have made use of a recently developed novel immune-competent ex vivo organ culture (EVOC) assay to evaluate the activity of CD3-based T-Cell Bispecific Antibodies (TCBs). In particular, we assessed the anti-tumor effects of a CEACAM5 TCB, which is designed to simultaneously bind CEA-expressing cancer cells and CD3-expressing T cells, activating the latter. Fresh tissues obtained during resection of NSCLC, CRC and PDAC tumors, were sliced and cultured ex vivo. The cancer tissue and microenvironment, including endothelial and immune cells, was preserved at high viability for 5 days. We then evaluated cytokine secretion in the assay after 24 and 48 hours.

Results

We found robust expression of T-Cell specific activation markers in response to treatment with CEACAM5 TCB antibodies. NSCLC and CRC tissues with abundant T-cells showed significant cytokine over-expression, while PDAC with low T-Cell presence showed none. Notably, comparing the histology of the fresh sample with the treated sample after 5 days, showed T-Cell infiltration from the tissue periphery into the cancer foci, demonstrating the efficacy of the bispecific antibody to recruit T-cells to tumor cells.

Conclusions

These results show the applicability of our ex vivo organ culture to evaluate immune activating compounds and suggests the ability to use EVOC, in the future, as a predictive biomarker for TCB therapy.

Legal entity responsible for the study

The authors.

Funding

Curesponse Ltd. & F. La-Hoffman Roche.

Disclosure

A. Zundelevich, B. Ibrahim, L. Turovsky, S. Aharon, V. Bar: Financial Interests, Personal, Member: Curesponse. S. Salpeter, G. Neev: Financial Interests, Personal, Leadership Role: Curesponse. R. Straussman: Financial Interests, Personal, Advisory Board: Curesponse. I. Silva, S. Li, S. Herter, M. Bacac, S. Sadok, N. Gjorevski: Financial Interests, Personal, Member: Roche.

Collapse
Poster Display Poster Display session

18P - Systematic evaluation of published predictive gene expression signatures in pan-cancer patient cohorts treated with immune checkpoint inhibitors in a real-world setting (ID 119)

Presentation Number
18P
Lecture Time
12:30 - 12:30
Speakers
  • Ahmad A. Tarhini (Tampa, PA, United States of America)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Previous studies developed predictive biomarkers related to immune checkpoint inihibitors (ICI) based on clinical trials cohorts. Here, we leverage the ORIEN data conducted under the Total Cancer Care protocol across 18 collaborating cancer centers to evaluate the predictive value of published gene expression signatures in real-world data (RWD) of patients with cancer treated with ICI.

Methods

Overall Survival (OS) was the primary endpoint. RNA-seq performed on tumors was aligned to human reference genome (GRCh38) following RSEM pipeline. Gene expressions were quantified as TPM and log2(TPM+1) transformed. 28 published predictive gene expression signatures were collected and evaluated in our cohort (Table). Mann-Whitney U-test was used to compute the difference between groups, and Kaplan-Meier survival analysis was performed. Test with p<0.05 was considered statistically significant.

Results

659 patients treated with ICI for kidney cancer (n = 151), lung cancer (n = 138), melanoma (n = 123), head and neck cancer (n = 121), sarcoma (n = 78) and bladder cancer (n = 48) were included. We defined “good” and “poor” outcomes if OS was >24 or <24 months, respectively. Twelve immune active gene signatures were associated with ICI responses in melanoma (p < 0.05); Table. An angiogenesis signature (p = 0.0281) and a tertiary lymphoid structure signature (p= 0.0133) were associated with ICI responses in kidney cancer and head and neck cancer, respectively. None of the 28 evaluated gene signatures were associated with ICI responses in lung cancer, bladder cancer or sarcoma.

Conclusions

We validated the predictive value of immune related gene signatures in melanoma, kidney, head and neck cancers utilizing RWD. Ongoing analyses are taking on a discovery approach for predictive genes and related pathways to better understand the underlying mechanisms related to tumor immunogenicity across the different tumor types.

Gene expression signatures and correlation with survival (>24 vs. <24 months)

Gene Signature Reference P-value Cancer
Tertiary Lymphoid Str. Cabrita 2020 0.0133 Head & neck
Angiogenesis Cristescu 2022 0.0281 Kidney
IFNg/Effector T cell Fehrenbacher 2016 0.001 Melanoma
Effector T cell Bolen 2011 0.0013
IFNg-6 Ayers 2017 0.0017
Immune Cytolytic Activity Rooney 2015 0.002
IFNg-18 Ayers 2017 0.0035
TIP Hot Wang 2021 0.004
Cytotoxic Immune Signature Davoli 2017 0.0047
Tertiary Lymphoid Str. Chaurio 2022 0.0061
Chemokine Coppola 2011, Mule 2012 0.0073
T & B cell interplay Tarhini 2017 0.0114
Roh Immune Score Roh 2017 0.0127
MHC-II Liu 2021 0.0180

Clinical trial identification

NCT03977402.

Legal entity responsible for the study

Oncology Research Information Exchange Network (ORIEN) and M2GEN.

Funding

Oncology Research Information Exchange Network (ORIEN), M2GEN, Community Foundation of Tampa Bay.

Disclosure

A.A. Tarhini: Financial Interests, Personal, Advisory Board: Bristol Myers Squib, Genentech/Roche, Easai, Instil Bio, Clinigen, Regeneron, Sanofi-Genzyme, Novartis, Partner Therapeutics, BioNTech, Merck; Financial Interests, Personal and Institutional, Invited Speaker: Bristol Myers Squibb; Financial Interests, Institutional, Invited Speaker: Genentech-Roche, Nektar, Checkmate, InflaRx; Financial Interests, Institutional, Research Grant: Regeneron, Sanofi-Genzyme, Clinigen, Acrotech, Pfizer, OncoSec. M.L. Churchman: Financial Interests, Institutional, Full or part-time Employment: M2GEN. W.S. Dalton: Financial Interests, Personal, Full or part-time Employment: M2GEN. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

19P - Effects of CTLA-4 Single Nucleotide Polymorphisms (SNPs) on toxicity of ipilimumab-containing regimens in patients with advanced stage melanoma (ID 120)

Presentation Number
19P
Lecture Time
12:30 - 12:30
Speakers
  • Karlijn D. Joode (Rotterdam, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

SNPs of the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) gene, an inhibitor of T cell priming, are associated with auto- and allo-immunity. Previously, studies implied a role for these SNPs as surrogate markers for outcome in melanoma patients treated with immune checkpoint inhibitors. However, no predictive SNPs are defined to date. The primary aim of this study was to analyze different CTLA-4 SNPs in a large real-world cohort of melanoma patients treated with ipilimumab and to correlate these SNPs with toxicity and survival.

Methods

Archival blood and/or tumor-tissue samples were collected from 317 advanced stage melanoma patients treated with ipilimumab (+/- nivolumab) in 5 Swiss and Dutch hospitals. DNA was extracted and used for MALDI-TOF MS based genotyping for 11 different SNPs. Associations between different allele genotypes and occurrence of grade ≥3 toxicity, treatment response and survival outcomes were tested using univariable logistic regressions or Cox proportional hazard models.

Results

DNA of 216/317 patients could be analyzed. The cohort was representative of a real-life population of metastatic melanoma patients receiving ipilimumab (+/- nivolumab): among the remaining 216/317 evaluable patients, 65.3% were male, median age at diagnosis was 59 years, 37% of patients had partial or complete response and 59% had ≥1 immune-related adverse events. A TT-genotype in the -1722 C/T SNP (rs733618, T=0.8345 (1000Genomes)), located in the promoter region of CTLA-4, was significantly associated with a decreased rate of ≥ grade 3 toxicity [odds ratio=0.40; CI95=0.16 - 1.00; p=0.049]. The TT-genotype in the Jo27 T/C SNP (rs11571297, T=0.5575 (1000Genomes)) [hazard ratio (HR)=0.54; CI95=0.28-1.02, p=0.056] and the GG-genotype in the Jo31 SNP (rs11571302, G=0.5713 (1000Genomes) [HR=0.54; CI95 = 0.29-0.99, p=0.046], both located at the 3’ untranslated region, were associated with increased overall survival.

Conclusions

CTLA-4 SNPs might be predictive of toxicity and/or survival in melanoma patients receiving ipilimumab. Confirmatory studies are needed to exploit findings of this exploratory study and to investigate the exact role of CTLA-4 SNPs as possible biomarkers.

Legal entity responsible for the study

The authors.

Funding

BMS.

Disclosure

K.D. Joode: Financial Interests, Personal, Other, Travel expenses congress: Ipsen. A.A.M. Van der Veldt: Financial Interests, Institutional, Advisory Board: BMS, MSD, Merck, Pfizer, Ipsen, Eisai, Pierre Fabre, Roche, Novartis, Sanofi. R.H. Mathijssen: Financial Interests, Personal, Invited Speaker: Novartis; Financial Interests, Personal, Advisory Role: Servier; Financial Interests, Personal, Licensing Fees, Patent pending: Pamgene; Financial Interests, Institutional, Research Grant, Investigator-initiated research: Astellas, Bayer, Boehringer Ingelheim, Cristal Therapeutics, Pamgene, Pfizer, Novartis, Roche, Servier. Y. Metaxas: Financial Interests, Institutional, Research Grant: Bristol Myers Squibb. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

20P - Human virome epitope-level antiviral antibody profiling identified the cytomegalovirus (CMV) as the main driver of Senescent Immune Phenotype (SIP) in patients with advanced non-small cell lung cancer patients (aNSCLC) (ID 121)

Presentation Number
20P
Lecture Time
12:30 - 12:30
Speakers
  • Marie Naigeon (Villejuif, Cedex, France)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immunosenescence is a progressive remodeling of immune functions with a multifactorial etiology including aging and chronic antigenic stressors (inflammation, infections, cancer). We showed that a high pretreatment SIP (CD28CD57+KLRG1+CD8+ circulating T cells>39.5%, SIP+) was associated with resistance to ICB in patients with aNSCLC. Chronic viral infections may speed up immune aging, especially CMV which affects blood T cells phenotype (loss of CD28, overexpression of CD57). We aimed to assess antiviral serological profile and its association with SIP status.

Methods

Baseline SIP status was assessed by flow cytometry on fresh blood samples from ICB-treated and polychemotherapy-treated (PCT) aNSCLC patients. Sera from patients were analysed with VirScan (CDI Labs, USA), a high-throughput antiviral antibody (Ab) method (VirScan™) enabling simultaneous epitope-level antiviral antibody profiling of most viruses with human tropism (206 species, 1000 strains) via phage-display and immunoprecipitation sequencing (PhIP-Seq).

Results

132 aNSCLC patients were evaluable for SIP and VirScan™ assay. The antiviral serological profile seems similar between SIP+ and SIP- patients, except for CMV where the mean enrichment of anti-CMV antibodies was higher in SIP+. Of the 74 antiviral Ab associated with a higher SIP, 70 (94.6%) recognized CMV-peptides and CMV was the only virus globally associated with a higher SIP. SIP+ patients were predominantly CMV+ compared to SIP- (93% vs 57%, p<0.001), but 70.5% of CMV+ remained SIP-. In CMV+ patients, no difference was observed in the number of CMV epitodes targeted by antibodies (p=0.62) nor in protein immunisation profile between SIP+ and SIP- patients. Among all clinic-biological parameters studied only median age was higher in SIP+ (71 years vs 63 years, p=0.01) compared to SIP- in CMV+ patients.

Conclusions

Among 206 species, only anti-CMV Ab were associated with SIP+ status, which is associated with an older age in CMV+ patients. Further work investigating the predictive value of CMV in the response to ICB in older patients is ongoing.

Clinical trial identification

NCT03984318, NCT02105168.

Legal entity responsible for the study

Gustave Roussy.

Funding

ANR-21-RHUS-0013, Malakoff Médéric.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

21P - Peripheral pre-existing T-cell immunity as predictive biomarker in cancer immunotherapy for NSCLC patients (ID 122)

Presentation Number
21P
Lecture Time
12:30 - 12:30
Speakers
  • Anastasia Xagara (Larissa, Greece)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Pre-existing immunity that describes the endogenous tumor-specific adaptive immunity, before treatment may represent a valuable novel predictive biomarker for ICI treatment. In this study we estimate the potential value of pre-existing cancer-antigen specific CD8+ T cells as circulating predictive biomarkers. Additionally, we evaluate the major differences of known immune cell phenotypes between Pre-existing positive (PreI+) and Pre-existing negative (PreI-) NSCLC patients in circulation.

Methods

Blood was collected before initiation of immunotherapy from 24 NSCLC patients stage IIIb, PD-L1+ that receiving Durvalumab. PBMCs were isolated with Ficoll density gradient centrifugation including 15 healthy donors (HD). PreI was calculated by detecting endogenous IFNg expressing cells after in-vivo co-cultures of PBMCs with hTERT, MAGEA1, NY-ESO-1 and Survivin antigens. Immunophenotyping was performed by multi-color flow cytometry using antibodies against CD3, CD4, CD8, CD45RA, CD45RO, CCR7, PD-1, PD-L1 for CD4 and CD8 T-cells and CD3, CD4, FoxP3, CD25, CD127, CTLA-4, CD39 for Tregs.

Results

From 24 patients receiving Durvalumab 10/24 (41,6%) had peptide specific T-cells (PreI + patients). 60% had detectable peptide specific T cells for all (4/4) antigens tested. Survival analysis revealed better PFS in PreI+ than PreI patients (Log-rank = 0.06, median 333,5 and 185 days ) and better OS (*Log-rank = 0.032, median undefined and 254 days). Patients with PreI+ and low CD25+CD127-CTLA-4+ Tregs (n=8) had better OS (*Log-rank 0,036, median undefined and 251 days) than those with PreI- and high Tregs (n=9). The levels of CD3CD4PD-1 were higher in NSCLC patients compairing to HD (*0,019) but there was not a difference between PreI+ and PreI- patients. PreI+ patients had significantly higher numbers of CD3CD8PD-1 cells comparing to PreI- (*p= 0,024) but not CD3CD8PD-L1. Prei+ patients had high numbers of Teff (CD3+CD8+CD45RA+CD45RO-CCR7-) compairing with both HD (**0.0039) and PreI- (*0.032). Both T cm and Tem do not differ between all tests.

Conclusions

Pre-existing tumor antigen specific T-cells in circulation before initiation of immune checkpoint inhibitors in NSCLC patients serve as a good prognostic factor of response.

Legal entity responsible for the study

The authors.

Funding

This research has been co-financed by the European Union and Greek national funds through the Operational Program Competitiveness, Entrepreneurship and Innovation, under the call Research-Create-Innovate (project code: T2EΔK-02218).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

22P - Mean platelet volume to lymphocyte ratio: a new biomarker predictive of response in patients treated with immunotherapy (ID 123)

Presentation Number
22P
Lecture Time
12:30 - 12:30
Speakers
  • Hasan C. Yildirim (Ankara, Turkey)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Although the use of immunocheckpoint inhibitors (ICIs) in cancer treatment has become widespread, the factors predicting the response to these treatments have not yet entered our clinical practice. Considering that the progression under ICIs is especially in the first 3-6 months, there is a need for biomarkers to help us determine which patients do not benefit from these treatments. It is important to detect an easy and inexpensive biomarker in order to be widespread in our clinical practice. In our study, we aimed to examine the relationship between mean platelet volume/lymphocyte ratio, which can be evaluated by hemogram test, and overall survival in cancer patients treated with nivolumab.

Methods

A total of 131 adult metastatic cancer patients treated with nivolumab for malignant melanoma (MM), renal cell carcinom (RCC), non-small cell lung cancer (NSCLC) and head and neck (HNC) cancer patients were included. Baseline demographics, ECOG performance status, type of tumors, and baseline blood count parameters were recorded. Possible predisposing factors were evaluated with univariate and multivariate analyses.

Results

The median age was 59.87 ± 11.97 years, and the median follow-up was 20.20 (IQR 12.80-27.60) months. Renal cell carcinoma (43.5%) and melanoma (25.9%) were most common diagnoses. Patients with ECOG 0-1 (mOS: 20.60 months (95% CI: 14.94-25.29) vs 5.24 months (95% CI: 0-16.42), p<0.001), LDH levels within the normal range (mOS: 24.54 months (95% CI: 14.13-34.96) vs 13.10 months (95% CI: 4.49-21.72), p=0.038) and low MPVLR (mOS: 33.70 months (95% CI: 25.99-41.42) vs 11.07 months (95% CI: 6.89-15.24), p<0.001) had a longer median overall survival. In multivariate Cox regression analysis, high MPV/Lymphocyte ratio, ECOG score 2-3 and high LDH level were associated with shorter overall survival time (p<0.001, p=0.001 and p=0.046 respectively).

Conclusions

In this study, we determined that MPVLR could be a new biomarker predicting response to nivolumab treatment.

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 Poster Display session

23P - Enriching for response: patient selection criteria for A2AR inhibition by EXS-21546 through ex vivo modelling in primary patient material (ID 124)

Presentation Number
23P
Lecture Time
12:30 - 12:30
Speakers
  • Gregory Vladimer (Vienna, Austria)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Novel immunotherapies targeting the adenosine pathway are in clinical trials, however, only modest monotherapy activity has been observed in non-prioritised patient groups. To optimise the chance of success with our A2AR-selective antagonist, EXS-21546 (‘546; NCT04727138, discovered in collab. with Evotec), we work to identify an adenosine-induced immunosuppression biomarker signature for clinical trial patient selection. Here we present initial transcriptional and functional data mapping the adenosine suppressed immune potential at the single cell level, and subsequent modulation through antagonism of A2AR with ‘546, along with first patient-selection modelling to prioritise patients for ‘546 therapy.

Methods

By leveraging disease-relevant primary human tissues, we model the patient specific anticancer immune potential, and begin to validate patient selection methods functionally with a translatable high content imaging platform amenable to primary human material. This platform is supported by end-to-end deep learning-driven image analysis; work is combined with orthogonal multi-omic characterisation of single cell effects induced by ‘546.

Results

We present preclinical mechanistic studies of A2AR antagonism on tumour infiltrating immune cells, leading to a foundational predictive adenosine suppression signature. The patient selection gene signature is correlated to functional profiling using a high content imaging platform with proven translational capabilities (Kornauth et al, Cancer Disc, 2022), demonstrating association of immune activity with antagonism of adenosine signalling by ‘546. Signatures and patient selection algorithms are cross-validated with publicly available data.

Conclusions

Combining deep learning of single cell functional and multi-omics profiling data of disease relevant primary model systems, we model the association of the immune response potential to A2AR antagonism in cancer to define a biomarker signature with the potential to identify patients likely to benefit from A2AR antagonism. This could be implemented in future studies of our clinical candidate ‘546 to deliver the right drug at the right time to the right patients.

Legal entity responsible for the study

Exscientia.

Funding

Exscientia.

Disclosure

G. Vladimer, I. Alt, R. Sehlke, A. Lobley, C. Baumgärtler, M. Stulic: Financial Interests, Personal, Full or part-time Employment: Exscientia; Financial Interests, Personal, Stocks/Shares: Exsciencia. K. Hackner, L. Dzurillova, E. Petru, L. Hadjari, J. Lafleur, J. Singer: Non-Financial Interests, Institutional, Other: Exscientia. N. Krall: Financial Interests, Personal, Full or part-time Employment: Exscientia; Financial Interests, Personal, Stocks/Shares: Exsciencia. J. Šufliarsky, L. Hefler, T. Fuereder: Non-Financial Interests, Institutional, Other: Exscientia. C. Taubert: Financial Interests, Personal, Stocks/Shares: Exscientia; Financial Interests, Personal, Full or part-time Employment: Exscientia. C. Boudesco, A. Payne: Financial Interests, Personal, Full or part-time Employment: Exscientia; Financial Interests, Personal, Stocks/Shares: Exscientia.

Collapse
Poster Display Poster Display session

24P - The predictive and prognostic role of single nucleotide gene variants in PD-1 and PD-L1 in patients with advanced melanoma treated with PD-1 inhibitors (ID 125)

Presentation Number
24P
Lecture Time
12:30 - 12:30
Speakers
  • Andrea Boutros (Genova, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The introduction of immune-checkpoint inhibitors (ICIs) has revolutionized the treatment paradigm for advanced melanoma, leading to substantial benefit in overall survival (OS). However, several patients still do not benefit from ICIs and to date there are no predictive biomarkers for PD-1 inhibitors. PD-1 axis single nucleotide variants (SNVs) may affect receptor/ligand interactions in the tumor microenvironment and consequently the immune response and efficacy of ICIs. Based on this rationale, their predictive and prognostic role was investigated.

Methods

We analyzed, in metastatic melanoma patients treated with nivolumab or pembrolizumab, five PD-1 SNVs, namely PD1.3G>A (rs11568821), PD1.5 C>T (rs2227981), PD1.6 G>A (rs10204525), PD1.7 T>C(rs7421861), PD1.10 C>G (rs5582977) and three PD-L1 SNVs:+8293 C>A (rs2890658), PD-L1 C>T (rs2297136) and PD-L1 G>C (rs4143815) by pyrosequencing methods. Association of SNV genotypic frequencies with best overall response (BOR) to PD-1 inhibitors and development of immune-related adverse events (irAEs) were estimated through a modified Poisson regression. A Cox regression modelling approach was applied to evaluate the SNV association with OS. All regression analyses were adjusted for individual confounding factors.

Results

A total of 125 patients with advanced melanoma were included in the analysis. In patients carrying the PD-L1 C>T variant, a trend towards a lower relative risk (RR) of having progressive disease was observed in the C/T genotype (RR=0.60; 95%CI 0.25-1.41). In addition, a trend for a reduction in irAEs occurrence was observed in patients with PD1.7 C/C and PD-L1 +8293 C/A genotypes (RR = 0.35 [95%CI 0.09-1.31] and RR=0.45 [95%CI 0.22-0.93], respectively). Finally, a trend for a survival benefit was observed in PD1.7 C/C (HR=0.41 [95%CI 0.16-1.00]) and PD-L1 C/T (HR=0.53 [95%CI 0.24-1.18]) SNV genotypes.

Conclusions

Our study showed that SNV of PD-1 and PD-L1 may play a role as a predictive biomarker of response and development of irAEs to PD-1 inhibitor therapy. Some of the investigated SNVs were also associated with a reduction of the risk of death, although a larger group of patients is needed to confirm these results.

Legal entity responsible for the study

The authors.

Funding

Italian Ministry of Health RF-2016-02362288 and Ricerca Corrente 2019-2021.

Disclosure

F. Spagnolo: Financial Interests, Personal, Invited Speaker: Sanofi Genzyme, Roche, BMS, Novartis, Merck, Sunpharma, MSD, Pierre Fabre; Financial Interests, Personal, Advisory Board: Novartis, Philogen, Sunpharma, MSD. P. Queirolo: Financial Interests, Personal, Advisory Board: Roche/Genentech, Novartis, MSD, BMS, Pierre Fabre, Sanofi, Sun Pharma, Merck Serono; Financial Interests, Personal, Sponsor/Funding: MSD Oncology, Sanofi/Regeneron. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

25P - Platinum-based chemotherapy (PCT) addition to 1st-line PD-1/PD-L1 inhibitors (ICI) prevent hyperprogressive disease (HPD) in non-small cell lung cancer (NSCLC) patients (pts) by reducing circulating immature neutrophils (ID 126)

Presentation Number
25P
Lecture Time
12:30 - 12:30
Speakers
  • Roberto Ferrara (Milan, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

HPD has been described in ≃14-25% of pretreated NSCLC pts upon single-agent (SA) ICI and has not been reported upon PCT-ICI. So far, no predictive biomarkers are available for HPD early detection.

Methods

NSCLC pts treated with 1st line SA-ICI or PCT-ICI were assessed for HPD and circulating neutrophils. HPD was defined as delta tumor growth rate (TGR) >50% and/or TGR ratio ≥2. Circulating low density neutrophils (LDNs) were assessed by flow cytometry on peripheral blood mononuclear cells (PMBCs). LDNs were defined as CD66b+CD15+ cells among CD11b+ PBMCs and immature subtypes as CD10- LDNs. The LDNs predictive role was assessed by penalized model-based tests. LDNs’ survival and sensitivity to cisplatin induced cell death were tested in-vitro.

Results

144 NSCLC pts were included: 75 treated with SA-ICI, 69 with PCT-ICI. In the SA-ICI cohort, HPD occurred in 8 (11%) pts. Baseline immature circulating CD10- LDNs were significantly higher (p <0.01) in HPD [median (ME): 39.3, interquartile range (IQR): 28.7] vs pts with progression [ME: 7.4, IQR: 14.9] or response/stable disease [ME: 3.7, IQR: 12.6]. CD10- LDNs were associated with HPD [odds ratio (OR): 1.17, 95% CI: 1.06; 1.29], with a good prediction capability [cross-validated AUC 0.97 (95%CI: 0.94;1.00)]. A 30.5% cut-off value was identified by Youden index to discriminate HPD from others. In the PCT-ICI cohort, 14 pts had CD10- LDNs ≥30.5% being at high HPD risk. However, no HPD was observed in PCT-ICI cohort and dynamic LDNs evaluation in high HPD risk pts showed a 52.3% median reduction in CD10- LDNs upon PCT-ICI, vs only an 8.9% reduction in HPD pts upon SA-ICI, suggesting that PCT prevents HPD by reducing immature LDNs. In vitro experiments showed that immature CD10- LDNs had prolonged survival compared to mature CD10+ LDNs (alive cells after 5-days: 18.7% vs 0%), however, they were more sensitive to cisplatin induced necrotic cell death, confirming the role of PCT in killing preferentially immature LDNs.

Conclusions

Baseline immature CD10- LDNs is a circulating easy to measure biomarker to early detect HPD upon 1st line SA-ICI and could select NSCLC pts to be addressed to PCT-ICI.

Legal entity responsible for the study

Istituto Nazionale dei Tumori di Milano.

Funding

International Association for the Study of Lung Cancer (IASLC).

Disclosure

R. Ferrara: Financial Interests, Personal, Advisory Board: MSD, Beigene. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

26P - Impact of SF3B1 mutation on response to immunotherapy (ID 127)

Presentation Number
26P
Lecture Time
12:30 - 12:30
Speakers
  • Byungho Lim (Daejeon, Korea, Republic of)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Tumor-derived genetic alterations can regulate tumor microenvironment (TME) and host immune responses via activation of oncogenic pathways. Therefore, detection of somatic mutations associated with TME and tumor immunity may be useful in identifying patients who could benefit from immunotherapy. Immune checkpoint inhibitors (ICIs), a type of immunotherapy, are considered to be the emerging standard of care for various cancers. However, biomarkers that can help in predicting response to ICIs remain unknown.

Methods

To identify genetic markers associated with sensitivity to ICIs, we established a transcriptome-based predictive model followed by integrative genomics analyses. The impact of in silico derived biomarkers on the TME and response to ICIs was then assessed using in vitro assays and an in vivo mouse model study.

Results

We generated a transcriptome signature associated with response to ICIs based on a small but well-defined immunotherapy study (GSE78220) and applied the signature to large melanoma cohort data from The Cancer Genome Atlas (TCGA) by estimating transcriptome similarity. Based on the analysis, TCGA melanoma cohort was classified into potential responders and non-responders, and subsequent genomic interrogation revealed that the distribution of splicing factor SF3B1 mutations was significantly higher in potential responders. Furthermore, comparative analyses based on patients genotypes and loss- and gain-of-function studies based on manipulation of SF3B1 mutations revealed that SF3B1 mutations promoted transcriptional reprogramming in response to ICIs. To substantiate these results in vivo, we established a syngeneic mouse model, wherein immune resistant B16F10 melanoma cells were implanted into mice. Strikingly, in comparison to mice bearing wild-type SF3B1 tumors, mice bearing mutant SF3B1 tumors showed improved TME and immune profiles suitable for immunotherapy, which were characterized by accumulation of CD4 and CD8 T cells, activated dendritic cells, and concomitant induction of immune effectors.

Conclusions

Our findings suggest that SF3B1 mutations serve as biomarkers that can used to preemptively predict response to ICIs and ensure rational use of immunotherapy in cancer patients.

Legal entity responsible for the study

Korea Research Institute of Chemical Technology (KRICT).

Funding

This work was supported by the Korea Research Institute of Chemical Technology grant [KK2231-20]; and the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) [2022R1C1C100616211].

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

27P - Gene ontology enrichment analysis of peripheral CD3+CD4+T-cells predicts the immune-mediated behavior of melanoma (ID 128)

Presentation Number
27P
Lecture Time
12:30 - 12:30
Speakers
  • Dimitrios Ziogas (Athens, Greece)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Melanoma, from its early stages, has a natural propensity to spread, that is regulated via constant immune counteraction. Unleashing adaptive immunity via ICIs has improved melanoma prognosis, but not all patients behave similarly under immunotherapy. Some relapse in the first few months of immunotherapy, while others experience durable disease control, even after discontinuation of ICIs.

Methods

In order to identify any cellular/molecular signature in the peripheral blood that could predict primary immune escape, we examined 100 consecutive patients with stage III or IV melanoma who were scheduled to start ICIs. A multiparameter flow cytometry was used to detect differences in immune subpopulations, gating them by standard conjugated antibodies and mRNA was extracted after sorting of CD3+CD4+T-cells. A gene ontology (GO) enrichment analysis was performed to recognize which specific genes and key pathways are upregulated in stage III and IV melanoma and between long responders (>36 months) and early progressors(<6 months) to ICIs.

Results

Although circulating immune cells did not differ numerically in patients with stage III and IV melanoma as well as in responders and non-responders to ICIs, transcriptomic analysis of CD3+CD4+T-cells from 24 selected patients showed that 189 genes were upregulated in stage IV vs. 92 in stage III; while 101 genes were upregulated in early progressors vs. 47 in long responders. These differentially expressed genes (e.g., HLA-DRB5, BCL3, TRIM37, NCAM1, FGFR1) were functionally implicated in distinct cellular pathways that were particularly activated in these two settings. In fact, biological GO pathways of adaptive immunity, negative regulation of DNA-binding transcription factor activity, and lipid metabolism were significantly more upregulated in stage IV compared to stage III melanoma; while phosphorylation of CD3 and TCR chains, PD-1 and IFN signaling were more activated in early progressors compared to long responders to ICIs.

Conclusions

Extrapolating the rules of intra-tumoral plasticity to the blood, specific epigenetically altered pathways in the peripheral CD3+CD4+T-cells may differentiate the melanoma evolution from stage III to IV and drive the resistance to ICIs.

Legal entity responsible for the study

Internal Review Board of Laikon General Hospital.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

28P - Epithelial Mesenchymal transition confers resistance to dual PDL1-VEGF inhibition in relapsed mesothelioma (ID 129)

Presentation Number
28P
Lecture Time
12:30 - 12:30
Speakers
  • Essa Y. Baitei (Leicester, United Kingdom)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immune checkpoint blockade (ICB) improves survival outcomes for patients with mesothelioma, however the mechanisms underpinning response remain elusive with only a minority of patients exhibiting response to ICB. Better understanding could facilitate therapeutic advances in both patient stratification and the rational treatment of relapse. The goal of this study was to gain greater insight into the cellular and molecular regulation of response to dual PDL1-VEGF inhibition (atezolizumab/bevacizumab) in patients with relapsed mesothelioma treated in the MiST4 single arm phase II trial (NCT03654833).

Methods

Diagnostic FFPE tumour blocks acquired from the MIST4 cohort, were subjected to RNA extraction and RNA sequencing(n=18) STAR and featureCounts were used for alignment and read counts Differential gene expression (DEseq2), gene set enrichment analysis (broadinstitute.org/gsea, Clusterprofile) were conducted pathway signatures scores for individual patient were calculated by single-sample GSEA. Immune microenvironment cells type deconvolution were deconvoluted using CIBERSORTx (https://cibersortx.stanford.edu/) and was compared with multiplex immunofluorescence as ground truth. Response was dichotomised into those patients exhibiting tumour growth (G) or reduction (R) measured by mRECIST1.1. Random forest machine learning, and non-parametric statistical testing was used to identify enriched transcriptional patterns in R (n=9) vs G (n=9) groups.

Results

The G group exhibited significant enrichment of epithelial-mesenchymal transition (EMT) as estimated by GSEA compared with R. Conversely, inflammatory signatures were highly enriched in the R group versus G. Immune cell deconvolution revealed that NK cells are more abundant in R group (p value = 0.03). Chemokine analysis revealed a significant increase in CXCL12 (p = 0.039) and CXCL17 (p = 0.013) in R group.

Conclusions

EMT and inflammatory transcriptional signatures are exhibit reciprocal association with response to dual anti PDL1-VEGF therapy in relapsed mesothelioma. Further exploration is needed and ongoing to fully understand the genomic, immune microvironment and gut microbiome and their influence on these potentially predictive features.

Legal entity responsible for the study

University of Leicester.

Funding

Asthma and Lung UK.

Disclosure

A. Thomas: Other, Institutional, Expert Testimony: Bristol Myers Squibb; Other, Institutional, Speaker’s Bureau: Bristol Myers Squibb; Other, Institutional, Advisory Board: Bristol Myers Squibb. D.A. Fennell: Other, Institutional, Sponsor/Funding, Honoraria: Bayer; Boehringer Ingelheim; Bristol Myers Squibb; Inventiva Pharma; Novocure; RS Oncology; Targovax; Other, Institutional, Advisory Role: Bristol Myers Squibb; Inventiva Pharma; Novocure; Roche; Targovax; Other, Institutional, Speaker’s Bureau: Bristol Myers Squibb; Boehringer Ingelheim; Other, Institutional, Sponsor/Funding: Astex Pharmaceuticals; Bayer; Boehringer Ingelheim (Inst); Bristol Myers Squbb (Inst); Fuji Pharma (Inst); lab21 (Inst); Financial Interests, Institutional, Sponsor/Funding, Travel, Accommodations, Expenses: Bristol Myers Squibb; Janssen Oncology. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

29P - Circulating proteins associated with immunotherapy efficacy in patients with pancreatic ductal adenocarcinoma. (ID 130)

Presentation Number
29P
Lecture Time
12:30 - 12:30
Speakers
  • Troels D. Christensen (Herlev, Denmark)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Recent trials have shown a clinical efficacy of checkpoint inhibitors combined with radiotherapy for a subset of patients with pancreatic ductal adenocarcinoma (PDAC). However, predictive biomarkers are needed. Multiple proteins are released into the bloodstream due to cancer and immune reactions, and several circulating proteins have been associated with response to immunotherapy in other cancers. We investigated 90 immuno-oncology (I-O) related serum proteins in 78 patients with PDAC treated with nivolumab with or without ipilimumab combined with radiotherapy in a randomized phase 2 study (CheckPAC, NCT02866383).

Methods

Proteins were measured in serum samples collected at baseline and after 2 months of treatment using Olink Target 96 I-O panel (Olink Proteomics, Uppsala, Sweden). Serum protein levels were correlated with efficacy data using Wilcoxon test or t-test. Survival was analyzed with univariate and multivariate Cox-regression adjusting for performance status, Glasgow prognostic score, bilirubin, study arm, and weight loss. An unadjusted p-value of 0.05 was considered significant.

Results

Patients with clinical benefit (partial response (n=7) or stable disease (n=15)) had significantly higher levels of fas ligand (FASLG) and galectin 1 (Gal-1), and decreased C-C motif chemokine (CCL) 4 compared to patients without clinical benefit. High Gal-1 and FASLG and low angiopoietin-2 and mucin-16 were associated with longer progression-free survival in univariate analysis. In multivariate analysis, the association remained significant for Gal-1 (Hazard ratio (HR) = 0.25, confidence interval (CI) 0.12-0.56, p<0.001), and borderline significant for FASLG (HR=0.52, CI: 0.26-1.04, p=0.06). For 14 proteins, an increase in protein level during treatment was associated with progressive disease, whereas a decreasing level of 3 proteins (including CCL4) was associated with clinical benefit. Changes in FASLG and Gal-1 were not associated with clinical benefit rate.

Conclusions

The study identified Gal-1 and possibly FALSG as potential novel predictive biomarkers of immunotherapy efficacy in patients with PDAC. The results need to be confirmed in future studies.

Clinical trial identification

NCT02866383.

Legal entity responsible for the study

The authors.

Funding

Bristol Myers Squibb.

Disclosure

I. Chen: Financial Interests, Personal, Advisory Board: Amgen; Financial Interests, Institutional, Research Grant: Roche, Bristol Myers Squibb, Celgene, Genis, Varian Medical Systems. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

30P - Frequency of peripheral CD8+ T cells expressing chemo-attractant receptors increases in NPC patients with EBV clearance upon radiotherapy (ID 131)

Presentation Number
30P
Lecture Time
12:30 - 12:30
Speakers
  • Shweta Mahajan (Rotterdam, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Endemic nasopharyngeal carcinoma (NPC) is associated with persistent Epstein Barr Virus (EBV) infection. Radiotherapy (RT) is standard-of-care for NPC, where clearance of EBV DNA in plasma is a recognized predictor of response. The relationship between post-RT clearance of EBV and concomitant changes in circulating immune cell populations in blood is not known.

Methods

In this study, serial blood samples were prospectively collected from 24 NPC patients before, during and post-RT, and peripheral blood mononuclear cells (PBMCs) were analysed with multiplex flow cytometry to assess the frequency of T cell subsets according to >20 markers of differentiation, co-signalling and chemotaxis. Sera were analysed for the number of EBV DNA copies by PCR, and patients were classified either as having cleared or not cleared EBV post-RT.

Results

We observed that patients with EBV clearance demonstrated a lower frequency of PD1+ CD8 +T cells as well as CXCR3+ CD8+ T cells during and post-RT when compared to patients with no EBV clearance. Most notably, these patients showed a temporal increase in frequencies of CD8+ T cells expressing the chemo-attractant receptors CCR1, 4 and/or 5 upon RT, which was not observed in patients with no EBV clearance. The increase in frequency of CCR+ CD8+ T cells was accompanied by a drop in frequency of naïve CD8+ T cells and an increase in frequency of OX40+CD8+ T cells. Moreover, blood of non-recurrent NPC patients contained higher quantities of CCL14 and CCL15, constituting ligands for CCR1 and CCR5, prior to RT when compared to recurrent NPC patients.

Conclusions

Our findings suggest that plasma clearance of EBV DNA post-RT is preceded by increased frequencies of CCR1, 4 and/or 5+ CD8+ T cells, potentially serving as a predictor for EBV clearance in NPC patients, which requires validation in larger cohorts.

Legal entity responsible for the study

Erasmus MC.

Funding

Erasmus MC.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

31P - The IOpener study: tyrosine kinase activity profiling to predict response to immune checkpoint inhibitors in patients with advanced stage non-small cell lung cancer (ID 132)

Presentation Number
31P
Lecture Time
12:30 - 12:30
Speakers
  • Karlijn D. Joode (Rotterdam, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Treatment with immune checkpoint inhibitors (ICIs) is successful, but only in a subset of patients with advanced stage non-small cell lung cancer (NSCLC). Blood-based kinase profiling of peripheral blood mononuclear cells (PBMCs) has previously shown its predictive value for response to ICI treatment in a discovery cohort (Hurkmans et al., JITC 2020). The aim of the current prospective study was to perform a blind validation of the predictive value of kinase activity profiles in PBMCs for ICI response in patients with NSCLC in a standardized setting.

Methods

PBMCs from patients with advanced stage NSCLC included in this multicenter study were collected prior to ICI treatment. Tyrosine kinase activity of PBMCs were profiled using a micro-array with 144 different peptide-substrates (Pamchip). Classification analysis was based on grouping of patients with no progression vs. progression (RECIST v1.1) ≤24 weeks after start of treatment. Only patients treated with first-line pembrolizumab (+/- chemotherapy) and ≥24 weeks follow-up were included.

Results

The model was first established based on tyrosine kinase activity profiles, determined in a calibration cohort (N = 61) and then applied to an independent validation cohort (N = 63). The model had a predictive accuracy of 60% (CI95 = 47-72%), which was improved when the model was combined with PD-L1 expression: for patients with a PD-L1 score <50%, the accuracy was 72% (CI95 = 55-86%). For this subgroup of patients, the kinome analysis showed a significant lower progression-free survival for patients for whom progression was predicted vs. those for whom no progression was predicted (p <0.01, hazard ratio= 3.6, CI95 = 1.6-8.1).

Conclusions

The tyrosine kinome of PBMCs before ICI treatment can be applied to predict progressive disease in patients with advanced stage NSCLC, in particular when combined with low PD-L1 expression. This may imply enhanced predictive value of kinome profiling for tumors with low presence of CD8 T cells. The latter needs confirmation, for which data from an ongoing extension study will be used. The final aim is to implement the kinome analysis for clinical use.

Legal entity responsible for the study

The authors.

Funding

Pamgene International B.V.

Disclosure

K.D. Joode: Financial Interests, Personal, Other, Travel expenses congress: Ipsen. C.H. Van Der Leest: Financial Interests, Personal, Advisory Board: MSD, Janssens, BMS. B. Pinedo: Financial Interests, Personal, Advisory Board, 2017-2021: Pamgene International B.V., Vitromics; Financial Interests, Personal, Advisory Board: Glycostem B.V., Dulisco U.A., Biox Biosciences B.V., ORCA Therapeutic B.V., Qurin Diagnostics; Financial Interests, Personal, Stocks/Shares: Qurin Diagnostics. E. Kapiteijn: Financial Interests, Personal and Institutional, Advisory Role: Bristol Myers Squibb, Novartis, Merck; Financial Interests, Personal, Advisory Role: Pierre Fabre; Financial Interests, Personal, Research Grant, not related to this paper: Bristol Myers Squibb; Financial Interests, Institutional, Research Grant, not related to this paper: Pierre Fabre. R. Debets: Financial Interests, Institutional, Research Grant, paid to institution: MSD, Bayer; Financial Interests, Personal and Institutional, Advisory Role, paid to institution: Bluebird Gio; Financial Interests, Personal and Institutional, Advisory Board, paid to institution: Genticel; Other, Institutional, Other, Paid to institution: Pan Cancer T; Other, Institutional, Other, no's 21152822.9 and 21184727.2 (pending to Erasmus MC): European patent application. J.P. Groten: Financial Interests, Personal, Advisory Board: Single Cell Discoveries bv; Financial Interests, Personal, Advisory Role: Vitromics Health Care bv; Financial Interests, Personal, Other, employee: Pamgene International B.V.; Non-Financial Interests, Personal, Advisory Role: AstraZeneca Corporation, MSD Europe, Charles River Corporation, Vitromics Healthcare; Non-Financial Interests, Personal, Invited Speaker: European Society of Toxicology; Non-Financial Interests, Personal, Invited Speaker, Visiting professor: Wageningen University research. J.G. Aerts: Financial Interests, Personal, Advisory Board, Speakers fee: Eli Lilly, MSD, Takeda; Financial Interests, Personal, Advisory Board: Amphera, Pfizer, BMS; Financial Interests, Personal, Advisory Role, DMC member, speakers fee: BIOCAD; Financial Interests, Personal, Stocks/Shares: Amphera; Financial Interests, Personal, Stocks/Shares, Patent pending: Pamgene, Amphera; Financial Interests, Institutional, Principal Investigator, present participation in >60 clinical trials related to oncology (all compensation paid to intitution): Multiple; Non-Financial Interests, Personal, Member, Board member IASCL: IASLC. R.H. Mathijssen: Financial Interests, Personal, Invited Speaker: Novartis; Financial Interests, Personal, Advisory Role: Servier; Financial Interests, Personal, Stocks/Shares, patent pending: Pamgene International B.V.; Financial Interests, Institutional, Research Grant, Investigator-initiated research: Astellas, Bayer, Boehringer Ingelheim, Cristal Therapeutics, Pamgene, Pfizer, Novartis, Roche, Servier. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

32P - Discovering markers that identify pro-metastatic immune cell subsets. (ID 133)

Presentation Number
32P
Lecture Time
12:30 - 12:30
Speakers
  • Ansooya A. Bokil (Trondheim, Norway)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Due to lack of reliable markers that determine metastatic potential and guide cancer treatment; metastasis remains a major cause of cancer-related deaths. Immune cells, especially myeloid cells, contribute significantly to metastasis and could function as potential prognostic markers. However, due to their tremendous heterogeneity and plasticity; no reliable and clinically translatable markers to define pro-metastatic immune cell subsets have been identified. With this study we aim to gain deeper insights into myeloid immune cell activities in metastatic tumors to reveal potential markers that define pro-metastatic immune cells. We expect such markers will improve assessment of prognosis and further help devise therapeutic strategies.

Methods

To identify pro-metastatic functions and markers, we performed RNA sequencing on immune cells of 67NR (non-metastatic) and 66cl4 (metastatic) primary tumors, derived from the syngeneic 4T1 mouse breast cancer model. After data analysis, we validated the candidate of interest using flow cytometry, immunoblotting, ELISA, qPCR among other techniques. The translational potential of the identified marker was studied by immunohistochemistry on 487 breast cancer patient biopsies.

Results

We identified several biological processes specifically upregulated in immune cells of metastatic tumors. Interestingly, a metabolic enzyme- arginase1 (ARG1), with known wound healing and immunosuppressive functions was associated with the top enriched processes. Therefore, we investigated it further and found that ARG1 was expressed in myeloid cells, specifically immature neutrophils and macrophages, infiltrating the metastatic tumors but not in non-metastatic ones. This indicated the potential of myeloid cells that express ARG1 to serve as prognostic markers. Further, we analyzed breast cancer patient biopsies and showed that a higher number of ARG1+ infiltrating cells were associated with breast cancer tumors that are more prone to metastasis (basal and HER2 type).

Conclusions

We identified ARG1-containing myeloid immune cells as pro-metastatic markers and indicated their translational potential to clinic.

Legal entity responsible for the study

The authors.

Funding

NTNU and Helse Midt-Norge.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

33P - Changes in serum cytokine CXCL12 level can predict the survival of patients with non-small-cell lung cancer receiving anti-PD-1 treatment (ID 134)

Presentation Number
33P
Lecture Time
12:30 - 12:30
Speakers
  • Yanjun Xu (Hangzhou, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The circulating predictive factors for survival of advanced non-small-cell lung cancer (NSCLC) patients receiving immune checkpoint inhibitors (ICIs) remain elusive. We aimed to identify the predictive value of circulating cytokines and programmed cell death ligand-1 (PD-L1) expression for survival.

Methods

Serum samples from 102 advanced-stage NSCLC patients who underwent immunotherapy were collected at baseline. PD-L1 expression was also analyzed.

Results

Positive PD-L1 expression (≥1%) was detected in 49.0% of patients. Compared with PD-L1-negative patients, PD-L1-positive patients had a significantly higher ORR (70% vs. 28.8%, p<0.05) and an increased mPFS (25.35 vs. 4.64 months, p=0.003), and tended to have increased mOS (44.84 vs. 20.42 months, p=0.087). CXCL12 levels in the top 33% (1/3) was a poor prognostic factor for durable clinical benefit (DCB, 23.5% vs. 72.1%, p<0.001), PFS (3.76 vs. 14.4 months; p<0.001) and OS (12.2 vs. 44.84 months; p=0.008), and a signature comprising PD-L1<1% and top 33% CXCL12 was associated with lowest ORR (27.3% vs. 73.7%, p<0.001), DCB (27.3% vs. 73.7%, p<0.001) and the worst mPFS (2.44 vs. 25.35 months, p<0.001) and mOS (11.97 vs. 44.84 months, p=0.007). Area under the curve (AUC) analysis for PD-L1 expression, CXCL12 level or PD-L1 expression plus CXCL12 level to predict DCB or NDB showed AUC value as 0.680, 0.719 and 0.794 respectively.

Conclusions

Our findings suggest that the combination of circulating cytokine CXCL12 level and PD-L1 status can predict survival of advanced NSCLC patients treated with ICIs.

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 Poster Display session

34TiP - Prospective Analysis of INteStinal Microbiome and Autoimmune PanEls as PrediCtors of Toxicity in ImmunOncology Patients (INSPECT-IO Study) (ID 135)

Presentation Number
34TiP
Lecture Time
12:30 - 12:30
Speakers
  • Abdulazeez T. Salawu (Toronto, ON, Canada)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Despite potential life-changing morbidity or even mortality, there are no validated biomarkers for identifying patients at risk of developing serious immune-related adverse events (irAE) from immune checkpoint inhibitor (ICI) therapy. Autoimmune conditions and irAE both involve loss of tolerance to endogenous antigens with similar clinical manifestation. We have previously shown that individuals with irAE have higher baseline IgG autoantibody (autoAb) levels with a greater increase in IgG and IgM autoAb after ICI administration, compared to those without irAE. Previous studies have identified associations and potential mechanistic relationships between the gut microbiome and development of irAE. We hypothesize that changes in gut microbiome, autoAb profiles, and peripheral immunophenotype with ICI therapy are associated with the development of irAE and are influenced by immunosuppressive treatment for irAE.

Trial Design

INSPECT-IO is a prospective Princess Margaret Cancer Centre initiative that aims to identify correlates of irAE in adults (>18 years) with advanced solid tumors [NCT04107311]. Patients receiving ICI-based combination immunotherapy are included given their higher risk of irAE compared to those on ICI monotherapy. Those with a history of autoimmune disease and a flare episode within one year are excluded. Whole blood and stool samples are collected from all patients at baseline (≤ 28 days prior to ICI), at an early timepoint (2 – 6 weeks after starting ICI) and end of treatment (≤ 28 days after completion of ICI). Additional blood and stool samples are collected within 96 hours of onset, and upon resolution of every significant irAE (≥ grade 2 by CTCAE v5.0 or requiring systemic immunosuppression). Shotgun sequencing will be used to determine gut microbial taxonomic and metagenomic composition; autoAb profiling will be performed by multiplex proteomic arrays and immunophenotyping will be done using flow cytometry. Archival tumor tissue and organ-specific tissue biopsies for histologic irAE diagnosis will be stored for additional molecular analyses. Study enrolment is ongoing (37 patients to date) with a target accrual of 120 patients.

Clinical trial identification

NCT04107311.

Legal entity responsible for the study

University Health Network.

Funding

GSK.

Disclosure

A.R. Abdul Razak: Non-Financial Interests, Personal, Advisory Role: Adaptimmune, Bayer, GlaxoSmithKline; Non-Financial Interests, Institutional, Research Grant: Deciphera, Karyopharm Therapeutics, Pfizer, Roche/Genentech, Bristol Myers Squibb, MedImmune, Amgen, Blueprint Medicines, Merck, AbbVie, Interion Therapeutics, 23 and me, Rain Therapeutics, Neoleukin, Daiichi Dankyo, Symphogen; Non-Financial Interests, Personal and Institutional, Research Grant: GlaxoSmithKline, Adaptimmune; Non-Financial Interests, Personal, Expert Testimony: Medison. P.L. Bedard: Financial Interests, Institutional, Invited Speaker: AstraZeneca, Bicara, BMS, Amgen, Novartis, Genentech/Roche, Sanofi, Merck, Pfizer, Zymeworks, Nektar Therapeutics, Lilly, SeaGen; Financial Interests, Institutional, Research Grant: Pfizer; Financial Interests, Institutional, Funding: Servier; Non-Financial Interests, Personal, Invited Speaker, Executive Board Member: Breast International Group; Non-Financial Interests, Personal, Leadership Role, Chair: AACR Project GENIE; Non-Financial Interests, Personal, Leadership Role, Past Chair IND Committee Member, Breast Site Steering Committee: Canadian Clinical Trials Group; Non-Financial Interests, Personal, Advisory Role: SeaGen, Lilly, Amgen, Merck, BMS, Pfizer, Gilead. L.L. Siu: Financial Interests, Personal, Advisory Board: Merck, AstraZeneca, Roche, Oncorus, Seattle Genetics, Voronoi, Arvinas, Tessa, Navire, Relay Therapeutics, Janpix, Amgen, Marengo, InterRNA, Medicenna, Hoopika, Coherus; Financial Interests, Personal, Other, Spouse is co-founder: Treadwell Therapeutics; Financial Interests, Personal, Stocks/Shares, Spouse has stock ownership: Agios; Financial Interests, Institutional, Invited Speaker: Novartis, Bristol Myers Squibb, Pfizer, Boehringer Ingelheim, GlaxoSmithKline, Roche/Genentech, AstraZeneca, Merck, Astellas, Bayer, Amgen, Symphogen, Intensity Therapeutics, Shattucks, EMD Serono; Non-Financial Interests, Personal, Advisory Role: ICR. B. Coburn: Non-Financial Interests, Personal and Institutional, Other, In Kind: Nubiyota. A. Spreafico: Financial Interests, Personal, Advisory Board: Merck, Bristol Myers Squibb, Novartis, Oncorus, Janssen, Medison & Immunocore; Financial Interests, Institutional, Research Grant: Novartis, Bristol Myers Squibb, Symphogen, AstraZeneca/MedImmune, Merck, Bayer, Surface Oncology, Northern Biologics, Janssen Oncology/Johnson & Johnson, Roche, Regeneron, Alkermes, Array Biopharma/Pfizer, GlaxoSmithKline, Treadwell, Amgen. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

37P - Synthetic tumour-infiltrating interleukin(IL)-12 for targeted and tolerable immunotherapy reduces metastasis in pancreatic cancer (ID 136)

Presentation Number
37P
Lecture Time
12:30 - 12:30
Speakers
  • Melanie Herpels (London, United Kingdom)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Metastatic Pancreatic Ductal Adenocarcinoma (PDAC) is a root cause of lethality in patients. The metastasis is promoted by abundant proteases in the PDAC microenvironment. Synthetic immuno-oncology opens new avenues towards overcoming limitations of standard immunotherapy agents, previously unsuccessful in PDAC. Anti-tumour cytokines such as IL-12 poorly infiltrate the stroma of solid tumours and trigger toxicity. Here, by exploiting the abundant proteases in PDACs, a targeted and tolerable masked Collagen-Binding Domain CBD-Il12 (mCBD-Il12) was produced by fusing the mask to the p35 subunit of Il-12 through a cleavable linker. This linker is cleaved by PDAC-specific proteases, leading to activation of mCBD-Il12 upon tumour infiltration.

Methods

Human PDAC samples were analysed for the expression of MMP2/9 and PLAU in established subtypes (classical and quasi-mesenchymal). mCBD-IL12 with a linker sensitive to such protease activity was therefore produced in HEK293F cells and purified using immobilised metal affinity and size exclusion chromatography. The ability of various murine tissues and their associated proteases to cleave the linker was verified through western blotting. Finally, the molecule was tested in a metastatic and mesenchymal pancreatic syngeneic mouse model.

Results

The quasi-mesenchymal subtype was found to express MMP2/9/PLAU to a higher degree than the other Collisson/Sadanandam PDAC subtypes. In addition, we found the linker to be readily cleaved by murine pancreatic tumour lysate in contrast with non-cancerous pancreatic tissue in vitro. Interestingly, mCBD-Il12 was found to considerably reduce the number of liver metastases and ascites volume compared to wild-type Il-12 or controls in a murine PDAC model. The ascites colour varied from white in wild-type Il-12 to light red in mCBD-Il12, and dark red in controls. Analysis of the blood-based cytokines is pending.

Conclusions

MCBD-Il12 is a novel bioengineered immunotherapy molecule that seems to suppress liver metastases in murine PDAC models. Further studies are required to investigate the changes in the tumour microenvironment and immune infiltrates involved.

Legal entity responsible for the study

The authors.

Funding

Cancer Research UK - Convergence Science Centre.

Disclosure

A. Mansurov, J. Ishihara: Financial Interests, Institutional, Stocks/Shares: Arrow Immune Inc; Financial Interests, Institutional, Other, Inventor: Patent number: US62/878,574, 2019. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

39P - The expanded CD8+ tumor-infiltrating lymphocyte tumor-specific granzyme response is dominated by granzyme B (ID 138)

Presentation Number
39P
Lecture Time
12:30 - 12:30
Speakers
  • Anne-Christine K. Rasmussen (Herlev, Denmark)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Recent ground-breaking results from a randomized phase III clinical trial showed tumor-infiltrating lymphocyte (TIL)-therapy to be superior to ipilimumab in anti-PD-1 refractory advanced melanoma patients, paving the way for TIL-therapy to become a standard treatment (Ref 1). Responses to TIL-therapy are thought to be primarily mediated by expanded CD8+ TILs (CD8+ REP-TILs) via secretion of cytotoxic molecules, including granzymes (Gzms) of which five distinct types (A, B, H, K, and M) have been identified in humans. To better understand the role of Granzymes in TIL-therapy, we characterized the granzyme profile of CD8+ REP-TILs during tumor cell recognition.

Methods

Matched CD8+ REP-TILs and autologous tumor cell line (TCL) pairs were generated using fresh metastatic melanoma samples. TIL/TCL pairs with known high tumor-reactivity were selected and employed in in vitro co-culture systems reproducing TIL: tumor recognition. Gzm mRNA upregulation was assessed via bulk mRNA (n=10) and single-cell RNA sequencing (n=6) of expanded TILs, +8 hours post co-culture. Gzm protein secretion was quantified by ELISA and flow cytometry post co-culture (+2, +8, and +24 hours, n=11).

Results

Bulk and single-cell RNAseq data analysis demonstrated distinct upregulation of only GzmB by CD8+ REP-TILs post TCL recognition. Protein levels were similarly dominated by GzmB, representing 61%, 73%, and 48% of total secreted Gzm at +2, +8 and +24 hours, respectively. In addition, GzmB showed by far the greatest post TCL recognition fold induction, up to 23.8-fold, whereas other Gzms were below 6.0-fold. Overall, GzmA was secreted at high levels, although the response was largely unspecific and only minimally induced by TCL recognition (up to 4.9-fold increase). All remaining granzymes were secreted and induced at minimal levels.

Conclusions

GzmB dominates the tumor-specific granzyme-related response of CD8+ REP-TILs, suggesting GzmB as the primary effector molecule in patients treated with TIL-therapy. GzmB secretion is therefore a potential candidate for a TIL-therapy potency biomarker. The potential of less inducible Gzms, such as GzmA, requires further clarification. Ref 1: Haanen et al. 2022 ESMO Congress (LBA3, Presidential Symposium I).

Legal entity responsible for the study

The authors.

Funding

Lundbeck Foundation (grants R286-2018-991, R307-2018-3636), The Danish Cancer Society (R309-A17896), Herlev and Gentofte Research Council (Clinician-Scientist grant to Marco Donia).

Disclosure

I. Svane: Financial Interests, Personal, Advisory Board: BMS, Pierre Fabre, Novartis; Financial Interests, Personal, Invited Speaker: MSD, Pierre Fabre, Novartis, Roche, BMS, 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, Roche, TILT Biotherapeutics, Lytix Biopharma, Novartis. M. Donia: Financial Interests, Personal, Invited Speaker, Teaching: Roche, Novartis; Non-Financial Interests, Personal, Other, Sub-investigator of clinical trial with connected translational research: Bristol Myers Squibb; Non-Financial Interests, Personal, Proprietary Information: Genentech, Bristol Myers Squibb; Financial Interests, Personal, Other, Advisor: Achilles Therapeutics; Other, Institutional, Other, Chairman of the Melanoma and Non-melanoma Skin Cancer Scientific Committee: Danish Medicines Council. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

40P - Can we predict which TIL products will react against NSCLC tumours based on the immune infiltrates? (ID 139)

Presentation Number
40P
Lecture Time
12:30 - 12:30
Speakers
  • Suzanne Castenmiller (Amsterdam, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Adoptive transfer of tumour infiltrating lymphocytes (TIL therapy) has shown high efficacy in a phase III trial for melanoma patients (M14TIL), and in phase I trials for other solid cancers. However, not all patients respond to TIL therapy. Good prediction tools would help to select which patients may benefit most. The tumour microenvironment is infiltrated by different types of lymphoid and myeloid cells, which communicate with each other. We therefore hypothesized that the presence of specific immune cell types may explain the variation in TIL products.

Methods

To define tumour-specific alterations of immune infiltrates, we characterized the myeloid and lymphoid cell populations present in tumour lesions and healthy adjacent tissue from 26 early-stage NSCLC patients by flow cytometry. We generated TIL products according to the clinical expansion protocol and determined their tumour reactivity based on cytokine production upon co-culture with the autologous tumour digest. Polyfunctional T cells were defined as cells producing at least two cytokines. Data were analysed using Cytotree, a R/Bioconductor package to analyse flow data in an unbiased fashion. Spearman’s Rank Correlation was used to correlate immune infiltrates with expansion rate and percentage of polyfunctional T cell.

Results

The majority of immune cells in the tumour tissue consisted of lymphoid cells (T cells 67%; B cells 16%; NK(T) cells 3.5%), which were all increased compared to the healthy adjacent tissue (T cells 33.6%; B cells 2.6%; NK(T) cells 13%). This increase was at cost of monocyte and dendritic cell infiltrates, which were decreased in tumour tissue (9%) compared to the healthy adjacent tissue (18%). None of the immune infiltrates correlated with the expansion rate of TILs. Monocytes, dendritic cell and NK cell infiltrates showed a negative association with the percentage of polyfunctional T cells. Interestingly, the presence of B cells was positively associated with the percentage of polyfunctional T cells.

Conclusions

We found that specific immune infiltrates in the tumour tissue associate with the functionality of TIL products, which may help select for TIL products with highly responsive T cells against NSCLC tumours.

Legal entity responsible for the study

The authors.

Funding

Sanquin.

Disclosure

J.B.A.G. Haanen: Financial Interests, Institutional, Advisory Board: Bristol Myers Squibb, Achilles Therapeutics, Ipsen, Merck Sharpe & Dohme, Merck Serono, Pfizer, Molecular Partners, Novartis, Roche, Sanofi, Third Rock Venture, Iovance Biotherapeutics; 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, Stocks/Shares: Neogene Therapeutics; Financial Interests, Institutional, Research Grant: Bristol Myers Squibb, BioNTech US, Merck Sharpe & Dohme, Amgen, Novartis, Asher Bio; Non-Financial Interests, Personal, Member: ASCO, AACR, SITC; Other, Personal, Other, Editorial Board ESMO Open: ESMO; Other, Personal, Other, Editor-in-Chief IOTECH: ESMO; Other, Personal, Other, Editorial Board: Kidney Cancer. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

42P - Tumor-reactive CD8+ T cells in ovarian and colon cancer in tumors and cell products (ID 141)

Presentation Number
42P
Lecture Time
12:30 - 12:30
Speakers
  • Sara Bobisse (Lausanne, Switzerland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Several pivotal studies established the high prognostic value of tumor-infiltrating lymphocytes (TILs) in patients with solid tumors, including colorectal (CRC) and ovarian (OvCa) cancer. Albeit the association between clinicopathological outcomes and tumor infiltration supports the intervention with immunotherapeutic strategies, TIL-adoptive cell transfer (ACT) only led to clinical results in a subset of patients with non-melanoma tumors, raising the question of the abundance of tumor-reactive T cells in these cancers. In the present study, we interrogated CRC and OvCa patients for the presence and features of neoantigen (neoAg) and tumor-specific T cells in in vitro-expanded and also in situ TILs.

Methods

We collected tumors from 30 patients with either early and advanced CRC or advanced OvCa. Neoepitope- and tumor-specific CD8 T cells were identified and FACS-sorted from blood or in vitro-expanded TILs. In parallel, T-cell receptor sequencing (TCR-Seq), including single cell TCR-seq (scTCR-seq) was performed on tumor samples followed by TCR annotation based on a robust in-house pipeline for TCR cloning and screening.

Results

Tumor-specific in vitro-expanded TILs were identified in most CRC and OvCa patients and originated from clonotypes of a broad range of intratumoral frequencies. In CRC, the presence of TILs targeting NeoAg correlated with the density of activated and exhausted TILs in both tumor and stroma. Furthermore, higher structural affinity TCRs preferentially infiltrated tumors. By interrogating libraries of scRNA/TCRseq from tumor samples, several additional tumor-specific TCRs were identified and their transcriptomic profile led to a signature and a predictor of tumor specific TCRs.

Conclusions

Although tumor-specific TILs are consistently detected in CRC and OvCa patients, they are not clonally expanded in tumors and do not efficiently proliferate in conventional cell culture conditions. Their transcriptomic profiles represent a unique dataset that can be exploited to determine cell culture conditions to guide next generation immunotherapies as well as biomarkers discriminating patients eligible for TIL-ACT.

Legal entity responsible for the study

The authors.

Funding

Ludwig Institute for Cancer Research.

Disclosure

L. Kandalaft: Financial Interests, Institutional, Full or part-time Employment: CHUV Lausanne University Hospital, Ludwig Cancer Research Institute Lausanne Branch; Financial Interests, Institutional, Research Grant: Research Foundation for the treatment of Ovarian cancer; Financial Interests, Institutional, Research Grant, Development of a Novel B Cell-based Vaccine for Metastatic Solid Cancers: ISREC Foundation; Financial Interests, Institutional, Research Grant, Development of a lung personalized Dendritic Cell Vaccine: Impact Cancer Foundation; Financial Interests, Institutional, Research Grant, Development of a Pancreatic personalized Dendritic Cell Vaccine in combination with PDL-1: BMS. G. Coukos: Financial Interests, Institutional, Research Grant: Celgene, Boehringer Ingelheim, Roche, BMS, Iovance Therapeutics, Kite Pharma; Financial Interests, Institutional, Advisory Board: Genentech, BMS, AstraZeneca, NextCure, Geneos Tx, Sanofi/Aventis, Roche. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

43P - Development of a production process to generate CD8+ T cell-enriched tumor infiltrating lymphocyte (TIL) products with increased cytotoxic potential for the treatment of patients with solid cancers. (ID 142)

Presentation Number
43P
Lecture Time
12:30 - 12:30
Speakers
  • Liselotte Tas (Amsterdam, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Treatment with autologous tumor-infiltrating lymphocytes (TILs) can induce remarkable clinical responses. The absolute numbers of CD8+ T cells in TIL products have been shown to correlate with clinical responses upon TIL therapy. With the current production process, the numbers of CD8+ T cells in the TIL products vary greatly between patients. By using a targeted cytokine that preferentially activates CD8+ T cells, we aimed to increase the cytotoxic potential of the TIL products.

Methods

A cis-targeted CD8-IL2 molecule (Asher Biotherapeutics) was used to promote CD8+ T cell outgrowth from tumor digests. In the rapid expansion protocol (REP), TILs were subjected to polyclonal stimulation using anti-CD3 antibodies (OKT-3) or anti-CD3/CD28 polymers (TransAct™) in the presence of CD8-IL2. The standard ‘young TIL’ production process using high dose IL-2 and OKT-3 was used as a standard comparison. TIL product composition, T cell differentiation phenotype and tumor-reactivity was assessed by flow cytometry and ELISA.

Results

7 tumors (5 melanoma, 1 cervical carcinoma and 1 endometrial carcinoma) were subjected to enzymatic digestion. At the end of the REP phase, TIL products cultured with CD8-IL2 contained 97.5% (range 92-98%) CD8+ T cells, compared to 77.2% (range 35-81%) for products cultured with conventional IL-2 (p = 0.036). Highest total CD8+ T cell numbers were obtained using CD8-IL2 in combination with TransAct in the REP. T cell differentiation phenotypes of the TIL products generated with CD8-IL2 were similar to the standard production process, showing that the improved expansion and CD8+ T cell enrichment did not come at the expense of a more exhausted phenotype. Upon restimulation with tumor digest clear anti-tumor reactivity of the TIL products could be demonstrated based on IFN-y production and tumor cell kill.

Conclusions

This study shows that CD8 cis-targeted IL-2 can be used to generate TIL products mainly comprising CD8+ T cells, thereby potentially improving cytotoxic potential and therapeutic efficacy. The use of CD3/28 TransAct, compared to anti-CD3 stimulation (OKT-3) improved the final yield of CD8+ T cells in the TIL products.

Legal entity responsible for the study

Netherlands Cancer Institute.

Funding

Asher Biotherapeutics.

Disclosure

L. Tas: Financial Interests, Institutional, Funding: Asher Biotherapeutics. M. van Zon: Financial Interests, Institutional, Funding: Asher Biotherapeutics. I. Djuretic: Financial Interests, Institutional, Full or part-time Employment: Asher Biotherapeutics. K.D. Moynihan: Financial Interests, Institutional, Full or part-time Employment: Asher Biotherapeutics. Y.A. Yeung: Financial Interests, Institutional, Full or part-time Employment: Asher Biotherapeutics. N. Mathewson: Financial Interests, Institutional, Full or part-time Employment: Asher Biotherapeutics. J.B.A.G. Haanen: Financial Interests, Institutional, Advisory Board: Bristol Myers Squibb, Achilles Therapeutics, Ipsen, Merck Sharpe & Dohme, Merck Serono, Pfizer, Molecular Partners, Novartis, Roche, Sanofi, Third Rock Venture, Iovance Biotherapeutics; 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, Stocks/Shares: Neogene Therapeutics; Financial Interests, Institutional, Research Grant: Bristol Myers Squibb, BioNTech US, Merck Sharpe & Dohme, Amgen, Novartis, Asher Bio; 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. Jedema: Financial Interests, Institutional, Funding: Asher Biotherapeutics.

Collapse
Poster Display Poster Display session

44P - In situ CAR Therapy Using oRNA™ Lipid Nanoparticles Regresses Tumors in Mice (ID 143)

Presentation Number
44P
Lecture Time
12:30 - 12:30
Speakers
  • Robert Mabry (Cambridge, MA, United States of America)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

LNP-mediated delivery of long coding RNA has been clinically validated for vaccines and gene editing. We have been developing a novel, synthetic, circular coding RNA platform (oRNA technology) which exhibits significant improvements in production, expression and formulation compared to mRNAs. Lacking the cap structure of mRNA, our oRNA technology uses a proprietary sequence-based IRES element to initiate protein translation in target cells. At the same time, ex vivo generated chimeric antigen receptor (CAR) T cell therapies have had tremendous success in treating hematologic malignancies, yet manufacturing, safety and efficacy challenges remain. At Orna Therapeutics, we are combining oRNA technology with novel immunotropic LNPs to address these challenges, by creating off-the-shelf “autologous” in situ CAR (isCAR™) therapies.

Methods

Orna’s immunotropic LNPs show preferential biodistribution to the spleen, with oRNA reporter expression detected in multiple immune cell subsets, including T cells, macrophages and NK cells. Delivery to immune cells is preserved across mice, rats and non-human primates. In vitro, expanding human T cells expressing an anti-human CD19 CAR oRNA show potent and sustained cytotoxicity and pro-inflammatory cytokine production compared to controls. To maximize protein expression, we developed FoRCE (Formulated oRNA Cell-based Evaluation)[AB1] [AW2] : a robust high-throughput platform that enables parallel arrayed synthesis, purification, lipid nanoparticle (LNP) formulation, and cell-based screening of oRNAs. We applied FoRCE to almost 3,000 unique oRNAs containing UTRs extracted from viral genomes and discovered hundreds of IRESs that drive translation from synthetic oRNA in primary human T cells, hepatocytes, and myotubes.

Results

Select IRESs from this screen drove high levels of CAR expression in primary human T cells. This elevated CAR expression translated to signficantly improved tumor regression in a human PBMC-engrafted NALM6 tumor-bearing mouse model. Tumor regression was dose-dependent, and the novel immunotropic LNP was well tolerated. oRNA-enabled isCAR therapies promise a re-dosable and scalable immune cell therapy.

Conclusions

This off-the-shelf treatment dose not require leukapheresis or lymphodepletion, and the transient expression of isCAR may provide better management of cytokine release syndrome (CRS) and complexities associated with tumor lysis as compared to conventional autologous cell therapy. Future opportunities exist to expand targeting strategies and leverage a payload capacity (up to 12 kb) well beyond the current cell therapy delivery space.

Legal entity responsible for the study

The authors.

Funding

Orna Therapeutics.

Disclosure

R. Mabry: Financial Interests, Institutional, Stocks/Shares: Orna Therapeutics. A. Becker, A. Wesselhoeft, A. Horhata: Financial Interests, Personal, Stocks/Shares: Orna Therapeutics.

Collapse
Poster Display Poster Display session

45P - The RevCAR T cell platform: a switchable and combinatorial therapeutic strategy for glioblastoma (ID 144)

Presentation Number
45P
Lecture Time
12:30 - 12:30
Speakers
  • Nicola Mitwasi (Dresden, Germany)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Glioblastoma (GBM) is a very aggressive brain tumor, associated with poor prognosis and survival. So far, the efficiency of available therapies is limited. After proving their effectiveness in targeting hematological malignancies, chimeric antigen receptor (CAR) T cells might provide a promising therapeutic approach for GBM. Here, we present our switchable Reverse CAR technology (RevCAR). Unlike conventional CAR T cells, RevCAR T cells contain an epitope in their extracellular receptor domain, and can only be activated via bispecific target modules (RevTM) which recognize the RevCAR T cells on one side and tumor cells on the other side. Once these RevTMs are eliminated, RevCARs are switched off. In addition, we have developed dual-targeting RevCARs, allowing the control of T cells according to the AND gate logic of Boolean algebra.

Methods

Novel RevTMs specific for GBM were developed. Subsequently, they were expressed in eukaryotic cells and purified with affinity chromatography. The ability of these RevTMs to bind GBM cells and RevCAR T cells was analyzed using flow cytometry. Moreover, the capability of the mono-specific and the dual-targeting RevCAR T cells to kill GBM cells was analyzed using luminescence-based cytotoxicity assay in the absence or presence of a range of RevTM concentrations. Secreted pro-inflammatory cytokines were also evaluated by ELISA. In addition to the in vitro assays, a proof of concept co-injection experiment was performed in vivo.

Results

In this study, we show that GBM-specific RevTMs can bind both the RevCAR T cells and GBM cells. Importantly, the RevCAR T cells can be activated to secrete pro-inflammatory cytokines and to efficiently kill GBM cells via the RevTMs. Moreover, we were able to prove that dual-targeting RevCAR T cells can only be activated upon recognition of two different GBM targets, thereby allowing a highly specific and selective killing of GBM both in vitro and in vivo.

Conclusions

In conclusion, the switchable RevCAR platform is a novel therapeutic approach that provides improved safety, and allows combinatorial targeting of GBM.

Legal entity responsible for the study

Helmholtz-Zentrum Dresden-Rossendorf.

Funding

EU-Europa fördert Sachsen-Europäischer Fonds für regionale Entwicklung-der Sächsische Landtag.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

46P - Development of an allogeneic CAR-T targeting MUC1-C (MUC1, cell surface associated, C-terminal) for epithelial derived tumors (ID 145)

Presentation Number
46P
Lecture Time
12:30 - 12:30
Speakers
  • David Oh (San Francisco, CA, United States of America)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Most solid tumors are of epithelial origin and express Mucin 1 (MUC1), a heterodimer of MUC1-N and the oncogenic subunit MUC1-C. Many drugs targeting MUC1 in clinical trials have been primarily directed against MUC1-N. Since MUC1-C is present broadly in tumor due to loss of cell polarity, exposure via hypoglycosylation and MUC1-N shedding, it may represent a more tumor-selective target than MUC1-N. P-MUC1C-ALLO1 is an allogeneic CAR-T targeting MUC1-C and is manufactured using transposon-based integration (piggyBac® DNA Delivery System) and the Cas-CLOVER™ Gene Editing System to knockout the TCR and MHC class I proteins resulting in an enriched T stem cell memory product. Thus, P-MUC1C-ALLO1 addresses multiple common solid tumor indications.

Methods

MUC1-C epitope expression was investigated by IHC using the scFv binder for P-MUC1C-ALLO1 CAR in epithelial tumor and normal frozen tissue arrays. Pre-clinical efficacy of P-MUC1C-ALLO1 was tested in xenograft models for triple-negative breast (TNBC) and ovarian cancers. Clinical safety has been evaluated in three patients in a phase I trial (NCT05239143).

Results

MUC1-C epitope was positive in multiple tumor samples. While tumor expression was relatively nonpolarized, normal tissue expression was restricted to the apical surface. P-MUC1C-ALLO1 demonstrated robust infiltration and activity in TNBC and ovarian cancer xenografts, with >90% of tumor mass comprised of CAR-Ts at day 10, and 100% tumor elimination at 2 weeks. In the phase I trial, 4 pts (esophageal, colorectal, breast, and pancreatic carcinomas) have been infused either at 0.75x106 (pts 1-3) or 2x106 cells/kg (pt 4). No P-MUC1C-ALLO1 related toxicities were observed. Early efficacy was seen at the low dose with one partial response in pt 3 (HR+, Her2- Breast cancer).

Conclusions

MUC1-C epitope is highly expressed across common epithelial cancers and is apically restricted in normal tissues. Potent anti-tumor activity is seen in preclinical models. In early phase I experience, P-MUC1C-ALLO1 is safe and tolerable with an early signal of efficacy at a low starting dose. P-MUC1C-ALLO1 phase I trial enrollment is on-going.

Clinical trial identification

NCT05239143.

Legal entity responsible for the study

Poseida Therapeutics, Inc.

Funding

Poseida Therapeutics, Inc.

Disclosure

D. Oh: Financial Interests, Personal, Invited Speaker, Licensing fees relating to a patent on the use of T cell receptor sequencing as a predictive marker of immune-related adverse events with immunotherapy: Regents of the University of California; Financial Interests, Personal and Institutional, Invited Speaker, Research support: Merck Sharp and Dohme, PACT Pharma, Poseida Therapeutics, TCR2 Therapeutics, Roche/Genentech, Nutcracker Therapeutics; Non-Financial Interests, Institutional, Proprietary Information, Proprietary materials for laboratory experimentation: Nutcracker Therapeutics, 3T Biosciences. J. Henry: Financial Interests, Personal, Full or part-time Employment, Associate Director/Oncologist: Sarah Cannon Research Institute; Financial Interests, Personal, Stocks/Shares: HCA; Financial Interests, Institutional, Invited Speaker: Abbiscko Therapeutics, ABl Bio, Accutar biotech ADC therapeutics, Agenus, Aileron Therapeutics, Amgen inc, Artios, AstraZeneca, Bicycle Therapeutics, BioAlta, BioInvent Pharma, Biosplice Therapeutics, Black Diamond Therapeutics, Boehringer, Ingelheim, Cyteir, Daiichi Sankyo, Eli Lilly, Epizyme, Erasca, Exelixis, FujiFilm, GSK, Hutchison MediPharma, ICON plc, IGM Biosciences, Immunogen, Jacobio Pharmaceuticals, Jounce Pharma, Jubilant therapeutics, Loxo Oncology, Merck& CO, Metabomed, Molecular templates, Navire Pharma, Nikang pharmaceuticals, Oncorus, Prelude therapeutics, Poseida, PureTech, Pyramid, Rascal Therapeutics, Regeneron, Relay Therapeutics, Rgenix inc, Ribon therapeutics, Sapience, Sarah Cannon Development Innovations, Sarah Cannon Research Institute, Seagen, Simcha Therapeutics, Siranomics, Stingthera, Synthorx inc, Takeda pharmaceuticals, Tallac therapeutics, Tarveda, Teneothree, Tesaro, Turning Point Pharma, Xencor. J.C. Baranda: Financial Interests, Institutional, Other, Consultant: Sanofi; Financial Interests, Personal, Stocks/Shares: Aprea, Moderna, Zyme, Merus, Hutchmed; Financial Interests, Institutional, Invited Speaker: Astellas, Nektar, Sanofi, Takeda, Pfizer, SQZ, Synermore, Changchun Intellicrown, Genome and Company, Sumitomo Pharma, Xencor. E.E. Dumbrava: Financial Interests, Personal and Institutional, Research Grant: Bayer Healthcare Pharmaceuticals Inc, Immunocore Ltd, Amgen, NCI, Aileron Therapeutics, Compugen Ltd, TRACON Pharmaceuticals Inc, Unum Therapeutics, Immunomedics, BOLT Therapeutics, Aprea Therapeutics, Bellicum Pharmaceuticals, PMV Pharma, Triumvira, Seagen Inc, Mereo BioPharma 5 Inc, Sanofi, Astex Therapetics ; Financial Interests, Personal, Advisory Board: BOLT Therapeutics, Mersana; Financial Interests, Personal, Advisory Role: Catamaran Bio. E. Cohen: Financial Interests, Personal, Other, Consulting: Axelia, Cel Sci, Eisai, Hoopika, ImmunoSensor, Istari, Janssen, Kahr Medical, Mana Therapeutics, Merck, Mirati, MSD, Nectin Tx, Pangea Therapeutics, Roche; Financial Interests, Personal, Other, DSMB: Ayala, Kura; Financial Interests, Personal, Stocks/Shares: Kinnate Biophama, Primmune Therapeutics. J.D. Eskew: Financial Interests, Personal, Full or part-time Employment: Poseida Therapeutics; Financial Interests, Personal, Stocks/Shares: Poseida Therapeutics. R. Belani: Financial Interests, Personal, Stocks/Shares: Poseida, Amgen; Financial Interests, Personal, Full or part-time Employment: Poseida. J. McCaigue, H. Namini, C. Martin, A. Murphy: Financial Interests, Personal, Full or part-time Employment: Poseida; Financial Interests, Personal, Stocks/Shares: Poseida. E. Ostertag: Financial Interests, Personal, Member of the Board of Directors: Poseida Therapeutics; Financial Interests, Personal, Stocks/Shares: Poseida Therapeutics; Financial Interests, Personal, Full or part-time Employment: Poseida Therapeutics. J. Coronella, D. Shedlock: Financial Interests, Personal, Full or part-time Employment: Poseida Therapeutic; Financial Interests, Personal, Stocks/Shares: Poseida Therapeutics. I.I. Rodriguez Rivera: Financial Interests, Institutional, Full or part-time Employment: Next Oncology; Non-Financial Interests, Personal, Principal Investigator: Omega Therapeutics, Poseida Therapeutics, Werewolf Therapeutics, Merck KGaA.

Collapse
Poster Display Poster Display session

47P - Phase 1 Study to Assess the Safety and Efficacy of P-BCMA-ALLO1, a Fully Allogeneic CAR-T Therapy, in Patients with Relapsed / Refractory Multiple Myeloma (RRMM) (ID 146)

Presentation Number
47P
Lecture Time
12:30 - 12:30
Speakers
  • Mehmet H. Kocoglu (Baltimore, MD, United States of America)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

P-BCMA-ALLO1 is an allogeneic Chimeric Antigen Receptor T-cell (CAR-T) targeting B-cell Maturation Antigen (BCMA) being investigated in RRMM. P-BCMA-ALLO1 is manufactured using non-viral transposon-based integration (piggyBac®DNA Delivery System) that introduces a humanized anti-BCMA VH-based CAR producing a highly enriched T stem cell memory product. The Cas-CLOVER™ Site-Specific Gene Editing System eliminates endogenous T cell receptor (TCR) expression via knockout of the TCR beta chain 1 gene to prevent graft-vs-host disease, and the beta-2 microglobulin gene to reduce MHC class I expression to eliminate host-vs-graft responses. P-BCMA-ALLO1 demonstrated compelling activity in MM xenografts, providing rationale for this first-in-human phase I study.

Methods

The primary objective is to assess the safety and maximum tolerated dose based on dose limiting toxicity (DLT) in RRMM patients who have received a proteasome inhibitor, immunomodulatory drug and anti-CD38 monoclonal antibody. Secondary objectives will assess the anti-myeloma effect. The protocol utilizes standard 3+3 dose escalation to treat 40 patients. Patients receive lymphodepleting chemotherapy (LDC) with cyclophosphamide (300 mg/m2/day) / fludarabine (30 mg/m2/day) on days -5, -4 and -3 followed by a single dose of P-BCMA-ALLO1 on Day 0.

Results

As of 21SEP2022, 7 patients were treated with P-BCMA-ALLO1. Six patients received the cohort 1 dose of 0.75 X 106 CAR-T cells/kg and 1 patient received the cohort 2 dose of 2 X 106 cells/kg. To date, 4 cohort 1 patients have completed the DLT evaluation period and are evaluable for response. Most adverse events (AE) were grade 1 and 2. One patient had a serious AE of G3 febrile neutropenia. DLTs, cytokine release syndrome and neurotoxicity have not been observed. To date, 1 patient achieved very good partial response, 2 patients achieved partial response, and 1 patient had stable disease. Responses were seen starting at week 2, and overall response rate is 75%.

Conclusions

Early results demonstrate acceptable toxicity profile and promising efficacy for P-BCMA-ALLO1. Dose escalation is ongoing. Updated safety and efficacy results will be presented.

Clinical trial identification

NCT04960579.

Legal entity responsible for the study

Poseida Therapeutics.

Funding

Poseida Therapeutics.

Disclosure

M.H. Kocoglu: Financial Interests, Personal and Institutional, Principal Investigator: Poseida Therapeutics. A. Asch: Financial Interests, Personal and Institutional, Principal Investigator: Poseida Therapeutics; Financial Interests, Personal and Institutional, Research Grant: Gilead Sciences. A. Ramakrishnan: Financial Interests, Personal and Institutional, Principal Investigator: Poseida Therapeutics. C. Bachier: Financial Interests, Personal and Institutional, Principal Investigator: Poseida Therapeutics. T. Martin: Financial Interests, Personal and Institutional, Principal Investigator: Poseida Therapeutics; Financial Interests, Personal and Institutional, Research Grant: Sanofi, Amgen, Janssen, Seattle Genetics; Financial Interests, Personal, Advisory Role: GSK, Juno, Roche. T. Rodriguez: Financial Interests, Personal and Institutional, Principal Investigator: Poseida Therapeutics; Financial Interests, Personal, Speaker’s Bureau: Kite Pharma, Sanofi, Jazz Pharmaceuticals. K. McArthur, C. Martin, H. Namini, E. Ostertag: Financial Interests, Personal and Institutional, Full or part-time Employment: Poseida Therapeutics. M. Spear: Financial Interests, Personal, Stocks/Shares: Poseida Therapeutics; Financial Interests, Personal and Institutional, Full or part-time Employment: Denovo Biopharma. E. Christie: Financial Interests, Personal and Institutional, Full or part-time Employment: Poseida Therapeutics. R. Belani: Financial Interests, Personal and Institutional, Full or part-time Employment: Poseida Therapeutics; Financial Interests, Personal, Stocks/Shares: Amgen. M. Zhang: Financial Interests, Personal and Institutional, Full or part-time Employment: Poseida Therapeutics; Financial Interests, Personal, Stocks/Shares: BeiGene. S. Cranert, J. Coronella, D. Shedlock: Financial Interests, Personal and Institutional, Full or part-time Employment: Poseida Therapeutics. C. Costello: Financial Interests, Personal and Institutional, Principal Investigator: Poseida Therapeutics; Financial Interests, Personal and Institutional, Research Grant: BMS, Takeda, Janssen, Pfizer.

Collapse
Poster Display Poster Display session

48P - Phase II trial on vaccination with Autologous Dendritic cells loaded with Autologous Tumour homogenate in resected Glioblastoma (COMBI-GVAX): clinical results of the first step. (ID 147)

Presentation Number
48P
Lecture Time
12:30 - 12:30
Speakers
  • Laura Ridolfi (Meldola, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Glioblastoma (GBM) is a poor prognosis malignant grade IV glioma. After surgical resection, standard therapy consists of concomitant radiotherapy (RT) and temozolomide (TMZ) followed by TMZ alone. Multiple phase I/II trials and at least 3 meta-analysis showed improved survival (OS) and progression free survival (PFS) with dendritic cell (DC) vaccination in high-grade gliomas (HGGs) patients (pts). In those developing antitumor immunity, DC vaccine increases the amount of intratumoral activated cytotoxic T lymphocytes and decreases the number of FoxP3 positive regulatory T cells. Based on these data we have developed a phase II study with DC vaccine concomitant to standard RT-CT in pts undergoing radical surgery for GBM.

Methods

This is a single-arm, monocentric, phase II trial evaluating progression free-survival (PFS) and safety of a DC vaccination integrated to standard therapy in resected GBM. All pts receive a DC vaccine loaded with autologous tumor homogenate for up to one year. The vaccine administration starts at the end of the RT-CT (Induction Phase) and then is alternated to TMZ (Maintenance Phase). Primary end points are PFS and safety, among secondary end points the in vitro (Elispot, Plasma Cytokines, Tumor tissue analysis) and in vivo (DTH skin test) immune response biomarkers are evaluated. A Simon's two-stage design has been used for the sample size calculation. In the first stage, 9 pts will be accrued and a total of 28 pts will be enrolled.

Results

The first 9 pts have been enrolled since October 2021, 4 females and 5 males with a median age of 58 years. Four pts had no MGMT methylation. Eight out of 9 pts concluded the induction phase and 1 is ongoing. To date 4 pts have progressed with a median PFS from the date of diagnosis of 7.5 months (range 5-11). DTH test became positive in 4 out of 7 evaluable pts. No gr3-4 vaccine related toxicities have been observed and gr1-2 toxicities were mostly due to local skin reactions.

Conclusions

This combination therapy seems very well tolerated. The 2 end points of the first step have been reached so the study will proceed to enrol the remaining 19 pts.

Clinical trial identification

EudraCT 2020-003755-15.

Legal entity responsible for the study

Ridolfi Laura.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

49P - Exploiting multi-omic integrated data from DC-vaccinated melanoma patients for the generation of an advanced adoptive T cell therapy (ID 148)

Presentation Number
49P
Lecture Time
12:30 - 12:30
Speakers
  • Jenny Bulgarelli (Meldola, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Dendritic Cells (DCs) account for the best adaptive immune stimulators promoting their usage as advanced therapy medicinal products (ATMP). Despite the well tolerated profile and the ability to induce anti-tumor immunity, DC vaccine has nowadays failed to translate into prolonged long-term clinical benefit, requiring an update of its therapeutic application.

Methods

We conducted a retrospective study correlating clinical and multiple biologic data with the aim to define biomarkers of DC vaccine clinical activity and to exploit its capacity to expand peripheral autologous tumor-specific effector cells. Immunohistochemistry on pre and post FFPE samples, proteomic analysis on plasma and tumor homogenate, multiparametric flow cytometry on the starting apheresis and on DCs, RNAseq and scRNAseq on peripheral monocytes and manufactured DCs respectively, were applied.

Results

Analysis of post-vaccine biopsies highlighted the vaccine ability to induce intratumoral CD8 up-regulation (p=0.0195). The presence of multiple checkpoint molecules was a common denominator of our ATMP (PD-L1 90,1%-98,9%; PD-L2 94,9%-%99,5; VISTA 21,9%-%39,3; TIM-3 17%-59,4%; B7-H3 86,3%-98,8%). Intriguingly, high dimensional analysis of the DC final product revealed the coexistence of B cell clusters (CD19, MS4A1), higher (p=0.0286) in patients displaying clinical response. Of note, GMPc DCs induced a sustained expression of CD137 in co-cultured CD8+ T cells, and anti-PD1 addition enhanced the fraction of CD137+ T cells.

Conclusions

In vivo mechanisms of adaptive immune resistance (e.g. tumoral PDL1 upregulation p=0.0353) as well as the observed expression of checkpoint molecules by our ATMP support the clinical investigation of DC/ICI combination. The association of B cells with tumor response has to be deepened and the B cell frequency assessed also in pretreatment blood samples. Of note, the expansion of bona fide antigen-reactive CD137+ T cells resulting from the designed autologous DC ex vivo platform could translate into the manufacturing of a novel GMPc T cell product.

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 Poster Display session

50P - Enhancing TIL and NK cells adoptive therapies with an engineered oncolytic adenovirus encoding a human vIL-2 cytokine for the treatment of human ovarian cancer (ID 149)

Presentation Number
50P
Lecture Time
12:30 - 12:30
Speakers
  • Dafne C. Quixabeira (Helsinki, Finland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Tumor-infiltrating lymphocytes (TILs) and natural killer (NK) adoptive cell therapies have shown promising results in advanced-stage melanoma and haematological malignancies, respectively. However, their limited persistence in vivo in absence of an exogenous source of IL-2, and migration to neoplastic sites have impaired their effectiveness in immunosuppressive tumor microenvironments, such as ovarian cancer. Here, we propose the use of an engineered oncolytic adenovirus encoding a vIL-2 cytokine, Ad5/3-E2F-d24-vIL2 (vIL-2 virus), to improve said cell therapies. Oncolytic adenoviruses are immunogenic agents that lyse infected cancer cells and recruit immune cells to the neoplastic site. Moreover, the vIL-2 virus continuously expresses a vIL-2 cytokine that preferentially stimulates effector lymphocyte proliferation over T regulatory cells.

Methods

Fragments of resected human ovarian cancer tumors were received and processed into single-cell suspensions. Autologous TILs were expanded from said sample fragments, while PBMC cells from healthy donors were used as the source of allogeneic NK cells. To evaluate cancer cell killing, ovarian cancer ex vivo tumor cultures were co-cultured either with autologous TILs or with allogeneic NK cells in the presence or absence of the vIL-2 virus. Additionally, in vivo combination therapies efficacy was assessed with a patient-derived xenograft (PDX) ovarian cancer model. Immune cell profiling was performed following patient sample co-cultures and PDX tumor treatments.

Results

The addition of vIL-2 virus to the ovarian cancer ex vivo tumor cultures improved both TIL and NK cell therapies efficacy in vitro. Similarly, significantly better tumor control was achieved when the vIL-2 virus was given in conjunction with cell therapies compared to their respective controls. Mechanistically, vIL-2 virus treatment enhanced cell cytotoxicity of adoptively transferred TILs and NK cells in PDX tumors, and in ovarian cancer tumor cultures.

Conclusions

Ad5/3-E2F-d24-vIL2 virus treatment seems to be a promising immunotherapeutic candidate to improve the response of adoptive cell therapies in human immunosuppressive solid tumors.

Legal entity responsible for the study

Cancer Gene Therapy Group (CGTG), Translational Immunology Research Program, University of Helsinki.

Funding

TILT Biotherapeutics.

Disclosure

J.M. Santos, J. Clubb, R. Havunen, V. Cervera-Carrascon: Financial Interests, Personal and Institutional, Stocks/Shares: TILT Biotherapeutics. A. Hemminki: Financial Interests, Personal and Institutional, Stocks/Shares: TILT Biotherapeutics; Financial Interests, Personal, Stocks/Shares: Targovax ASA. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

51P - Immunological analysis of blood from patients with solid tumors treated with TILT-123, an oncolytic adenovirus encoding for tumor necrosis factor alpha (TNFa) and interleukin 2 (IL-2) (ID 150)

Presentation Number
51P
Lecture Time
12:30 - 12:30
Speakers
  • Santeri Pakola (Helsinki, Finland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

In recent years, cancer therapy has witnessed the successful development and clinical implementation of a wide array of tools to fight cancer, including, for example, monoclonal antibodies and adoptive cell therapies (ACT). Immunotherapies employing such tools to reinvigorate T-cells continue to change patient care by providing durable benefit in patients with advanced solid tumors. The latter is offset, however, by a large share of patients that present little response to immunotherapy, in part due to T-cell dysfunction. To overcome this issue, we constructed a chimeric oncolytic adenovirus (TILT-123; Ad5/3-E2F-D24-TNFa-IRES-IL2) capable of expressing tumor necrosis factor alpha (TNFa) and interleukin-2 (IL-2). The use of this agent in preclinical cancer models led to complete responses and favorable immunological effects, both as monotherapy or, in combination with immune checkpoint inhibitors and ACT using tumor-infiltrating lymphocytes (TIL) or chimeric antigen receptor T-cells.

Methods

Blood from patients enrolled in TILT-T115, a clinical study using TILT-123 as monotherapy in patients with injectable advanced solid tumors (NCT04695327), was collected at screening and during treatment, and peripheral blood mononuclear cells (PBMCs) and serum were extracted. Immunological effects were evaluated by detecting immune cell populations in PBMCs by flow cytometry, and by detecting cytokines in blood serum by a targeted multiplexed immuno-assay.

Results

Preliminary immunological data points at immune effects both in PBMCs and blood serum of patients undergoing treatment. Immunological data (e.g. flow cytometry and/or cytokine data) will be presented.

Conclusions

TILT-123 can induce systemic immunological effects supporting the continued development of the agent as cancer immunotherapy.

Clinical trial identification

NCT04695327 (first posted January 5, 2021).

Legal entity responsible for the study

TILT Biotherapeutics Ltd.

Funding

TILT Biotherapeutics Ltd.

Disclosure

J.M. Santos, V. Cervera-Carrascon, C. Kistler: Financial Interests, Institutional, Full or part-time Employment: TILT Biotherapeutics Ltd; Financial Interests, Personal, Stocks/Shares: TILT Biotherapeutics Ltd. S. Sorsa: Financial Interests, Personal, Stocks/Shares: TILT Biotherapeutics Ltd; Financial Interests, Institutional, Full or part-time Employment: TILT Biotherapeutics Ltd. R. Havunen: Financial Interests, Institutional, Full or part-time Employment: TILT Biotherapeutics Ltd. A. Hemminki: Financial Interests, Personal, Stocks/Shares: Targovax, TILT Biotherapeutics Ltd; Financial Interests, Institutional, Full or part-time Employment, CEO: TILT Biotherapeutics Ltd; Financial Interests, Institutional, Member of the Board of Directors: TILT Biotherapeutics Ltd. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

52P - MUC1 targeted immunotherapy with an oncolytic adenovirus coding for a bispecific T cell engager (ID 151)

Presentation Number
52P
Lecture Time
12:30 - 12:30
Speakers
  • Saru Basnet (Helsinki, Finland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Bispecific T cell engager (BsTe) is a fusion recombinant protein comprised of two single–chain variable fragments with dual specificity for a tumor–associated antigen (TAA) and T cell receptor (usually CD3ε). Immunotherapy with BsTe has shown efficacy in patients with hematologic malignancies and uveal melanoma. However, the antitumor efficacy of BsTe in most solid tumors has been limited due to their short serum half-life and insufficient tumor concentration. We designed a novel serotype 5/3 oncolytic adenovirus encoding for a BsTe cross-linking Mucin1 (MUC1) to CD3, Ad5/3–E2F–d24–aMUC1aCD3. The BsTe (aMUC1aCD3) is designed for the treatment of human solid tumors, where the BsTe links CD3 molecules on the surface of T cells and MUC1 on the target cancer cells.

Methods

Cell viability assays were used to assess the oncolytic potential of the novel Ad5/3–E2F–d24–aMUC1aCD3 construct. The functionality of virus-derived aMUC1aCD3 was evaluated in vitro using T cell activation, proliferation and cancer cell killing assays. The efficacy of the aMUC1aCD3 coding virus was investigated in vivo using a humanized MUC1 expressing lung cancer xenograft mouse model.

Results

The addition of aMUC1aCD3 transgenes did not compromise virus replication capacity. When Ad5/3–E2F–d24–aMUC1aCD3 is combined with allogenic T cells, rapid tumor cell lysis upon infection was observed using different cancer cell lines. TILT-321 infected cells secreted functional aMUC1aCD3 engagers as evidenced by increased T cell activity and competitive binding to MUC1+ cells. Ad5/3–E2F–d24–aMUC1aCD3 treatment also led to effective anti–tumor efficacy in vivo in a human MUC1 expressing lung cancer xenograft model. This response was associated with increased intratumoral T cell activation mediated by the aMUC1aCD3-armed adenovirus.

Conclusions

This study provides proof-of-concept for an effective strategy to overcome some of the limitations of recombinant BsTe delivery in the treatment of solid tumors. The proposed technology could be beneficial for the treatment of solid tumors preferentially those expressing MUC1.

Legal entity responsible for the study

The authors.

Funding

Ida-Montin Foundation, Orion Foundation, Minerva Foundation, Jane and Aatos Erkko Foundation.

Disclosure

J.M. Santos, V. Cervera-Carrascon, S.Sorsa: Financial Interests, Institutional, Full or part-time Employment: TILT Biotherapeutics Ltd; Financial Interests, Personal, Stocks/Shares: TILT Biotherapeutics Ltd. R. Havunen: Financial Interests, Institutional, Full or part-time Employment: TILT Biotherapeutics Ltd. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

53P - Combining cancer vaccines based on arenavirus vectors with 4-1BB (CD137) agonists enhances therapeutic efficacy in a non-inflamed tumor model (ID 152)

Presentation Number
53P
Lecture Time
12:30 - 12:30
Speakers
  • Judith Strauss (New York, NY, United States of America)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

One of the main reasons for the low efficiency of immunotherapy with immune checkpoint inhibitors in non-inflamed (cold) tumors is the lack of anti-tumoral T cell responses. Arenavirus-based cancer vaccines are ideally suited to induce tumor specific T cells but are hampered by the presence of immunosuppressive factors within the tumor microenvironment. Due to their capacity to promote activation, expansion, and effector function of activated T cells, we hypothesized that 4-1BB agonists could help to overcome intratumoral immune suppression by maintaining or even enhancing the functionality of vector-induced T cell responses.

Methods

To test this hypothesis, we initially combined replicating LCMV based vectors (artLCMV) encoding the tumor-associated antigens gp70 or Trp2 with agonistic 4-1BB monoclonal antibody (mAb).

Results

Single administration of vector and 4-1BB mAb prolonged tumor growth control and increased the number of complete responders in the cold B16.F10 tumor model. The combination induced a moderate increase of tumor-infiltrating CD8+ T cells, and positively affected expression of granzyme B, Ki67 and Bcl-XL in tumor-infiltrating antigen-specific T cells. Next, we generated artLCMV vectors co-expressing 4-1BBL together with gp70 or Trp2. In the immunogenic MC38 model, vector-encoded 4-1BBL did not further enhance the already high anti-tumor efficacy of artLCMV-gp70. In the cold B16.F10 model, however, 4-1BBL co-expression increased median survival time and number of complete responders, especially after intratumoral application. This contrasted with systemic application of vector and 4-1BB mAb, as described above. These data indicate that 4-1BB costimulation of T cells is most effective within the tumor, supporting the development of tumor-targeted 4-1BB agonists, which could be applied systemically, as intratumoral injections are not applicable to all cancer patients.

Conclusions

Overall, these experiments confirm the potential of combination therapies with arenavirus vectors and 4-1BB agonists, especially for the treatment of cold tumors, which lack functional T cells.

Legal entity responsible for the study

Hookipa Pharma Inc.

Funding

Hookipa Pharma Inc.

Disclosure

J. Strauss, D. Reckendorfer, K. Pojar, T. Pölzlbauer, S. Ahmadi-Erber, S. Schmidt, J. Raguz, J.C. Lampert, K.K. Orlinger, H. Lauterbach: Financial Interests, Personal, Full or part-time Employment: Hookipa Pharma Inc; Financial Interests, Personal, Stocks/Shares: Hookipa Pharma Inc. M. Habbeddine, M. Scheinost: Financial Interests, Personal, Stocks/Shares: Hookipa Pharma Inc.

Collapse
Poster Display Poster Display session

54P - Oncolytic virus combined PD-1 antibody Toripalimab in advanced lung cancer with liver metastases: An Early Stage, Single Arm, Study (TROJAN 2201) (ID 153)

Presentation Number
54P
Lecture Time
12:30 - 12:30
Speakers
  • Jing Yang (Shanghai, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The efficacy of checkpoint inhibitors are far from satisfied in lung cancer patients with liver metastases. Oncolytic virus can enhance the efficacy of PD-1/PD-L1 antibody in vitro/vivo. In this study, we evaluate the safety and efficacy of oncolytic virus H101 plus PD-1 antibody Toripalimab for liver metastasis-lung cancer advanced patients who have progressed and failed after EGFR-TKIs, chemotherapy or checkpoint inhibitors treatment.

Methods

The patients refractory to previous several lines of standard treatment were injected with recombinant human adenovirus 5 (1012 vp or 2×1012vp) locally for liver metastases plus toripalimab (3mg/kg) systemic therapy every 2 weeks until progression or intolerable toxicity. The two primary end points were investigator-assessed safety and objective response rate (ORR) and the secondary end points included progression-free survival (PFS) and disease control rate (DCR). Efficacy was assessed every 2 months by investigators according to mRECIST v1.1 criteria. Blood samples were collected from patients prospectively.

Results

From January 26, 2021 to July 15, 2022, ten patients were enrolled and received at least 1 cycle of the combination regimen. The most common treatment-related adverse events (TRAEs) were grade 1-2 fever, which was occurred in 9 (90%) of 10 patients. No deaths and other SAE were judged to be treatment-related. Six among the 10 patients received at least 3 cycles of the combination regimen and 5 were evaluable for efficacy analyses as one patient was lost to follow-up. Two of five (40%) patients (one small cancer and one adenocarcinoma) acquired partial remission (PR) after 3 cycles of the combination therapy, including one patient remained remission for 11 months. Two patients (40%) (one small cancer and one adenocarcinoma) achieved stable disease (SD) and the other one adenocarcinoma patent (20%) had progressive diseases (PD).

Conclusions

Our results confirm that the combination of recombinant human adenovirus type 5 plus toripalimab has shown an acceptable safety profile and the preliminary efficacy in those who refractory to previous several lines of standard treatment, deserved further investigation in randomized trials.

Legal entity responsible for the study

The authors.

Funding

Shanghai Science and Technology Committee, Grant/Award Number: 18DZ1910102.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

55P - Mutations Localized at the Membrane Predict Immunotherapeutic Efficacy in Cancer Treatment (ID 154)

Presentation Number
55P
Lecture Time
12:30 - 12:30
Speakers
  • Zoe Goldberger (Montreal, QC, Canada)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

In the clinic, immune checkpoint immunotherapy (ICI) is used to re-activate immune reactions against tumor neoantigens, leading to striking remission in cancer patients’ tumors. However, complete or durable responses to ICI treatment only occur in a minority of patients. While the level of tumor mutational burden (TMB) can be used as a predictive marker for responsiveness, we questioned whether the subcellular localization of the neoantigens within the tumor cell additionally plays a role. Using 3 human datasets with 1722 patients treated with ICI, we previously highlighted that patients bearing a high proportion of tumor neoantigens at the membrane of cancer cells responded better to anti-PD1. To decipher underlying immunological mechanisms, we developed a melanoma mouse model that expresses membrane-bound or soluble antigens and analyzed local and systemic anti-tumor immune responses upon anti-PD1 immunotherapy.

Methods

We engineered B16F10 melanoma cells to express membrane-bound or soluble OVA and analyzed intratumoral immune cell infiltration upon implantation in C57BL6 mice. We then compared tumor growth upon anti-PD1 treatment in the wild-type mice or in mice depleted for specific immune cells population. In addition, we compared systemic responses upon tumor implantation via ex vivo antigen-specific restimulation of the splenocytes.

Results

We demonstrated that mice bearing tumors with membrane-bound OVA have increased local and systemic anti-tumor immune reactions compared to soluble OVA, which rendered these tumors highly susceptible to ICI, leading to complete tumor rejection in mice. We additionally observed that tumor rejection was dependent on the level of antigen expressed in the cancer cells, with an increased rejection rate observed for the high dose membrane-bound OVA tumors compared to low ones. We surprisingly found that tumor rejection was independent of immunoglobulin G (IgG), of NK cells and of BatF3+ cross-presenting dendritic cells, and mostly relied on CD8+ T cells cytotoxicity, and partially on CD4+ T cells.

Conclusions

In this study, we show that the subcellular localization of tumor neoantigens plays an important role in the immunogenicity of tumors and subsequent responsiveness to anti-PD1 immunotherapy.

Legal entity responsible for the study

Priscilla S. Briquez, Jeffrey A. Hubbell.

Funding

This work was funded by the Chicago Immunoengineering Innovation Center of the University of Chicago and NIH R01CA219304 (to M.A.S).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

63P - Activity and safety of first-line treatments for advanced melanoma: a network meta-analysis (ID 155)

Presentation Number
63P
Lecture Time
12:30 - 12:30
Speakers
  • Andrea Boutros (Genova, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Treatment options for advanced melanoma have recently increased with the introduction of the anti-LAG3 and anti-PD-1 relatlimab/nivolumab combination. To date, ipilimumab/nivolumab is the benchmark of overall survival (OS), despite a high toxicity profile. Furthermore, in BRAF-mutant patients, BRAF/MEK inhibitors and the atezolizumab/vemurafenib/cobimetinib triplet are also available treatments, making the first-line therapy selection more complex. To address this issue, we conducted a systematic review and network meta-analysis of the available first-line treatment options in advanced melanoma.

Methods

Randomised clinical trials (RCTs) of previously untreated, advanced melanoma were included if at least one intervention arm contained a BRAF/MEK or an immune-checkpoint inhibitor (ICI). The aim was to indirectly compare the ICIs combinations ipilimumab/nivolumab and relatlimab/nivolumab, and these combinations with all the other first-line treatment options for advanced melanoma (irrespective of BRAF status) in terms of activity and safety. The co-primary endpoints were progression-free survival (PFS), overall response rate (ORR) and grade ≥3 treatment-related adverse events (TRAEs) rate, defined according to Common Terminology Criteria for Adverse Events (CTCAE).

Results

A total of 9070 patients treated in 18 first-line clinical trials of metastatic melanoma were included in the network meta-analysis. No difference in PFS nor ORR between ipilimumab/nivolumab and relatlimab/nivolumab was observed (HR=0.99 [95%CI 0.75 – 1.31] and RR=0.99 [95%CI 0.78 – 1.27], respectively). The PD-(L)1/BRAF/MEK inhibitors triplet was superior to ipilimumab/nivolumab in terms of PFS (HR=0.56 [95%CI 0.37 – 0.84]) and ORR (RR=3.07 [95%CI 1.61 – 5.85]). Relatlimab/nivolumab showed a trend to a lower risk of grade ≥3 TRAEs (RR=0.71 [95%CI 0.30 – 1.67]) compared to ipilimumab/nivolumab, which showed the highest probability of grade ≥3 TRAEs.

Conclusions

Relatlimab/nivolumab showed similar PFS and ORR compared to ipilimumab/nivolumab, with a trend for a better safety profile. The PD-(L)1/BRAF/MEK inhibitors triplet combinations showed the highest probability to achieve better PFS and ORR.

Clinical trial identification

PROSPERO registration number: CRD42022303279.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

L. Del Mastro: Financial Interests, Personal, Invited Speaker, Educational meeting: Novartis, Symposia, Andromeda E20, Vyvamed srl; Financial Interests, Personal, Invited Speaker, Lecture: Ipsen; Financial Interests, Personal, Advisory Board, Her2+ and TN breast cancer: Roche; Financial Interests, Personal, Invited Speaker, Consultancy for TNBC text: Roche; Financial Interests, Personal, Advisory Board, denosumab: Amgen; Financial Interests, Personal, Advisory Board, Early and metastatic BC: Eli Lilly; Financial Interests, Personal, Invited Speaker, CDK4-6 inhibitors: Eli Lilly; Financial Interests, Personal, Advisory Board, tucatinib: Seagen Int; Financial Interests, Personal, Advisory Board, Oncotype dx: Exact sciences, Havas life; Financial Interests, Personal, Advisory Board, Neratinib: Pierre Fabre; Financial Interests, Personal, Invited Speaker, Internal training: MSD; Financial Interests, Personal, Invited Speaker, Educational meetings: Accademia Nazionale Medicina; Financial Interests, Personal, Invited Speaker, Author for BC text: Pensiero Scientifico Editore; Financial Interests, Personal, Advisory Board, Breast cancer: Uvet; Financial Interests, Personal, Other, Author slide kits and interviews: Think2it; Financial Interests, Personal, Advisory Board, Palbociclib: Pfizer; Financial Interests, Personal, Invited Speaker, Breast cancer: Aristea, Meeting SrL; Financial Interests, Personal, Other, Author slide kits: Forum service; Financial Interests, Personal, Other, Author text about biosimilars: Edizioni Minerva Medica; Financial Interests, Personal, Other, consultant: Kardo srl; Financial Interests, Personal, Invited Speaker, Breast cancer meetings: Delphi international, Over srl; Financial Interests, Personal, Invited Speaker: Prex Srl, Editree; Financial Interests, Personal, Advisory Board: Uvet, Collage SpA, Daiichi Sankyo, AstraZeneca; Financial Interests, Personal, Other, Interview: Infomedica srl; Financial Interests, Personal, Other, Consultant: Sharing progress in cancer care - Switzerland; Financial Interests, Institutional, Funding, National coordinating PI: Roche; Financial Interests, Institutional, Funding, Local PI: AstraZeneca, Roche, Eli Lilly, Daiichi Sankyo, Novella Clinical, Novartis; Non-Financial Interests, Institutional, Product Samples, Genomic Test: FoundationOne. M. Lambertini: Financial Interests, Personal, Advisory Board: Roche, AstraZeneca, Lilly, Novartis, Exact Sciences, MSD, Seagen, Gilead; Financial Interests, Personal, Invited Speaker: Pfizer, Takeda, Sandoz, Ipsen, Libbs, Knight. F. Spagnolo: Financial Interests, Personal, Invited Speaker: Sanofi Genzyme, Roche, BMS, Novartis, Merck, Sunpharma, MSD, Pierre Fabre; Financial Interests, Personal, Advisory Board: Novartis, Philogen, Sunpharma, MSD. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

64P - Safety and efficacy of immune checkpoint inhibitors (ICIs) in patients (pts) with solid tumors and cardiac metastases (ID 156)

Presentation Number
64P
Lecture Time
12:30 - 12:30
Speakers
  • Amin H. Nassar (New Haven, CT, United States of America)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Incidence of cardiac metastasis (mets) is rising among pts with cancer. Data on the safety and clinical outcomes of ICIs on pts with cardiac mets is limited.

Methods

In an international multi-center retrospective study, pts with solid tumors and mets to the heart who had received ICIs were included. Immune-related adverse events (irAEs) were graded according to CTCAE v5.0. Objective response rates (ORR) were evaluated by RECIST when available. Overall survival (OS) and progression-free survival (PFS) were calculated from time of ICI initiation using the Kaplan-Meier Method.

Results

62 pts across 16 institutions were identified with a median follow-up of 38.4 months (mo). Melanoma (33.9%, n=21) and non-small cell lung cancer (NSCLC, 22.6%, n=14) were the most common cancers. Median age at ICI start was 65 (23-89) years and 29.0% (n=18) received combination therapy with anti-PD1/PD-L1 and anti-CTLA4, and 58.1% (n=36) received ICIs as 1st line. Most common locations of cardiac mets were the ventricles (38.7%, n=24) and atria (37.1%, n=23). At ICI initiation, 20.1% (n=13) had a cardiac thrombus. Median diameter of cardiac mass was 4 (0.5-9.0) cm. Cardiology referrals and cardiac MRIs were done on 37/62 (59.7%) and 28/62 (45.2%) pts, respectively. 33.9% had cardiac complications from cancer mets, including arterial/venous emboli (8.1%, n=5), tamponade and arrhythmias (each 6.5%, n=4). For NSCLC, ORR, PFS, and OS were 28.7% (11.7-54.6), 4.3 mo (1.5-11.2), and 9.9 mo (2.0-14.4), respectively. For melanoma, ORR, PFS, and OS were 38.1% (20.8-59.1), 7.5 mo (2.2-19.3), and 35.0 mo (5.4-62.9), respectively. Cardiac mass ORRs for NSCLC and melanoma were 21.4% (7.6-47.6) and 42.9% (24.5-63.5), respectively. iRAEs are shown in the table.

irAE leading to discontinuation 10 (16.1%)
Systemic steroids 17 (27.4%)
Any grade Grade ≥3
Any irAE 28 (45.2%) 13 (19.4%)
Cardiac 1 (1.6%) 0 (0%)
Dermatitis 8 (12.9%) 1 (3.2%)
Pneumonitis 2 (3.2%) 1 (1.6%)
Diarrhea 8 (12.9%) 3 (4.8%)
Endocrine 6 (9.7%) 1 (1.6%)
Mucositis 1 (1.6%) 0 (0%)
Neurologic 1 (1.6%) 1 (1.6%)
Hepatitis 1 (1.6%) 1 (1.6%)
Other 8 (12.9%) 2 (3.2%)

Conclusions

Cardiac mets are frequently associated with cardiac complications. ICIs have activity in cardiac mets and are safe.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

T.K. Choueiri: Financial Interests, Personal, Advisory Board, Advice on GU/RCC drugs: BMS, Pfizer, Merck, Exelixis, AstraZeneca; Financial Interests, Personal, Advisory Board, Advice on Onc drugs: Lilly, EMD Serono, Infinity; Financial Interests, Personal, Advisory Board, Advice on RCC drug: Calithera; Financial Interests, Personal, Invited Speaker, RCC drug: Ipsen; Financial Interests, Personal, Advisory Board, Advice on GU Onc drugs: Surface Oncology; Financial Interests, Personal, Other, Consultant on onc drugs: Analysis Group; Financial Interests, Personal, Invited Speaker, CME, ww2.peerview.com: Peerview; Financial Interests, Personal, Invited Speaker, CME, gotoper.com: PER; Financial Interests, Personal, Invited Speaker, CME, researchtopractice.com: ResearchToPractice; Financial Interests, Personal, Invited Speaker, National Association of Managed Care: NAMC; Financial Interests, Personal, Invited Speaker, ASCO-related event: ASCO-SITC; Financial Interests, Personal, Other, Grant review to Orien Network ($400): ORIEN; Financial Interests, Personal, Advisory Board, Advising oncology strategy: Aptitude Health; Financial Interests, Personal, Invited Speaker, Best of ASCO19 talk: Advent health; Financial Interests, Personal, Invited Speaker, Best of ESMO20 talk ($1000): UAE Society of Onc; Financial Interests, Personal, Invited Speaker, CME, mjhlifesciences.com (OncLive): MJH life sciences; Financial Interests, Personal, Invited Speaker, Grand Rounds: MDACC; Financial Interests, Personal, Invited Speaker, RCC webinar: Cancernet; Financial Interests, Personal, Invited Speaker, CME, Kidney Cancer Association ($1300): France Foundation; Financial Interests, Personal, Invited Speaker, CME, RCC: Springer, WebMed; Financial Interests, Personal, Invited Speaker, CME, ImmunoOncology in RCC: ASiM, CE; Financial Interests, Personal, Invited Speaker, CME, PodCast in RCC ($500.00): Caribou Publishing; Financial Interests, Personal, Invited Speaker, Reimbursement ($432.00): Kidney Cancer Association; Financial Interests, Personal, Other, member of the DSMB for clinical trial: Aravive; Financial Interests, Personal, Invited Speaker, unpaidhttps://www.accru.org/main/public/index.xhtml: ACCRU; Financial Interests, Personal, Invited Speaker, Unpaidhttps://kidneycan.org: KidneyCan; Financial Interests, Personal, Other, External Advisory Board Member: Gustave Roussy; Financial Interests, Personal, Stocks/Shares: Pionyr (not publicly traded), Tempest (publicly traded), Osel (not publicly traded), Nuscan ((not publicly traded)); Financial Interests, Personal, Royalties, For writing and updating chapters in GU Oncology: Up-To-Date online textbook; Financial Interests, Institutional, Funding, National Chair: BMS, Merck, Exelixis, AstraZeneca, Takeda, Tracon, Peloton; Financial Interests, Institutional, Invited Speaker: Roche, Surface Oncology, GSK; Financial Interests, Institutional, Funding, National co-chair: Pfizer, EMD-Serono; Financial Interests, Institutional, Funding, Chair of trial: Lilly; Financial Interests, Institutional, Funding, SC member: Eisai; Financial Interests, Institutional, Funding, National co-chair on 3 ongoing trials: Alliance Cooperative Group; Financial Interests, Institutional, Research Grant, for GU oncology translational research through IION program: BMS; Financial Interests, Institutional, Research Grant, for GU oncology translational research: Exelixis; Financial Interests, Institutional, Research Grant, For Health outcomes research: Roche; Financial Interests, Institutional, Invited Speaker, leads trials as PI: Nikang; Non-Financial Interests, Personal, Leadership Role, Co-Chair of the meeting, 2019-: Kidney Cancer Research Summit of KidneyCAN; Non-Financial Interests, Personal, Principal Investigator, Trial Global and National PI with GU Cancers, mostly Kidney Cancer: Multiple Academic and Industry entities; Non-Financial Interests, Personal, Other, Track Leader/Session chair/Speaker/Discussant: ASCO; Non-Financial Interests, Personal, Other, Speaker/Discussant/Track Leader: ESMO; Non-Financial Interests, Institutional, Other, Access to genomic database: Foundation Med, Guardant, Invitae; Non-Financial Interests, Personal, Other, Grants reviewers: AACR; Non-Financial Interests, Personal, Other, Reviewer of papers: Various journals (e.g. NEJM, Lancet, JCO); Non-Financial Interests, Personal, Other, Medical writing and editorial assistance support (e.g. ClinicalThinking, Envision Pharma Group, Fishawack Group of Companies, Health Interactions, Parexel, Oxford PharmaGenesis, pharmagenesis, and others). However, first draft frequently initiated by myself when I am 1st author: Medical Communication; Non-Financial Interests, Personal, Member: ASCO, AACR; Non-Financial Interests, Personal, Other, Political vote usually as “independent”, not a member of any political party. I am an issue voter: General US Politics; Other, Personal, Other, No financial interest. Institutional.Filed patents related to biomarkers of immune checkpoint blockers, and circulating free methylated DNA. No money made and some patents were abandoned: Filed patents; Other, Personal, Other, Employee at DFCI. Please see https://www.dana-farber.org/ for mission statement (non-profit hospital). I am also the past President of Medical Staff at DFCI 2015-2018: Dana-Farber Cancer Institute (DFCI); Other, Personal, Other, Professor at HMS, Please see https://hms.harvard.edu/ for mission statement (non-profit school): Harvard Medical School (HMS). All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

65P - Atezolizumab plus bevacizumab versus Lenvatinib for unresectable hepatocellular carcinoma: a large real life worldwide population (ID 157)

Presentation Number
65P
Lecture Time
12:30 - 12:30
Speakers
  • Margherita Rimini (Milan, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Atezolizumab plus bevacizumab and lenvatinib have not been compared in a randomized controlled trial. We conducted a retrospective multi-center study to compare the clinical efficacy and safety of lenvatinib and atezolizumab with bevacizumab as a first-line treatment for patients with unresectable HCC in the real-world scenario.

Methods

Clinical features of lenvatinib and atezolizumab plus bevacizumab patients were balanced through inverse probability of treatment weighting (IPTW) methodology, which weights patients' characteristics and measured outcomes of each patient in both treatment arms. Overall survival was the primary endpoint.

Results

The analysis included 1,341 patients who received lenvatinib, and 864 patients who received atezolizumab plus bevacizumab. After IPTW adjustment, atezolizumab plus bevacizumab did not show a survival advantage over lenvatinib HR 0.97 (p=0.739). OS was prolonged by atezolizumab plus bevacizumab over lenvatinib in viral patients (HR: 0.76; p=0.024). Conversely, OS was prolonged by lenvatinib in patients with NASH/NAFLD (HR: 1.88; p=0.014) In the IPTW-adjusted population, atezolizumab plus bevacizumab provided better safety profile for most of the recorded adverse events.

Conclusions

Our study did not identify any meaningful difference in overall survival between atezolizumab plus bevacizumab and lenvatinib. Although some hints are provided suggesting that patients with NASH/NAFLD might benefit more from lenvatinib therapy and patients with viral etiology more from atezolizumab plus bevacizumab.

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 Poster Display session

66P - Real-world data of first-line chemo-immunotherapy for patients with extensive stage SCLC: A multicentre experience from Switzerland and the UK (ID 158)

Presentation Number
66P
Lecture Time
12:30 - 12:30
Speakers
  • Laura Moliner Jimenez (Manchester, United Kingdom)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The addition of immunotherapy (either atezolizumab or durvalumab) to platinum-etoposide for patients with extensive stage SCLC (ES-SCLC) has been recently established as standard first-line treatment based on IMPOWER133 and CASPIAN trials. Yet, the efficacy and safety in a real-world setting remains unclear.

Methods

We retrospectively evaluated patients with ES-SCLC treated with either atezolizumab or durvalumab plus platinum-etoposide between October 2018 and October 2021 in ten centres in the UK and ten centres in Switzerland. Responses were assessed using RECIST v1.1 criteria. Median PFS and OS were analyzed by the Kaplan-Meier method.

Results

A total of 436 patients were included. Median age was 67 years, 228 (52.3%) were males, 209 patients (47.9%) were current and 203 (46.6%) former smokers. 63 patients (14.4%) had an ECOG performance status (PS) ≥2. 385 patients (88.3%) were initially diagnosed with ES-SCLC. Liver and brain metastases were diagnosed in 170 (39.0%) and 87 (20.0%) of patients, respectively. At the time of analysis, 284 patients (65.1%) had died. Most of the patients received atezolizumab (n=427, 97.9%) in combination with chemotherapy, 2.1% received durvalumab. Most patients (n=422, 96.8%) were treated with carboplatin/etoposide. Overall response rate was 71.8% (95%CI 67.3-76.0%) with a median duration of response of 3.5 months (95%CI 3.3-4.0). Median PFS and OS were 5.5 (95%CI 5.3-5.7) and 9.3 months (95%CI 8.4-10.4), respectively. Immune related adverse events (AEs) were seen in 21.8% of patients. 5 patients (1.1%) developed a grade 5 AE. 174 patients (39.9%) received a subsequent systemic therapy. Among patients with brain metastases at diagnosis, mPFS and mOS were 4.8 months (95%CI 4.4-5.5) and 8.6 (95%CI 6.9-10.8), respectively.

Conclusions

Data from our large series show shorter OS than what reported in the trials. Known poor prognostic factors (mainly ECOG PS ≥2 and brain metastases) were more common in our cohort of patients at baseline and may have determined a shorter OS. This is the largest real-world evidence dataset evaluating patients with ES-SCLC treated with first-line chemo-immunotherapy, and could help to optimise clinical management of these 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 Poster Display session

67P - Real Word Data for Atezolizumab plus Bevacizumab in unresectable Hepatocellular Carcinoma (ID 159)

Presentation Number
67P
Lecture Time
12:30 - 12:30
Speakers
  • MARA PERSANO (Cagliari, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Atezolizumab plus bevacizumab has been recently approved as new first-line standard of care for patients with unresectable hepatocellular carcinoma (HCC). We perform a real word study to evaluate the impact in term of outcome of the inclusion criteria from the IMbrave150 trial on safety and efficacy of treatment.

Methods

We enrolled patients treated with atezolizumab plus bevacizumab for unresectable HCC from 4 different countries. No specific inclusion and exclusion criteria were applied, except for the absence of previous systemic therapies for HCC. The whole population was split in two groups according to the concordance with the inclusion criteria as reported in the IMbrave150 trial in “IMbrave150-in” and “IMbrave150-out” patients, and survival outcomes in the two groups of patients have been evaluated.

Results

766 patients were enrolled in the study: 561/766 (73%) were included in the “IMbrave150-in” group, and 205/766 (27%) were included in the “IMbrave150-out” group. mOS and mPFS were 16.3 Vs 14.3 months (HR 0.48, 0.35–0.65; p < 0.0001] and 8.3 Vs 6.0 months (HR 0.79, 0.63–0.99; p = 0.0431) in “IMbrave150-in” and “IMbrave150-out” patients, respectively. Multivariate analysis confirmed that patients included in the “IMbrave150-in” group had significantly longer OS compared to patients included in the “IMbrave150-out” group (HR 0.76, 0.47-0.97; p=0.0195). In “IMbrave150-in” patients the ALBI grade was not significantly associated to OS, whereas in “IMbrave150-out” patients, those with an ALBI grade 1 reported a significant benefit in terms of OS compared to those with an ALBI grade 2 (16.7 Vs 5.9 months HR 4.40,2.40-8.08, p>0.0001). No statistically differences were reported in “IMbrave150-in” and “IMbrave150-out” groups in terms of safety profile.

Conclusions

The accordance with the inclusion criteria of the IMbrave150 trial positively impact prognosis of patients receiving Atezolizumab plus bevacizumab. Between patients who do not meet the inclusion criteria, those reporting an ALBI grade of 1 could benefit from treatment with the combination.

Legal entity responsible for the study

M. Rimini.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

69P - A Real-World Analysis of Treatment-Free survival for Advanced Melanoma Patients treated with First-Line Immune Checkpoint Inhibitors (ID 161)

Presentation Number
69P
Lecture Time
12:30 - 12:30
Speakers
  • Mehul Gupta (Calgary, AB, Canada)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Conventional time to event endpoints, such as overall survival and progression-free interval fail to reflect quality of life when characterizing outcomes with immune checkpoint inhibitors (ICI). Treatment-free survival (TFS) represents an alternative approach, to more accurately characterize the time free of systemic therapy, providing a more patient-centric view of ICI therapy. There remains a paucity of studies evaluating TFS outcomes in advanced melanoma patients receiving immunotherapy, especially in the non-clinical trial context. Here, we present the first real-world observational analysis of TFS outcomes for patients with advanced melanoma receiving first-line ICI therapy.

Methods

Demographic, clinical and survival characteristics of patients with advanced melanoma receiving first-line ICI therapy (n=316) was collected retrospectively from a multi-center observational cohort study in Alberta, Canada. Treatment-free survival was defined as the difference in the 36-month restricted mean survival time between 2 survival endpoints, time to ICI cessation or censored at last follow up, and time to subsequent systemic anti-cancer therapy, death or censored at last follow up.

Results

The restricted mean TFS was longer for nivolumab plus ipilimumab (mean 12.38 months, 95% CI 8.79-16.00) when compared to nivolumab (mean 8.93 months 95% CI 4.38-13.47) and pembrolizumab (mean 11.14 months, 95% CI 8.47, 13.80). During the 36 month follow up, interval patients treated with Nivolumab plus ipilimumab spent 34.4% of their time off systemic anticancer treatments, compared to the 30.9% and 24.8% of their time for the pembrolizumab and nivolumab treatment groups respectively.

Conclusions

TFS represents a patient-centric, informative endpoint for advanced melanoma patients treated with first-line ICI therapy. Of note, patients treated with combination nivolumab plus ipilimumab spent more time alive and free from systemic anti-cancer therapy than those treated with anti-PD-1 monotherapy alone. This information may inform therapeutic decision making, given the number of treatment choices available to clinicians and patients in this context.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

D.Y.C. Heng: Non-Financial Interests, Personal, Advisory Role: Pfizer, Novartis, Bristol Myers Squib, Janssen, Astellas, Ipsen, Eisai, Merck; Financial Interests, Personal, Research Grant: Pfizer, Novartis, Exelixis, Bristol Myers Squibb, Ipsen. T. Cheng: Financial Interests, Personal, Research Grant: Pfizer. V. Navani: Non-Financial Interests, Personal, Advisory Role: Kyowa Kirin, Ipsos. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

70P - A retrospective real-world study of anlotinib plus PD-1 inhibitors in advanced esophageal squamous cell carcinoma (ESCC) previously treated with immune checkpoint inhibitors (ICIs). (ID 162)

Presentation Number
70P
Lecture Time
12:30 - 12:30
Speakers
  • Jun Liu (Shanghai, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immune checkpoint inhibitors (ICIs) combined with chemotherapy have become standard first-line treatments for advanced esophageal cancer. However, effective therapeutic strategies are limited after failure of immunotherapy. The combination of ICIs and anti-angiogenic agents may produce synergistic effects. Anlotinib, a multi-targeted anti-angiogenic agent, has been approved with good tolerance and efficacy in advanced ESCC. Therefore, we aimed to retrospectively analyze the efficacy and safety of anlotinib plus PD-1 inhibitors in advanced ESCC previously treated with ICIs in the real world (NCT 04984096).

Methods

We retrospectively analyzed the effects of anlotinib plus PD-1 inhibitors with unresectable locally advanced or recurrent/metastatic ESCC and must have received anti–PD-1 or anti–PD-L1 therapy previously. Efficacy was assessed using the Response Evaluation Criteria in Solid Tumors version 1.1. Baseline characteristics and adverse events (AEs) were recorded throughout the entire study.

Results

Between July 2020 to March 2022, 29 eligible patients were included in the final analysis. At data cutoff (July 3, 2022), the ORR was 31.0 % and the DCR was 86.2%, including 9 patients with partial response (PR), 16 patients with stable disease (SD) and 4 patients with progression disease (PD). 8 (27.6%) of 29 patients had received first-line treatment, 20 patients (69.0%) had received second-line treatment and 1 (3%) patient received three lines system chemotherapy. The median PFS was 5.33 months (95% CI 4.28-6.38) and the median OS was 10.38 months (95% CI 6.26-14.48). Any treatment-related adverse events (TRAEs) occurred in all patients, anemia and lymphopenia were the most two common TRAEs, only 2 patients (6.9%) with grade 3-4 TRAEs of lymphocytopenia. All AEs could be relieved after symptomatic treatment. No treatment-related deaths occurred.

Conclusions

Anlotinib plus PD-1 inhibitors showed encouraging anti-tumor activity and manageable safety in advanced ESCC previously treated with ICIs, providing a feasible and well-tolerated treatment option for this patient population. The data further need to be validated in prospective study.

Clinical trial identification

NCT 04984096.

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 Poster Display session

71P - Real-world Treatment Patterns and Clinical Outcomes for patients received Sintilimab (Sint) in First-line Treatment of Advanced Non-small Cell Lung Cancer (NSCLC) in China: A Multicenter Retrospective Observational Study (ID 163)

Presentation Number
71P
Lecture Time
12:30 - 12:30
Speakers
  • Yan Yu (Harbin, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Sint has been used to treat patients (pts) with advanced NSCLC. This observational study aims to describe the real-world use of Sint and to explore its real-world effectiveness in the first-line (1L) treatment of advanced NSCLC.

Methods

Pts with advanced NSCLC initiating 1L therapy with Sint between Aug 1st 2018 and Aug 1st 2020 in 4 tertiary hospitals in mainland China were included. Data in demographic and clinical characteristics, treatment patterns and outcomes were retrospectively collected through medical records. Descriptive analysis was used to describe the baseline characteristics and treatment patterns. Kaplan-Meier method was used to calculate the median progression-free survival (mPFS).

Results

102 pts (85.3% men) were included with mean age of 63.9±9.5 years (median follow-up time: 4.8 months (mo) (range 0.0-32.0)). The most commonly-used treatment regimen was Sint + platinum (Pt) + taxanes (56.0%) for squamous NSCLC (sqNSCLC) and Sint + Pt + pemetrexed (38.5%) for non-squamous NSCLC (nsqNSCLC) (Table). The mPFS was 15.8 mo (95% CI: 11.5-NA) and 12.0 mo (95% CI: 6.3-NA) respectively for sqNSCLC and nsqNSCLC pts. The mPFS was 17.0 mo (95% CI 11.5-NA) in sqNSCLC pts using Sint + Pt + taxanes as 1L therapy and 7.2 mo (95% CI: 0.7-NA) in nsqNSCLC pts. The ORR was 57.1% and 53.6% respectively for sqNSCLC and nsqNSCLC pts.

Baseline characteristics and treatment pattern of NSCLC

Total (N=102) sqNSCLC (N=50) nsqNSCLC (N=52)
Age (mean±SD), years 63.9±9.5 64.9±7.0 63.0±11.4
Male (%) 85.3 88.0 82.7
Stage at first diagnosis (%)
III 21.6 34.0 9.6
IV 42.2 28.0 55.8
Median of Sint cycle number 4.0 3.0 5.0
Treatment pattern (%)
Sint monotherapy 6.9 4.0 9.6
Sint + Pt + taxanes 36.3 56.0 17.3
Sint + Pt + pemetrexed 19.6 0.0 38.5
Sint + Pt + gemcitabine 5.9 12.0 0.0
Sint + Pt + anti-VEGF therapy + pemetrexed 5.9 0.0 11.5
Sint + others 25.5 28.0 23.1

Conclusions

For sqNSCLC pts with Sint as 1L therapy, taxanes and gemcitabine are more preferred, while for nsqNSCLC pts, pemetrexed is more often combined. The real-world effectiveness of 1L treatment of Sint for advanced NSCLC pts is promising from the study.

Legal entity responsible for the study

Eli Lilly China.

Funding

Lilly (Shanghai) Management Co., Ltd.

Disclosure

L. Zhang: Financial Interests, Personal and Institutional, Full or part-time Employment, employees of Lilly China and own Lilly stock shares: Eli Lilly China; Financial Interests, Personal and Institutional, Stocks/Shares, employees of Lilly China and own Lilly stock shares: Eli Lilly China. L. Liu: Financial Interests, Personal and Institutional, Full or part-time Employment, employees of Lilly China and own Lilly stock shares: Eli Lilly China; Financial Interests, Personal and Institutional, Stocks/Shares, employees of Lilly China and own Lilly stock shares: Eli Lilly China. J. Li: Financial Interests, Personal and Institutional, Full or part-time Employment, employees of Lilly China and own Lilly stock shares: Eli Lilly China; Financial Interests, Personal and Institutional, Stocks/Shares, employees of Lilly China and own Lilly stock shares: Eli Lilly China. W. Ding: Financial Interests, Institutional, Advisory Role, received Lilly’s sponsorship: Happy Life Technology. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

72P - Treatments response in non-small cell lung cancer patients according to BRCA status on liquid biopsy: a retrospective analysis (ID 164)

Presentation Number
72P
Lecture Time
12:30 - 12:30
Speakers
  • Leonardo Provenzano (Milan, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

NSCLC has to date a poor prognosis with life expectancy at five year of 26%. The development of precision medicine is going to improve the prognosis, but most of alterations are still lacking of clinical significance. Somatic BRCA alterations, is reported to be quite frequent in NSCLC, with a prevalence of 5.3-6.6%. However, no specific study to date specifically evaluated its prognostic and predictive role in this population of patients.

Methods

We evaluated BRCA status of aNSCLC patients undergone liquid biopsy at Istituto Nazionale dei Tumori in Milan and Luigi Vanvitelli Hospital in Naples. Guardant360CDx® was used and mutations pathogenicity were interpreted based on the COSMIC database. Only mutations interpreted as pathogenic were considered. Chi square and Fisher tests were used to match clinical characteristics with BRCA status and survival outcomes were analysed with Cox model.

Results

307 patients had BRCA status available on liquid biopsy, of which 33 (10.7%) had a pathogenic mutation in BRCA1/2 genes. BRCAm patients were older, more frequently male and smoker, had a higher PS and higher PDL1 expression on tissue samples; however, none of these features was significantly associated except for PS. Among patients treated with first-line platinum-based CT, a trend for better PFS was observed among BRCAm patients vs wt, without reaching statistical significance (HR 0.76, CI 95% 0.43-1.35, p=0.354). An increased platinum ORR was observed in BRCA mut vs wt patients (47.6% vs 34.0%, p=0.012). Conversely, among patients treated with any-line IOT, a worse PFS was observed among BRCAm patients vs wt (mPFS 3.5 vs 7.2 m; HR 2.14, CI 95% 1.35-3.40; p=0.001) and numerically reduced ORR was also observed (13.0% vs 26.8%, p=0.192). Finally, BRCA mutations seem to do not have impact on OS (BRCAm vs wt: 28.2 vs 28.8 m; HR 1.17, CI 95% 0.76-1.79; p=0.482).

Conclusions

In our retrospective cohort of aNSCLC patients, presence of a pathogenic mutation in BRCA1/2 genes resulted in reduced benefit to IOT, without affecting overall survival, probably due of the counterbalance effect of the increased sensitivity to platinum-CT. These data could be used to guide therapeutic choices and therefore deserve to be confirmed in a prospective cohort.

Legal entity responsible for the study

Fondazione IRCCS Istituto Nazionale dei Tumori, Milan.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

73P - A randomized phase II study of neoadjuvant immunotherapy or immunochemotherapy in locally advanced oral squamous cell carcinoma (ID 165)

Presentation Number
73P
Lecture Time
12:30 - 12:30
Speakers
  • Gang Chen (Wuhan, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

To evaluate the therapeutic efficacy and safety of neoadjuvant PD-1 inhibitor camrelizumab combined with TPF induction chemotherapy versus camrelizumab alone in patients with locally advanced resectable oral squamous cell carcinoma (OSCC). This study also aims to build predictive models for treatment decision based on a multivariable logistic regression analysis of multi-dimensional data.

Methods

A phase II study (NCT04649476) was conducted. Untreated stage III or IVA (UICC 8th edition) OSCC patients received neoadjuvant therapy with three 2-week cycles of camrelizumab (200 mg, d1, q2w) (arm A, n=34) or three 2-week cycles of camrelizumab (200 mg, d1, q2w) combined with two 3-week cycles of TPF induction chemotherapy [docetaxel (T) 75 mg/m2, cisplatin (P) 75 mg/m2 and 750 mg/m2 5-fluorouracil (F)] (arm B, n=34), followed by surgery and adjuvant radiotherapy. The primary end point was pathological response rate.

Results

Of the 68 patients enrolled into this trial, 60 completed the full treatment protocol. The pathological response rate in arm B was 83.4%, which was much higher than arm A (16.7%). The pathological complete response (pCR) rate was 30.0% in arm B, while no patient with pCR was observed in arm A. The median follow-up duration was 9.7 months, and the 1-year event-free survival (EFS) of arm A and arm B were 66.7% and 96.7%, respectively. Grade 3-5 neoadjuvant therapy-related adverse events were occurred in 7 patients from arm B and one patient from arm A. A decision tree, mainly based on lymph node metastasis, tumor site, and infiltrated immunocytes in biopsy tissue, was developed to screen patients with high responsivity and low adverse effects.

Pathological response rate

Pathological response Arm A Camrelizumab (n = 30) Arm B Camrelizumab + TPF (n = 30)
No. % No. %
pCR (0% viable tumor) 0 0 9 30.0
pMPR (≤10% viable tumor) 5 16.7 16 53.4
pPR (11-50% viable tumor) 4 13.3 3 10.0
pNR (51%-100% viable tumor) 21 70.0 2 6.6
Pathological response rate (pCR+pMPR) [95% CI interval] 5 (16.7) [3.3 – 30.0] 25 (83.4) [70.0 – 96.7]

Conclusions

Neoadjuvant camrelizumab plus TPF induction chemotherapy for locally advanced resectable OSCC showed high pathological response rate, with acceptable adverse effects. Newly developed predictive models may benefit precise treatment decision for OSCC patients in clinical practice.

Clinical trial identification

NCT04649476.

Legal entity responsible for the study

Gang Chen.

Funding

Jiangsu Hengrui Pharmaceuticals Co., Ltd.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

74P - The safety, tolerability, and preliminary antitumor activity of sitravatinib plus tislelizumab in patients (pts) with locally recurrent or metastatic triple negative breast cancer (TNBC): a multi-cohort, phase II trial (ID 166)

Presentation Number
74P
Lecture Time
12:30 - 12:30
Speakers
  • Linda Liu (Shanghai, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

This is a multi-cohort, phase II trial to evaluate the safety and preliminary antitumor activity of 70 mg (cohort A) or 100 mg (cohort B) sitravatinib QD plus tislelizumab in pts with locally recurrent or metastatic TNBC, and their combination with nab-paclitaxel in untreated locally recurrent inoperable or metastatic TNBC pts (cohort C). The preliminary result of cohort A has been reported with ORR of 38.1% (Lei Fan. JCO 2022 40:16_suppl). This analysis aims to report the updated analysis in cohort A and the interim results in cohort B.

Methods

Pts with locally recurrent or metastatic TNBC were included and received 70 mg (cohort A) or 100 mg (cohort B) sitravatinib QD PO and 200 mg tislelizumab IV Q3W until disease progression or intolerable toxicity. The primary endpoints included ORR (cohort A and B) and rate of grade ≥3 treatment-related adverse events (TRAEs) (cohort B). Secondary endpoints included DCR, PFS, and safety/tolerability. Updated analysis was provided for cohort A (Simon's 2 stage design). A Bayesian optimal phase II design with one interim analysis (on the first 20 pts out of the total 40 efficacy evaluable pts) was employed for cohort B to monitor both efficacy and safety primary endpoints simultaneously. The stopping boundary of interim analysis was the number of pts with ORR ≤1 or with grade ≥3 TRAEs ≥13.

Results

The data cutoff date of this analysis is 29 July 2022. In cohort A, with the median follow up of 10.7 months, the median PFS was 9.7 (95% CI: 2.8, 12.5) months among 21 efficacy evaluable pts. The rate of grade ≥3 TRAEs was 33.3% (7/21). In cohort B, among the first 20 efficacy evaluable pts with the median follow up of 4.1 months, 9 (45.0% [95% CI: 23.8%-67.9%]) pts were with confirmed ORR and 4 (20%) pts experienced grade ≥3 TRAEs, which met the interim goal. DCR was 80.0% (95% CI: 56.3%-94.27%). At the data cutoff, cohort B achieved the predefined enrollment target.

Conclusions

Sitravatinib combined with tislelizumab demonstrated clinically meaningful anti-tumor activity and had a manageable safety profile in the targeted pts population.

Clinical trial identification

NCT04734262.

Legal entity responsible for the study

The authors.

Funding

BeiGene (Beijing) Co., Ltd.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

75P - Effectiveness and safety data from IMreal Cohort 1: patients (pts) with urothelial cancer (UC) receiving atezolizumab after platinum-containing chemotherapy (plt chemo) under real-world conditions (ID 167)

Presentation Number
75P
Lecture Time
12:30 - 12:30
Speakers
  • Nicolas Girard (Paris, France)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Atezolizumab (atezo; anti–PD-L1) is used to treat pts with locally advanced/metastatic UC after plt chemo or who are cisplatin-ineligible and whose tumours have PD-L1 expression ≥5%. IMreal (NCT03782207) is a non-interventional, global, multi-centre, multi-cohort prospective study evaluating the short- and long-term outcomes and safety with atezo in the real-world setting. We report the second interim effectiveness and safety data for Cohort 1.

Methods

Eligible pts were adults with UC receiving atezo either as 2L+ treatment following plt chemo or after disease progression within 12 mo of adjuvant plt chemo. Enrolment occurred from 7 Feb 2019 to 15 Sept 2020. Clinical outcomes and safety data before, during and after treatment with atezo were collected. Pt history data were retrospectively collected. The primary endpoint is overall survival (OS) rate. Key secondary endpoints include progression-free survival (PFS), overall response rate (ORR), pt characteristics and safety.

Results

are based on 116 pts with a data cutoff of 1 Jun 2021. See table for key endpoints and pt characteristics. Overall, 105 pts (91%) had ≥1 AE. Treatment-related (TR) AEs occurred in 38 pts (33%), with Grade 3/4 TRAEs in 6 pts (5%). AEs of special interest occurred in 27 pts (23%); they were considered related in 14 pts (12%). Three pts (3%) had a Grade 5 AE, including 2 (2%) due to complication of device insertion/device obstruction and 1 (1%) due to a bacterial infection; neither was considered related.

Atezo (N=116)
Effectiveness (95% CI)
OS, median, mo 7.5 (5.5, 12.3)
1-y OS rate, % 41 (31.6, 50.1)
PFS, median, mo 4.4 (3.3, 5.7)
ORR, % 31 (21.1, 42.7)
Disease control rate, % 57 (45.4, 68.4)
Duration of response, median, mo 16.4 (12.0, NE)
Demographics, n (%)
Age ≥65 y 77 (66)
Male 94 (81)
White 113 (97)
ECOG PS n=90
0 23 (31)
1 46 (5)
2 15 (17)
3 1 (1)
PD-L1 statusa n=22
Negative 11 (50)
Positive 9 (41)
Unknown 2 (9)
Histology n=116
Urothelial 109 (94)
Non-urothelial 2 (2)
Mixed 5 (4)
Disease statusb
Metastatic 112 (97)
Locally recurrent 24 (21)
Locally advanced unresectable 8 (7)
Clinical stage n=114
IIIB 8 (7)
IVA 58 (51)
IVB 48 (42)

a Per various tests. b Categories are not mutually exclusive.

Conclusions

The OS rate with and safety profile of atezo remain consistent with those observed in pivotal studies, with no new or unexpected safety signals identified.

Clinical trial identification

NCT03782207.

Editorial acknowledgement

Medical writing support was provided by Marcia Gamboa, PhD of Health Interactions, funded by F. Hoffmann-La Roche.

Legal entity responsible for the study

F. Hoffmann-La Roche.

Funding

F. Hoffmann-La Roche.

Disclosure

N. Girard: Financial Interests, Personal, Invited Speaker: AstraZeneca, BMS, MSD, Roche, Pfizer, Mirati, Amgen, Novartis, Sanofi; Financial Interests, Personal, Advisory Board: AstraZeneca, BMS, MSD, Roche, Pfizer, Janssen, Boehringer, Novartis, Sanofi, AbbVie, Amgen, Lilly, Grunenthal, Takeda, Owkin; Financial Interests, Institutional, Research Grant, Local: Roche, Sivan, Janssen; Financial Interests, Institutional, Funding: BMS; Non-Financial Interests, Personal, Officer, International Thymic malignancy interest group, president: ITMIG; Other, Personal, Other, Family member is an employee: AstraZeneca. S. Popat: Financial Interests, Personal, Advisory Board: Boehringer Ingelheim, Novartis, Amgen, Janssen, Daiichi Sankyo, AstraZeneca, Bayer, BMS, Blueprint, Merck Serono, Guardant Health, Beigene, Takeda, Lilly, Seattle Genetics, Turning Point Therapeutics, Xcovery, GlaxoSmithKline, MSD, Pfizer, Sanofi; Financial Interests, Personal, Expert Testimony: Roche; Financial Interests, Personal, Invited Speaker: Medscape, VJ Oncology, Touch Medical; Financial Interests, Institutional, Invited Speaker: AstraZeneca, Ariad, Roche, Boehringer Ingelheim, Celgene, Daiichi Sankyo, Trizel, GlaxoSmithKline, Takeda, Turning Point Therapeutics, Roche, Janssen, BMS, Lilly; Financial Interests, Personal, Other, Journal Deputy Editor, Lung Cancer: Elsevier; Financial Interests, Institutional, Other, Sub-investigator: Amgen; Financial Interests, Institutional, Other, Sub-Investigator: MSD, Blueprint, Seattle Genetics; Financial Interests, Institutional, Research Grant: Guardant Health; Non-Financial Interests, Personal, Leadership Role, Foundation Council Member, Unpaid: European Thoracic Oncology Platform; Non-Financial Interests, Personal, Leadership Role, Chair of Steering Committee, Unpaid: British Thoracic Oncology Group; Non-Financial Interests, Personal, Officer, Thoracic Faculty, Unpaid: European Society of Medical Oncology; Non-Financial Interests, Personal, Advisory Role, Mesothelioma Task-force Member, Unpaid: International Association for the Study of Lung Cancer; Non-Financial Interests, Personal, Invited Speaker, Unpaid: Mesothelioma Applied Research Foundation; Non-Financial Interests, Personal, Advisory Role, Honorary Clinical Advisor, Unpaid: ALK Positive UK; Non-Financial Interests, Personal, Advisory Role, Research Advisory Group Member, Unpaid: Ruth Strauss Foundation; Non-Financial Interests, Personal, Advisory Role, Scientific Adivsory Board Member, Unpaid: Lung Cancer Europe. S. Shoshkova: Financial Interests, Personal, Full or part-time Employment: Roche. S. Fear: Financial Interests, Personal, Full or part-time Employment: Roche. J.L. Perez Gracia: Financial Interests, Institutional, Research Grant: Roche, BMS, MSD, Seattle Genetics; Financial Interests, Personal, Speaker’s Bureau: Roche, BMS, Ipsen, MSD, Seattle Genetics; Financial Interests, Personal, Other, Travel Support: Roche, MSD, BMS.

Collapse
Poster Display Poster Display session

76P - Safety profile of adjuvant pembrolizumab (pembro) in melanoma, nonÐsmall cell lung cancer (NSCLC), and renal cell carcinoma (RCC): pooled analysis of phase 3 clinical trials (ID 168)

Presentation Number
76P
Lecture Time
12:30 - 12:30
Speakers
  • Jason J. Luke (Pittsburgh, PA, United States of America)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Pembro has well characterized, manageable safety in the advanced setting. Clinical data suggest pembro has a similar profile in the adjuvant setting; further characterization is important given the extended life expectancy of pts with resectable disease.

Methods

Safety data from pts with resected stage IIIA-C (AJCC 7th ed) melanoma (KEYNOTE-054), resected stage IB-IIIA (AJCC 7th ed) NSCLC (PEARLS/KEYNOTE-091), RCC post-nephrectomy/metastasectomy (KEYNOTE-564), and resected stage IIB-C (AJCC 8th ed) melanoma (KEYNOTE-716) were pooled. Pts received adjuvant pembro 200 mg (2 mg/kg pediatric pts) Q3W or placebo for up to ∼1 yr. Safety was monitored for ≤30 days after treatment end (90 days for serious AEs). AEs were graded using NCI CTCAE v4.0.

Results

Data from 4125 pts were pooled (pembro, n=2060; placebo, n=2065). Median age was 61.0 yrs; 1351 (65.6%) pts treated with pembro and 1343 (65.0%) with placebo were male. Any-grade treatment-related AEs (TRAEs) occurred in 1620 (78.6%) pts treated with pembro and 1212 (58.7%) with placebo. Grade 3-5 TRAEs occurred in 336 (16.3%) pts treated with pembro and 72 (3.5%) with placebo; most common were diarrhea (n=23; 1.1%), ALT increased (n=20; 1.0%), and colitis (n=20; 1.0%) with pembro and lipase increased (n=13; 0.6%) and fatigue (n=6; 0.3%) with placebo. 4 pts in the pembro group died due to a TRAE. TRAEs led to discontinuation in 326 (15.8%) pts treated with pembro and 44 (2.1%) with placebo. Immune-mediated AEs (imAEs) and infusion reactions occurred in 761 (36.9%) pts treated with pembro and 193 (9.3%) with placebo (grade 3-5: n=181 [8.8%] vs n=23 [1.1%]). Most common imAEs were hypothyroidism (n=382; 18.5%), hyperthyroidism (n=227; 11.0%), and pneumonitis (n=82; 4.0%) with pembro and hypothyroidism (n=76; 3.7%), pneumonitis (n=29; 1.4%), and hyperthyroidism (n=27; 1.3%) with placebo. Median time to onset of first imAE or infusion reaction was 2.6 mo with pembro and 4.1 mo with placebo.

Conclusions

Data from this pooled analysis of >4000 patients across several tumor types show that adjuvant pembro has a manageable safety profile consistent in severity and management with the established profile of pembro in advanced disease.

Clinical trial identification

NCT02362594 (first posted to clinicaltrials.gov: Feb 13, 2015); NCT02504372 (first posted to clinicaltrials.gov: July 21, 2015); NCT03142334 (first posted to clinicaltrials.gov: May 5, 2017); NCT03553836 (first posted to clinicaltrials.gov: Jun 12, 2018).

Editorial acknowledgement

Medical writing and/or editorial assistance was provided by Jemimah Walker, PhD, and Holly C. Cappelli, PhD, CMPP, of ApotheCom (Yardley, PA, USA). This assistance was funded by Merck Sharp & Dohme Llc, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA.

Legal entity responsible for the study

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

Funding

Funding for this research was provided by Merck Sharp & Dohme Llc, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA.

Disclosure

J.J. Luke: Financial Interests, Personal, Other, DSMB: AbbVie, Immutep, Evaxion; Financial Interests, Personal, Advisory Board: 7 Hills, Bright Peak, Exo, Fstar, Inzen, RefleXion, Xilio, Actym, Alphamab Oncology, Arch Oncology, Duke Street Bio, Kanaph, Mavu, NeoTx, Onc.AI, OncoNano, Pyxis, Saros, STipe, Tempest; Financial Interests, Personal, Advisory Role: AbbVie, Alnylam, Atomwise, Bayer, Bristol-Myers Squibb, Castle, Checkmate, Codiak, Crown, Cugene, Curadev, Day One, Eisai, EMD Serono, Endeavor, Flame, G1 Therapeutics, Genentech, Gilead, Glenmark, HotSpot, Kadmon, KSQ, Janssen, Ikena, Inzen, Immatics, Immunocore, Incyte, Instil, IO Biotech, Macrogenics, Merck, Mersana, Nektar, Novartis, Partner, Pfizer, Pioneering Medicines, PsiOxus, Regeneron, Ribon, Roivant, Servier, STINGthera, Synlogic, Synthekine; Financial Interests, Institutional, Research Grant: AbbVie, Astellas, AstraZeneca, Bristol-Myers Squibb, Corvus, Day One, EMD Serono, Fstar, Genmab, Ikena, Immatics, Incyte, Kadmon, KAHR, Macrogenics, Merck, Moderna, Nektar, Next Cure, Numab, Palleon, Pfizer, Replimmune, Rubius, Servier, Synlogic, Takeda, Trishula, Tizona, Xencor. G.V. Long: Financial Interests, Personal, Other, Consultant/Advisor: Agenus, Amgen, Array Biopharma, Boehringer Ingelheim International GmbH , Bristol Myers Squibb , Evaxion Biotech A/S , Hexal AG (Sandoz Company) , Highlight Therapeutics S.l., Innovent Biologics USA Inc, Merck Sharp & Dohme (Australia) Pty Limited , Merck Sharp & Dohme, Novartis Pharma AG, OncoSec Medical Australia, PHMR Limited, Pierre Fabre , Provectus Australia , Qbiotics Group Limited , Regeneron Pharmaceuticals Inc. C. Robert: Financial Interests, Personal, Advisory Board: BMS, Roche, Pierre Fabre, Novartis, Sanofi, MSD, Pfizer, AstraZeneca. M.S. Carlino: Financial Interests, Personal, Advisory Board: Amgen, Bristol Myers Squibb, Eisai, Ideava, Merck Sharp & Dohme, Nektar, Novartis, Oncosec, Pierre-Fabre, Obiotics, Regeneron, Roche. T.K. Choueiri: Financial Interests, Personal and Institutional, Funding: AstraZeneca, Aravive, Aveo, Bayer, Bristol Myers-Squibb, Calithera, Circle Pharma, Eisai, EMD Serono, Exelixis, GlaxoSmithKline, IQVA, Infinity, Ipsen, Jansen, Kanaph, Lilly, Merck, Nikang, Nuscan, Novartis, Pfizer, Roche, Sanofi/Aventis, Surface Oncology; Financial Interests, Personal and Institutional, Invited Speaker: AstraZeneca, Aravive, Aveo, Bayer, Bristol Myers-Squibb, Calithera, Circle Pharma, Eisai, EMD Serono, Exelixis, GlaxoSmithKline, IQVA, Infinity, Ipsen, Jansen, Kanaph, Lilly, Merck, Nikang, Nuscan, Novartis, Pfizer, Roche, Sanofi/Aventis, Surface Oncology; Financial Interests, Personal and Institutional, Advisory Board: AstraZeneca, Aravive, Aveo, Bayer, Bristol Myers-Squibb, Calithera, Circle Pharma, Eisai, EMD Serono, Exelixis, GlaxoSmithKline, IQVA, Infinity, Ipsen, Jansen, Kanaph, Lilly, Merck, Nikang, Nuscan, Novartis, Pfizer, Roche, Sanofi/Aventis, Surface Oncology; Financial Interests, Personal and Institutional, Principal Investigator: AstraZeneca, Aravive, Aveo, Bayer, Bristol Myers-Squibb, Calithera, Circle Pharma, Eisai, EMD Serono, Exelixis, GlaxoSmithKline, IQVA, Infinity, Ipsen, Jansen, Kanaph, Lilly, Merck, Nikang, Nuscan, Novartis, Pfizer, Roche, Sanofi/Aventis, Surface Oncology; Financial Interests, Personal and Institutional, Advisory Role: AstraZeneca, Aravive, Aveo, Bayer, Bristol Myers-Squibb, Calithera, Circle Pharma, Eisai, EMD Serono, Exelixis, GlaxoSmithKline, IQVA, Infinity, Ipsen, Jansen, Kanaph, Lilly, Merck, Nikang, Nuscan, Novartis, Pfizer, Roche, Sanofi/Aventis, Surface Oncology; Non-Financial Interests, Personal and Institutional, Invited Speaker: Peerview, OncLive, MJH. N.B. Haas: Financial Interests, Personal, Advisory Board: Aveo, Eisai, Roche Genentech, Merck; Financial Interests, Personal, Invited Speaker: Exelixis. M.E.R. O'Brien: Financial Interests, Personal and Institutional, Advisory Board: Royal Marsden Hospital; Financial Interests, Personal and Institutional, Invited Speaker: Royal Marsden Hospital; Financial Interests, Personal and Institutional, Funding: Royal Marsden Hospital; Financial Interests, Personal and Institutional, Principal Investigator: Royal Marsden Hospital. S. Peters: Financial Interests, Institutional, Other, Consultant/Advisor: AbbVie, AiCME, Amgen, Arcus, AstraZeneca, Bayer, Beigene, Biocartis, BioInvent, Blueprint Medicines, Boehringer Ingelheim, Bristol-Myers Squibb, Clovis, Daiichi Sankyo, Debiopharm, Provectus Australia, Eli Lilly, Elsevier; Financial Interests, Institutional, Other, Consultant, Advisory: F-Star, Fishawack, Foundation Medicine, Genzyme, Gilead, GSK, Illumina; Financial Interests, Institutional, Other, Consultant/Advisory: Imedex, IQVIA, Incyte, iTeos, Janssen, Medscape, Medtoday, Merck Sharp and Dohme, Merck Serono, Merrimack, Novartis, Novocure, OncologyEducation, Pharma Mar, Phosplatin Therapeutics, PER, Peerview, Pfizer, PRIME, Regeneron, RMEI, Roche/Genentech, RTP, Sanofi, Seattle Genetics, Takeda, Vaccibody; Financial Interests, Institutional, Invited Speaker: AiCME, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, ecancer, Eli Lilly, Foundation Medicine, Illumina, Imedex, Medscape, Merck Sharp and Dohme, Mirati, Novartis, PER, Peerview, Pfizer, Prime, Roche/Genentech, RTP, Sanofi, Takeda; Financial Interests, Institutional, Research Grant: Amgen, AstraZeneca, Beigene, Bristol-Myers Squibb, GSK, Merck Sharp and Dohme, Roche/Genentech. M.A. Leiby: Financial Interests, Personal, Full or part-time Employment: Merck Sharp & Dohme Llc; Financial Interests, Personal, Stocks/Shares: Merck & Co., Inc. J. Lin: Financial Interests, Personal, Full or part-time Employment: Merck; Financial Interests, Personal, Stocks/Shares: Merck. Y. Zhao: Financial Interests, Personal, Full or part-time Employment: Merck; Non-Financial Interests, Personal, Member: Merck. C. Krepler: Financial Interests, Personal, Stocks/Shares: Merck; Financial Interests, Personal, Full or part-time Employment: Merck. R.F. Perini: Financial Interests, Personal, Stocks/Shares: Merck; Financial Interests, Personal, Full or part-time Employment: Merck Sharp & Dohme Llc. M.C. Pietanza: Financial Interests, Personal, Full or part-time Employment: Merck Sharp & Dohme Llc; Financial Interests, Personal, Stocks/Shares: Merck & Co., Inc. A. Samkari: Financial Interests, Personal, Stocks/Shares: Merck & Co., Inc.; Financial Interests, Personal, Full or part-time Employment: Merck Sharp & Dohme Llc. T. Gruber: Financial Interests, Personal, Full or part-time Employment: Merck, Clinical Safety and Risk Management; Financial Interests, Personal, Stocks/Shares: Merck, Clinical Safety and Risk Management; Financial Interests, Institutional, Leadership Role: Merck, Clinical Safety and Risk Management. N. Ibrahim: Financial Interests, Personal, Full or part-time Employment: Merck Sharp & Dohme Llc; Financial Interests, Personal, Stocks/Shares: Merck & Co., Inc. A.M.M. Eggermont: Financial Interests, Personal, Speaker’s Bureau: BMS, Merck; Financial Interests, Personal, Advisory Board: Agenus, BioInvent, Brenus,CatalYm, Clover, Ellipses, Galecto, GenOway, GSK, IO Biotech, IQVIA, ISA Pharmaceuticals, Merck/MSD, Sairopa, Scorpion, Sellas, SkylineDX, TigaTX, Trained Therapeutics Discovery; Financial Interests, Personal, Stocks/Shares: IOBiotech, Sairopa, SkylineDX; Financial Interests, Personal, Principal Investigator: KEYNOTE-054; Financial Interests, Personal, Other, IDMC: BioNTech, IQVIA, GSK, Pfizer. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

77P - A Study to Evaluate the Safety, Tolerability and Efficacy of IBI939 in Combination With Sintilimab in Patients with Previously Untreated Locally Advanced Unresectable or Metastatic PD-L1-Selected Non-Small Cell Lung Cancer (NSCLC) (ID 169)

Presentation Number
77P
Lecture Time
12:30 - 12:30
Speakers
  • Ying Cheng (Changchun, Ji, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

T-cell immunoreceptor with Ig and ITIM domains (TIGIT) plays an important role in tumor immunosurveillance in addition to well-established immunosuppressive checkpoint receptors such as PD-1 and CTLA-4. We report the preliminary safety and anti-tumor activity of IBI939 (anti-TIGIT mAb) in combination with sintilimab (anti-PD-1 mAb) in patients (pts) with previously untreated locally advanced unresectable or metastatic PD-L1-selected NSCLC.

Methods

Eligible pts with systemic treatment-naïve, advanced or metastatic NSCLC, PD-L1 TPS ≥50%, and driver gene negative were enrolled and randomized 2:1 to IBI939 20 mg/kg plus sintilimab 200 mg IV Q3W (arm A) or sintilimab 200 mg monotherapy IV Q3W (arm B). The primary objective was to evaluate the ORR per RECIST v1.1. The secondary objectives include evaluation of PFS per RECIST v1.1, OS, and safety.

Results

Of 42 pts enrolled, 28 pts (median age: 65; adenocarcinoma: n=19; brain metastasis: n=7) and 14 pts (median age: 58; adenocarcinoma: n=6; brain metastasis: n=1) were in arm A and arm B, respectively. As of June 30th, 2022, among 40 response-evaluable NSCLC pts (27 in arm A vs 13 in arm B), the confirmed ORR was 66.7% (95% CI, 46.0-83.5) vs 61.5% (95% CI, 31.6-86.1)(arm A vs B). Nine pts with the event (progressive disease or death) had occurred in both arms. The median PFS was not reached (95% CI, 6.8-NA) in arm A vs 6.0 months (95% CI, 1.4-NA) in arm B (HR: 0.43; 95% CI, 0.17-1.10). The incidence of TRAEs was 85.7% vs 71.4% (2 and 5 pts experienced ≥ grade 3 events in each arm), respectively. The most common TRAEs were hypothyroidism (35.7%), aspartate aminotransferase increased (21.4%), blood urea increased (17.9%), hyperthyroidism (17.9%) in arm A. Immune-related AEs (determined by the investigator) were reported in 64.3% of pts in arm A and 50.0% of pts in arm B.

Conclusions

IBI939 plus sintilimab demonstrated improved PFS benefit and manageable safety profile in PD-L1 TPS ≥ 50% NSCLC patients with no prior systemic treatment.

Clinical trial identification

NCT04672369.

Legal entity responsible for the study

Innovent Biologics, Inc., Suzhou, Jiangsu, China.

Funding

Innovent Biologics, Inc., Suzhou, Jiangsu, China.

Disclosure

Y. Chen, G. Han, J. Ye: Financial Interests, Personal, Sponsor/Funding, Employment: Innovent Biologics, Inc. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

78P - Clinical outcomes with atezolizumab plus bevacizumab (AB) or lenvatinib (L) in hepatocellular carcinoma (HCC). (ID 170)

Presentation Number
78P
Lecture Time
12:30 - 12:30
Speakers
  • MARA PERSANO (Cagliari, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The aim of this multicenter study is to compare response obtained by AB o L and identify any factors that may predict the emergence of this response to one of two treatments in a real-world setting.

Methods

Study population derived from retrospective analysis of prospectively collected HCC patients (P) treated with AB or L as first-line treatment. ORR with AB vs L was the primary endpoint. The secondary endpoint compared OS with L vs AB in terms of best response.

Results

1312 P were treated with L, and 823 P were treated with AB. ORR was 38.6% for P receiving L, and 27.3% for P receiving AB [p < 0.01; odds ratio (OR) 0.60)]. In responding P (CR + PR), OS was 22.3 months (m) for P receiving L, and 22.5 m for P treated with AB (p 0.20; HR 0.81, reference L). In non-responding P (SD + PD), OS was 10.8 m for P receiving L, and 11.5 m for P receiving AB (p 0.36; HR 0.85, reference L). For P who achieved CR, OS was not reached in both arms, but the result from univariate Cox regression model showed 62% reduction of death risk for P treated with AB (p 0.05). In all multivariate analyses, treatment arm was not found to be an independent factor conditioning OS. Comparing ORR achieved in the two arms, L was shown to be statistically significant in all subgroup P except for Child Pugh B, presence of portal vein thrombosis (PT), αFP ≥ 400 ng/mL, presence of extrahepatic disease (EHD), ALBI 2, and no previous locoregional procedures. P who achieved CR were compared to the rest of the population in the same way. The only statistically significant difference was in favor of L in P with NLR > 3 (p 0.03; OR 0.39). Finally, we evaluated separately which P had more response in AB arm and then in L arm. Only in L arm, three factors favoring response were highlighted: absence of EHD (p < 0.01; OR 1.88), BCLC B (p < 0.01; OR 0.54), absence of PT (p 0.05; OR 1.34), and NLR ≤ 3 (p < 0.01; OR 0.70).

Conclusions

L achieve higher ORR in all P subgroups. P who achieve CR with AB can achieve OS so far never recorded in HCC patients. This study didn’t highlight any factors that could identify HCC P subgroups capable of obtaining CR.

Legal entity responsible for the study

M. Persano.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

79P - Efficacy and Safety of Zimberelimab (GLS-010) Monotherapy in Patients with Recurrent or Metastatic CervicalCancer: A Multicenter, Open-Label, Single-Arm, Phase II Study (ID 171)

Presentation Number
79P
Lecture Time
12:30 - 12:30
Speakers
  • Xiaohua Wu (Shanghai, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Zimberelimab is a novel, fully human anti-PD-1 monoclonal antibody with high affinity and selectivity for PD-1.The objective of this study (NCT03972722) was to evaluate zimberelimab, a novel, anti-programmed cell death protein 1 monoclonal antibody, in patients with programmed death ligand-1-positive recurrent or metastatic cervical cancer that had progressed after first- or subsequent-lineplatinum containing standard chemotherapy.

Methods

In this single-arm, phase II study, eligible patients in 27 Chinese sites were assigned to receive intravenous zimberelimab 240 mg as monotherapy every 2 weeks until confirmed disease progression, death, intolerable adverse effects, or withdrawal from the study. The primary endpoint was the objective response rate (ORR) assessed per Response Evaluation Criteria in Solid Tumors (version 1.1) by an independent review committee. Secondary endpoints included duration of response (DoR), disease control rate (DCR), progression-free survival(PFS), overall survival (OS), and safety.

Results

Ninety participants were included in the full analysis set, with a median follow-up of 11.5 months. Complete and partial responses were achieved by 4 and 21 patients, respectively, corresponding to an ORR of 27.8% (95% confidence interval [CI], 18.85 to 38.22; P < .0001 vs historical controls). Median OS and DoR were not reached during the study: 12-month OS rates were 54% (95% CI, 41 to 66) and 6-month DoR rates were 84% (95% CI, 58 to 95). Median PFS was 3.7 months and the 12-month PFS rate was 15% (95% CI, 2 to 42). Treatment-related adverse events (TRAEs) occurred in 78.1% of participants, with hypothyroidism (25.7%) and anemia (19.0%) being the most frequently reported. Grade ≥ 3 TRAEs occurred in 22.9% of participants.

Conclusions

Zimberelimab monotherapy demonstrated durable antitumor activity and an acceptable safety profile in patients with recurrent or metastatic cervical cancer that had progressed after first- or subsequent-line platinum-containing standard chemotherapy. Further investigation of zimberelimab in patients with cervical cancer is warranted.

Clinical trial identification

NCT03972722.

Legal entity responsible for the study

The authors.

Funding

Guangzhou Gloria Biosciences Co., Ltd.

Disclosure

J. Zhu: Financial Interests, Personal, Member of the Board of Directors: Guangzhou Gloria Biosciences Co., Ltd. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

80P - Penpulimab (Anti-PD-1) combined with anlotinib as first-line therapy for unresectable hepatocellular carcinoma (uHCC): Updated overall survival results from a phase Ib/II study (ID 172)

Presentation Number
80P
Lecture Time
12:30 - 12:30
Speakers
  • Chun Han (Beijing, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The combination of immune checkpoint inhibitors and tyrosine kinase inhibitors (TKIs) manifested high efficacy in uHCC. Anlotinib was a novel oral multi-targeted TKI selective for VEGF receptors 1/2/3, FGF receptors 1-4, PDGF receptors α and β, and c-kit. Penpulimab, a humanized anti-PD-1 IgG1 monoclonal antibody, was engineered to eliminate FcγR binding, consequently eliminating ADCC, ADCP and ADCR effects. This AK105-203 trial (NCT04172571) aimed to explore the efficacy and safety of penpulimab plus anlotinib in patients (pts) with histologically or cytologically confirmed uHCC.

Methods

In this single-arm, multicenter phase Ib/II trial, pts with uHCC and no prior systemic treatment were eligible if they were 18-75 years, and classified as BCLC stage B (not amenable for locoregional therapy) or C, Child-Pugh score ≤7 and ECOG PS of 0-1. Pts received anlotinib (8 mg, p.o., qd, d1-14, q3w) plus penpulimab (200mg, iv, d1, q3w). The primary endpoint was ORR (RECIST v1.1). Secondary endpoints were safety, DCR, DoR, TTP, PFS and OS.

Results

31 pts (median age 56 years [23-74], ECOG 0/1 [64%/36%], BCLC B/C [23%/77%], HBV/HCV [61%/7%]) received combined therapy. As of August 5, 2022, median follow-up time was 23.0 months (range 3.7-31.9). The ORR was 31.0% (95% CI, 15.3-50.8%), and DCR was 82.8% (95% CI, 64.2-94.2%). The median PFS and TTP for 31 patients were 8.8 months (95% CI, 4.0-12.3) and 8.8 months (95% CI, 4.0-14) respectively. OS events were observed in 16 patients (51.6%), and the median OS was 23.0 months with 12-months OS rate was 67.9%. Treatment-related adverse events (TRAEs) occurred in 90.3% of pts (≥G3 in 25.8% [8/31]). No G5 AE occurred. Most common TRAEs (≥25%) were increased AST (41.9%) and ALT (35.5%), general disorders and administration site conditions (35.5%), skin and subcutaneous tissue disorders (32.3%), platelet count decreased (25.8%), asthenia (25.8%).

Conclusions

Anlotinib combined with penpulimab showed encouraging efficacy and acceptable safety in pts with uHCC. The further randomized, phase 3 study of penpulimab plus anlotinib at a higher dose (10 mg) in this setting is ongoing (NCT04344158).

Clinical trial identification

NCT04172571, release date November 21, 2019.

Legal entity responsible for the study

Akeso, Inc.

Funding

Akeso, Inc.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

81P - Is PD-1 inhibitor based treatment better than chemotherapy for metastatic NSCLC patients with PD-L1≥50% who develop EGFR-TKI resistance? A real-world investigation (ID 173)

Presentation Number
81P
Lecture Time
12:30 - 12:30
Speakers
  • Yujing Li (Shanghai, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Platinum-based chemotherapy is still the standard of care for Epidermal growth factor receptor (EGFR) mutated non-small cell lung cancer (NSCLC) patients after developing EGFR-TKI resistance. However, no study focusing on the role of PD-1 inhibitor based treatments for EGFR mutated NSCLC patients who carried PD-L1 TPS ≥ 50% progressed after EGFR-TKI therapy. Thus, we aimed to investigate the outcomes of PD-1 inhibitor based treatments for these patients and to explore the population that may benefited from PD-1 inhibitor based therapies.

Methods

We retrospectively collected data of EGFR mutated advanced NSCLC patients with PD-L1 TPS≥50% who have failed prior EGFR-TKI therapies without T790M mutation at Shanghai Chest Hospital between January 2018 and June 2021. Progression-free survival (PFS) and overall survival (OS) were utilized to evaluate the outcomes.

Results

A total of 146 patients were included. The median follow-up was 36.7 months (IQR, 12.5-44.2 months). Among the population, 66 patients (45.2%) received chemotherapy, the remaning (54.8%) received PD-1 inhibitor based therapies, including 56 patients(70.0%) received PD-1 inhibitor combined with chemotherapy (PC) and 24 patients (30.0%) received PD-1 inhibitor monotherapy (PM). Survival analysis shown that patients who received PD-1 inhibitor based therapies had better PFS and OS compared with those treated with other therapy (median PFS, 4.0 vs. 10.0 months, P<0.001; median OS, 39.5 vs. 24.2 months, P<0.001). What’s more, patients who treated with PC treatment had a superior survival time than those received PM treatment (median PFS, 10.3 vs. 7.0 months, P<0.001; median OS, 32.4 vs. 41.6 months, P<0.001). Subgroup analysis found that the PFS and OS benefit of PC was evident in all subgroups.

Conclusions

For advanced NSCLC patients with EGFR mutations and PD-L1 TPS≥50% who have failed prior EGFR-TKI therapies without T790M mutation, PD-1 inhibitor based treatment could provide a more favorable survival than classical chemotherapy. What's more, compared with PD-1 inhibitor monotherapy, PD-1 inhibitor combined with chemotherapy seems to be the preferred treatment.

Legal entity responsible for the study

The authors.

Funding

Shanghai Shenkang Organization.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

82P - Preliminary analysis of tislelizumab (TIS) and chemotherapy as neoadjuvant therapy for potentially resectable stage IIIA/IIIB non-small cell lung cancer (NSCLC) (ID 174)

Presentation Number
82P
Lecture Time
12:30 - 12:30
Speakers
  • Ting Wang (Chengdu, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The benefit of neoadjuvant immunotherapy and chemotherapy in resectable NSCLC indicated that this combination therapy may provide more surgical opportunities and survival benefits to potentially resectable locally advanced NSCLC. Herein, we initiated a phase II study to evaluate the feasibility of immunotherapy plus chemotherapy in stage IIIA/IIIB NSCLC.

Methods

We planned to recruit 33 patients (pts) with stage IIIA/IIIB EGFR/ALK/ROS wild-type NSCLC. Eligible pts received 2 cycles of neoadjuvant chemoimmunotherapy (PD-1 inhibitor TIS, nab-paclitaxel, and cisplatin/carboplatin) and were reassessed for surgery. Thereafter, pts underwent surgery within 6 weeks and continued 2 cycles of TIS plus chemotherapy, followed by up to 15 cycles of TIS monotherapy. The primary endpoint was the R0 resection rate. Secondary endpoints were major pathologic response (MPR), pathologic complete response (pCR), disease-free survival, and overall survival.

Results

From Jan 2021 to Sep 2022, 18 of 33 enrolled pts (54.5%) completed neoadjuvant therapy and underwent resection (13 with IIIA and 5 with IIIB disease). No treatment-related surgical delay occurred. 17 of 18 pts (94.4%) underwent successful R0 resection (Table). Of 18 pts who underwent resection,6 (33.3%) achieved pCR and 4 (22.2%) achieved MPR, resulting in an overall pathologic response rate of 55.6%. Of the 4 pts who achieved MPR, 3 had only 1% viable tumor cells in the resection specimen. The overall response rate (ORR) and disease control rate (DCR) were 88.9% (16/18) and 100 % (18/18), respectively. Both the clinical and pathological downstaging occurred in 16 of 18 pts (88.9%).

Outcomes Results, n (%, 95%CI); n = 18
Radiological response
PR 16 (88.9, 65.29-98.62)
SD 2 (11.1, 1.38-34.71)
ORR 16 (88.9, 65.29-98.62)
DCR 18 (100, 81.5-100.0)
Surgical resection
R0 17 (94.4, 72.71-99.86)
R1 1 (5.6, 0.14-27.29)
Downstaging rate
clinical 16 (88.9, 65.29-98.62)
pathologic 16 (88.9, 65.29-98.62)
Pathologic response 10 (55.6, 30.76-78.47)
MPR 4 (22.2, 6.41-47.64)
pCR 6 (33.3, 13.34-59.01)

Conclusions

Neoadjuvant TIS plus chemotherapy increased surgical opportunities in potentially resectable locally advanced stage IIIA/IIIB NSCLC. The encouraging R0 resection rate observed in this study supports further investigation.

Clinical trial identification

NCT04865705.

Editorial acknowledgement

Chunlu Shu from Medical Affairs, BeiGene, Ltd.

Legal entity responsible for the study

Qinghua Zhou and Yongsheng Wang.

Funding

BeiGene Ltd.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

83P - The Immune-related adverse event (IRAE) Likelihood Score (ILS) identifies ÒpureÓ IRAEs strongly associated with outcome in a phase 1-2 trial population (ID 175)

Presentation Number
83P
Lecture Time
12:30 - 12:30
Speakers
  • Luca Mazzarella (Milan, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immune-related adverse events (IRAE) pose a significant diagnostic and therapeutic challenge in patients treated with immuno-oncology (IO) drugs. IRAEs have been found to correlate with better outcome, but studies are conflicting on the magnitude and significance of this correlation. Estimating the true incidence of IRAEs is particularly difficult in the early phase 1/2 trial setting, with factors contributing to both over- and under-estimation. A key issue is the lack of IRAE diagnostic criteria, necessary to discriminate “pure” IRAEs from other treatment-related adverse events not sustained by an autoimmune process. We present the definitive analysis of a retrospective study conducted on patients treated with IO drugs within phase 1-2 trials at our institute.

Methods

We extensively reviewed clinical characteristics and temporal dynamics of IRAEs and empirically developed an IRAE Likelihood Score (ILS) based on availability of invasive or highly specific tests, response to immune suppression, temporal correlation with IO drug initiation, evidence ruling out alternative cause, known relationship with IO. We defined High Confidence (HC) or Low Confidence (LC) IRAEs by clinical consensus and estimated correlation with survival of treatment-related events by multivariate Cox analysis. To mitigate immortal time-bias, we also analysed data at 2-month landmark and modeling IRAEs as time-dependent covariate.

Results

29.2% of 202 patients developed ≥ 1 treatment-related adverse event. ILS ≥ 5 discriminated between HC and LC IRAEs with >93% specificity and sensitivity. HC IRAE patients (n=24) had significantly improved outcome for PFS and OS, irrespective of the model used (landmark, time-dependent or uncorrected, HR for PFS ranging 0.24-0.44, for OS 0.18-0.23, all p values <0.01), whereas LC IRAE patients (n=35) showed no statistically significant correlation.

Conclusions

ILS provides a simple system to identify bona fide IRAEs, pruning for other treatment-related events likely due to different pathophysiology. Applying stringent criteria leads to lower and more reliable estimates of IRAE incidence and identifies events with significant impact on survival.

Legal entity responsible for the study

The authors.

Funding

Italian Ministry of Health.

Disclosure

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

Collapse
Poster Display Poster Display session

84P - Ambulatory management of ICI-induced hepatitis: a safe and effective management approach (ID 176)

Presentation Number
84P
Lecture Time
12:30 - 12:30
Speakers
  • Anna C. Olsson-Brown (Liverpool, United Kingdom)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The incidence of immune related adverse events (irAEs) secondary to oncological immune checkpoint inhibitors (ICIs) are treated as standard with high dose corticosteroids (CST). Grade 3-4 hepatitis is managed with intravenous methylprednisolone (IVMP) as per international protocols. CST treatment is effective but if administered in the inpatient setting can lead to long hospital stays, psychological distress and increased risk of hospital related illness. Clatterbridge Cancer Centre (CCC), a UK tertiary cancer centre, has an established regional pan-tumour immunotherapy (IO) service to support all patients with irAEs and delivers a ambulatory IVMP pathway as part of that service.

Methods

A retrospective review of the ambulatory IVMP service since its introduction in 2018 was undertaken. The proportion of patients treated in an ambulatory setting was compared the situation prior to 2018. Additionally the responsiveness of the service and impact on admission, length of stay (LOS) and bed days (BD) was evaluated.

Results

Between 2018 and 2021 2017 patients were treated with checkpoint inhibitors regionally. 1027 patients experienced CST requiring irAEs of which 95 experienced grade 3/4 hepatitis requiring IVMP. Prior to the introduction of the service 100% (17/17) of patients required inpatient admission for IVMP associated with a median LOS of 12.5 days and accounting for 137.5 BD per annum. Following the introduction of the ambulatory service 15% (15/95) required admission for the introduction of IVMP, with 60% (9/15) of them completing their IVMP as an outpatient in the ambulatory setting. 75% (71/95) of patients had treatment commenced as a day case and there was an ambulatory to inpatient conversion of 9.9% (7/71), all of whom displayed CST insensitivity and required additional immunosuppresion. The median LOS was reduced to 4.75 days. Given the average number of patients requiring IVMP between 2020-22 was 34.5 (range 33-36) per year this is a saving of 268 bed days per annum resulting in a cost saving of £123,280 per annum.

Conclusions

The introduction of an ambulatory IVMP service for the management of immunotherapy induced hepatitis has been illustrated to be safe, effective, responsive to CST resistance and result in care provision efficiencies.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

A.C. Olsson-Brown: Financial Interests, Personal, Invited Speaker: BMS, MSD, Roche, Novartis, AZ, Eisai, B-I. T. Guinan: Financial Interests, Personal, Invited Speaker: BMS, MSD, Roche, Sanofi . All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

85P - Tislelizumab combined with apatinib and oxaliplatin plus S1 as neoadjuvant therapy for Borrmann IV, large Borrmann III type and Bulky N positive advanced gastric cancer: a single-arm multicenter trial (TAOS-3B-Trial) (ID 177)

Presentation Number
85P
Lecture Time
12:30 - 12:30
Speakers
  • Luchuan Chen (Fuzhou, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

We aimed to investigate the efficacy and safety of S1 plus oxaliplatin in combination with tislelizumab, a novel engineered anti-PD-1 monoclonal antibody, and apatinib, an inhibitor of VEGFR-2, as neoadjuvant therapy for Borrmann IV、large Borrmann III type(tumor size>5cm) and Bulky N positive advanced gastric cancer (GC).

Methods

This was a single-arm, multicenter, open-label phase II trial (NCT05223088). Eligible patients (pts) had histologically proven advanced HER-2 negativeGC with Borrmann IV, large Borrmann III type(tumor size>5cm) and Bulky N positive. The surgery was performed after 4 cycles of drug treatment (S1+oxaliplatin+tislelizumab+apatinib).

Results

Baseline Patient Characteristics: Among the 25 pts eligible which have been already postoperative efficacy evaluation in 40 pts, the median age was 57 years. The histological types were mainly poorly differentiated adenocarcinoma.

Characteristics N (%)
Age, years
Median 57
Mean 55.12±10.08
Range 39-73
Sex
Male 19 (76.0)
Female 6 (24.0)
Enrollment factors
Borrmann IV 3 (12.0)
Borrmann III 20 (80.0)
Bulky N positive 2 (8.0)
Histological classification
Moderately differentiated adenocarcinoma 2 (8.0)
Poorly differentiated adenocarcinoma 23 (92.0)
MSI status
MSS 25 (100.0)
MSI-H 0 (0.0)
PD-L1(28-8)CPS score
<1 5 (20.0)
1-5 13 (52.0)
>5 7 (28.0)
Tumor location
Stomach 25 (80.0)
Carcinoma of esophagogastric junction 5 (20.0)
ECOG
0 7 (28.0)
1 18 (72.0)

Efficacy: Among the 25 pts eligible for preoperative efficacy evaluation, 23 achieved partial response (PR) and 2 had stable disease (SD), resulting in an overall response rate (ORR) of 92% and a disease control rate (DCR) of 100%. The rate of R0 resection was 100%. Six cases were diagnosed with pathological complete response (pCR). Nine cases were diagnosed with major pathologic response (MPR). The pCR rate (TRG 0) and MPR rate (TRG 0-1) were 24% and 36% respectively. The TRG 0-2 rate were 88%. Safety and tolerability: The incidence of adverse events (AEs) was 100%. The most common hematologic AEs were leukopenia (72.0%) and granulocytopenia (72.0%). The most common nonhematologic AEs included fatigue (80.0%) and cutaneous adverse reactions (28.0%).

Conclusions

Tislelizumab combined with apatinib and oxaliplatin plus S1 chemotherapy showed clinical benefits in Borrmann IV large Borrmann III type and Bulky N positive advanced GC, with acceptable safety profile.

Clinical trial identification

NCT05223088.

Legal entity responsible for the study

The authors.

Funding

BeiGene.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

86P - Efficacy and safety of IBI110 (anti-LAG-3 mAb) in combination with sintilimab (anti-PD-1 mAb) in advanced squamous non-small cell lung cancer (sqNSCLC): updated results of the phase Ib study (ID 178)

Presentation Number
86P
Lecture Time
12:30 - 12:30
Speakers
  • Caicun Zhou (Shanghai, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Although PD-1/L1 inhibitors have shown efficacy in advanced/metastatic sqNSCLC, many patients (pts) do not achieve robust response or durable benefit to this treatment. Hence, novel therapies that meet this requirement are needed. The preliminary results of IBI110 combined with sintilimab and chemotherapy (paclitaxel plus carboplatin, TP) (combination therapy) in pts with treatment-naive sqNSCLC were previously presented (Caicun Zhou et al. ASCO 2022). Here, we report updated results of this phase Ib study.

Methods

Eligible pts with previously untreated, unresectable, locally advanced/metastatic sqNSCLC were enrolled. Pts received IBI110 combined with sintilimab and TP for 4-6 cycles. The primary objectives were to evaluate the safety, tolerability, and efficacy of the combination therapy.

Results

As of 19th August, 2022, we reanalyzed the data of 20 pts previously reported. The most common TRAEs included anemia (65%), alopecia (60%), white blood cell count decreased (55%), asthenia (45%), aspartate aminotransferase increased (45%), rash (45%), neutrophil count decreased (40%). The most common TRAEs ≥ grade 3 were neutrophil count decreased (35.0%), and white blood cell count decreased (20.0%). Immune-related AEs (irAEs) occurred in 14 pts (4 pts: grade ≥ 3). The safety profile was consistent with the primary analysis. The updated ORR was also 80% (16/20, 15 patients with ≥ 2 efficacy assessments were confirmed PR and 1 pt was unconfirmed PR). The 6-month PFS rate was 75.0% (95% CI, 50.0-88.7), 9 pts reached PFS events while 11 pts had the continuous benefit (median follow-up time: 9.9 mos), and 12 pts had PFS ≥ 8 mos. The median PFS was not reached.

Conclusions

IBI110 combined with sintilimab and chemotherapy in advanced sqNSCLC continues showing robust anti-tumor activity and favorable safety.

Clinical trial identification

NCT04085185.

Legal entity responsible for the study

Innovent Biologics, Inc., Suzhou, Jiangsu, China.

Funding

Innovent Biologics, Inc., Suzhou, Jiangsu, China.

Disclosure

J. Sun, H. Wang, Y. Chen: Financial Interests, Personal, Sponsor/Funding, Employment: Innovent Biologics, Inc. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

87P - Infliximab use in patients with checkpoint inhibitor toxicities - a tertiary centre experience (ID 179)

Presentation Number
87P
Lecture Time
12:30 - 12:30
Speakers
  • Shuai H. Zhang (London, United Kingdom)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immune checkpoint inhibitors (ICIs) have revolutionised the treatment of multiple cancers. However, they are associated with a spectrum of immune-related adverse events (irAEs). Infliximab is a recommended treatment for corticosteroid refractory irAEs, but real-world data is limited.

Methods

We conducted a retrospective review of patients in our institution who received infliximab for irAEs between May 2021 to July 2022. Data was correlated against internal guidelines which require upfront testing for TB, HIV, VZV and Hepatitis B and C.

Results

Over this period, 31 patients received infliximab (male n=20). Median age 66.7 years (range: 33-86). Seventeen patients (54.8%) had melanoma, 6 (19.4%) non-small-cell lung cancer, 4 (12.9%) renal cancer, 3 (9.7%) gastrointestinal cancer and 1 (3.2%) uterine cancer. Eighteen (58.1%) patients received ipilimumab/nivolumab combination, 7 (22.6%) pembrolizumab containing regimes, 3 (9.7%) nivolumab, 2 (6.5%) durvalumab and 1 (3.2%) patient received ipilimumab single agent. Highest grade irAE was Grade 3 in 71.0% (n=22), G2 in 19.4% (n=6), G4 in 6.5% (n=2) and G5 in 3.2% (n=1). Gastrointestinal irAE accounted for 87.1% (n=27) of the cohort, 9.7% of patients (n=3) had rheumatological irAE, with 1 case of pneumonitis. The median number of ICI cycles prior to infliximab treatment was 3 (range 1 to 29). Median number of infliximab doses was 2 (range 1 to 3). ICI treatment was discontinued after irAE requiring infliximab in 25 patients (80.6%). All patients received corticosteroids before infliximab. Twenty-four patients (77.4%) were tested for TB (T-Spot test) prior to infliximab initiation. All patients tested received negative results. Testing rates for Hepatitis B and C was 100%, while 93.5% of patients (n=29) were tested for HIV. Testing rates for VZV was 58.1% (n=18). Two patients developed shingles post infliximab and were treated with aciclovir. No patients experienced reactivation of TB or hepatitis post infliximab treatment.

Conclusions

In our institution, most patients treated with infliximab had received ICI therapy for melanoma. The most frequent indication for infliximab was gastrointestinal toxicity. Aside from shingles, no post-infliximab treatment infections were reported in this cohort.

Legal entity responsible for the study

The Royal Marsden NHS Foundation Trust.

Funding

Has not received any funding.

Disclosure

D. Cunningham: Financial Interests, Institutional, Research Grant: MedImmune/AZ, Clovis, Eli Lilly, 4SC, Bayer, Celgene, Leap, Roche; Non-Financial Interests, Personal, Advisory Role: OVIBIO. I. Chau: Financial Interests, Personal, Advisory Board: Bristol Myers Squibb, Eli-Lilly, MSD, Roche, Merck-Serono, AstraZeneca, OncXerna, Pierre Fabre, Boehinger Ingelheim, Astella, Incyte, GSK, Sotio, Daiichi Sankyo, Eisai; Financial Interests, Personal, Other, DMC chairman: Five Prime Therapeutics; Financial Interests, Personal, Invited Speaker: Eisai, Eli-Lilly, Servier; Financial Interests, Institutional, Invited Speaker: Cilag-Janssen, Eli Lilly. N. Starling: Financial Interests, Personal, Advisory Board: GSK, Novartis, MSD Oncology, Servier, AstraZeneca, Pfizer; Financial Interests, Personal, Invited Speaker: Clinical Options, Eli Lilly, Pierre Fabre, Amgen, Merck Serono; Financial Interests, Institutional, Research Grant, Sept 2017 (24m) Paid to institution research: Merck; Financial Interests, Institutional, Research Grant, Nov 2017 (48m) -Paid to institution research fund: AstraZeneca; Financial Interests, Institutional, Research Grant, Jan 2018 - Paid to institution research fund: Pfizer; Financial Interests, Institutional, Research Grant, July 2018 (36m) Paid to institution research fund: BMS. S. Popat: Financial Interests, Personal, Advisory Board: Boehringer Ingelheim, Novartis, Amgen, Janssen, Daiichi Sankyo, AstraZeneca, Bayer, BMS, Blueprint, Merck Serono, Guardant Health, Beigene, Takeda, Lilly, Seattle Genetics, Turning Point Therapeutics, Xcovery, GlaxoSmithKline, MSD, Pfizer, Sanofi; Financial Interests, Personal, Expert Testimony: Roche; Financial Interests, Personal, Invited Speaker: Medscape, VJ Oncology, Touch Medical; Financial Interests, Personal, Other, Journal Deputy Editor, Lung Cancer: Elsevier; Financial Interests, Institutional, Other, Sub-investigator: Amgen; Financial Interests, Institutional, Invited Speaker: Ariad, AstraZeneca, Roche, Boehringer Ingelheim, Celgene, Daiichi Sankyo, GlaxoSmithKline, Takeda, Trizel, Turning Point Therapeutics, Roche, Janssen, BMS, Lilly; Financial Interests, Institutional, Other, Sub-Investigator: MSD, Blueprint, Seattle Genetics; Financial Interests, Institutional, Research Grant: Guardant Health; Non-Financial Interests, Personal, Leadership Role, Chair of Steering Committee, Unpaid: British Thoracic Oncology Group; Non-Financial Interests, Personal, Officer, Thoracic Faculty, Unpaid: European Society of Medical Oncology; Non-Financial Interests, Personal, Leadership Role, Foundation Council Member, Unpaid: European Thoracic Oncology Platform; Non-Financial Interests, Personal, Advisory Role, Mesothelioma Task-force Member, Unpaid: International Association for the Study of Lung Cancer; Non-Financial Interests, Personal, Invited Speaker, Unpaid: Mesothelioma Applied Research Foundation; Non-Financial Interests, Personal, Advisory Role, Honorary Clinical Advisor, Unpaid: ALK Positive UK; Non-Financial Interests, Personal, Advisory Role, Research Advisory Group Member, Unpaid: Ruth Strauss Foundation; Non-Financial Interests, Personal, Advisory Role, Scientific Adivsory Board Member, Unpaid: Lung Cancer Europe. M.E.R. O'Brien: Financial Interests, Personal, Advisory Board, virtual ad board: Puma, Sanofi; Financial Interests, Personal, Advisory Board: iTeos, Amgen, MSD, Pierre Fabre; Financial Interests, Institutional, Invited Speaker, we host clinical trials, PI for MSD pearls trial: MSD; Non-Financial Interests, Personal, Advisory Role: Eisai; Other, Personal, Other, Chair this UK research group: NCRI CSG LORD subgroup. A.R. Minchom: Financial Interests, Personal, Advisory Board: Janssen Pharmaceuticals, Merck Pharmaceuticals, Takeda Pharmaceuticals, 4D Pharma , Genmab Pharmaceuticals; Financial Interests, Personal, Other, Honoraria: Chugai Pharmaceuticals, Novartis Oncology, Faron Pharmaceuticals, Bayer Pharmaceuticals, Merck Pharmaceuticals, GSK, Janssen Pharmaceuticals; Financial Interests, Personal, Other, Expenses: Amgen Pharmaceuticals; Financial Interests, Personal, Invited Speaker, Expenses: LOXO Oncology. L.M. Pickering: Financial Interests, Personal, Invited Speaker, Recorded teaching July 2020: BMS; Financial Interests, Personal, Invited Speaker, Group teaching, London, Dec 2019: BMS; Financial Interests, Personal, Advisory Board, Virtual, Oct 2020: BMS; Financial Interests, Personal, Invited Speaker, Virtual teaching to Eisai internal team, July 20: Eisai; Financial Interests, Personal, Advisory Board, Regional Advisory Board, Vienna, Nov 19: MSD; Financial Interests, Personal, Advisory Board, Regional Advisory Board, Munich, Apr 19: MSD; Financial Interests, Personal, Advisory Board, March 2019, London: Novartis; Financial Interests, Personal, Advisory Board, Virtual, Oct 20: Pfizer; Financial Interests, Personal, Invited Speaker, Virtual, Aug 20: Pfizer; Financial Interests, Personal, Invited Speaker, Budapest, Nov 19: Pfizer; Financial Interests, Personal, Advisory Board, London, Oct 19: Pfizer; Financial Interests, Institutional, Funding, Uncommon theme pump priming funds: NIHR; Financial Interests, Institutional, Funding, Tracking renal cancer evolution in blood: Rosetrees Trust; Other, Personal, Other, Charitable funding to support research students and internal departmental clinical research: Kidney and melanoma cancer fund of RMH charity. A. Furness: Financial Interests, Personal, Invited Speaker, Speaker fees: Bristol Myers Squibb, Ipsen, Eisai; Financial Interests, Personal, Advisory Board, Advisory Board: Immuncore; Non-Financial Interests, Personal, Advisory Role: Erase Mesothelioma. S. Turajlic: Financial Interests, Personal, Invited Speaker: IDEA Pharma, Roche, Ventana, MSD, Merck; Financial Interests, Institutional, Funding, Uncommon theme pump priming funds: NIHR; Financial Interests, Institutional, Funding, Digital theme funding: RMH/ICR/BRC/Imperial AHSC/Faculty of Medicine; Financial Interests, Institutional, Funding, Tracking renal cancer evolution in blood: Rosetrees Trust; Financial Interests, Institutional, Funding, Clinical PhD Fellowship over 3 years: CRUK Welcome Trust; Financial Interests, Institutional, Funding, Investigating the relationship between primary melanomas and their metastases: The Robert McAlpine Foundation; Financial Interests, Institutional, Funding, Innovation Grant Award for biomarker development: The Francis Crick Institute; Financial Interests, Institutional, Funding, Developing a novel method of representative tumour in sampling in clinical setting: Ventana; Financial Interests, Institutional, Funding, Mapping clonal evolution in renal cell carcinoma: CRUK training and career development board - clinician scientist fellowship; Financial Interests, Institutional, Funding: Harry J Lloyd Charitable Trust Career Development Award; Financial Interests, Institutional, Funding, Clinical Research Fellowship in melanoma: Andy Quick Charitable fund; Financial Interests, Institutional, Funding, Mechanisms of BRAF resistance: Complete Genomics; Financial Interests, Institutional, Funding, Molecular profiling of non-cutaneous melanoma: CRUK; Financial Interests, Institutional, Funding, Target discovery in acral melanoma: Rosetrees Trust. 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. K. Young: Non-Financial Interests, Personal, Principal Investigator, PI at RMH for Ultimovacs clinical trial: Ultimovacs. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

88P - Hepatic arterial infusion chemotherapy(HAIC) + Lenvatinib(len) and tislelizumab(tis) ±transhepatic arterial embolization(TAE) for unresectable hepatocellular carcinoma(uHCC) with portal vein tumor thrombus(PVTT) and high tumor-burden: A multicenter retrospective study (ID 180)

Presentation Number
88P
Lecture Time
12:30 - 12:30
Speakers
  • WENBO GUO (Guangzhou, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The triple combination therapy of HAIC combined with len. and ICIs showed promising results for advanced uHCC comparing with duplex treatment. However, for high tumor-burden lesions, the triple combination therapy is still insufficient in reduce tumor burden without TAE. This study aimed to assess the efficacy and safety of quadruple combination therapy comparing to the triple combination therapy.

Methods

This retrospective study comprised consecutive patients who had a primary uHCC with PVTT and high tumor-burden (max nodule diameter >10cm) from Jan 2019 to Feb 2022 in four Chinese medical centers. These pats were treated with either quadruple combination therapy (TAE, HAIC, len and tis; THLP group) or triple combination therapy (HAIC, len and tis; HLP group). HAIC involved oxaliplatin, fluorouracil, and leucovorin (FOLFOX). Efficacy was evaluated according to OS, PFS, ORRDCR. Besides, treatment-related AEs were compared.

Results

In total, 100 patients were included in this study: 50 patients in the THLP group and the remaining 50 patients in the HLP group respectively, which resulted in a 1:1 ratio between two groups. The median follow-up was 10.8 months (range, 4.6-20.2 m). The THLP group showed OS (14.1 vs 11.3 m; hazard ratio [HR] 0.60, 95% CI, 0.37-0.99; p=0.044), PFS (5.6 vs 4.4 month; HR 0.63, 95% CI 0.40-0.98; p=0.039), and a higher objective response rate (mRECIST: 72% vs 52%, p=0.039; RECIST 1.1: 60% vs 40%, p=0.046) than the HLP group. As for the DCR, though there was no significant difference, the THLP group was higher than the HLP group (mRECIST: 88% vs 76%, p=0.118; RECIST 1.1: 86% vs 72%, p=0.086). Grade 3/4 treatment-related AEs were more frequent in the THLP group than in the HLP group including abdominal pain (34% vs 18%), Nausea (36% vs 22%), and elevated transaminases (10% vs 2%), but there AEs were completely controllable and no treatment-related death happened.

Conclusions

Additional TAE to triple combination therapy of HAIC, len and tis might improve prognosis compared with triple combination therapy and had accepted toxic effects.

Legal entity responsible for the study

The authors.

Funding

BeiGene Ltd.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

89P - Durvalumab could be effective in combination with anti-HER2 agents in HER2-low breast cancer (ID 181)

Presentation Number
89P
Lecture Time
12:30 - 12:30
Speakers
  • Maryam Arshad (London, United Kingdom)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The clinical challenge for treating HER2 (human epidermal growth factor receptor 2)-low breast cancer is the paucity of actionable drug targets. However, the discovery of immune checkpoint inhibitors has made immunotherapy an emerging new treatment modality for breast cancer. Moreover, several chemotherapeutic agents are known to induce immunogenic cell death by activating the immune system. Therefore, we hypothesized that modulating the tumour microenvironment using trastuzumab and or trastuzumab deruxtecan (T-Dxd) in breast organoids co-cultured with T-cells might enhance the response to immunotherapy.

Methods

We established a panel of HER2-low breast cancer patient-derived organoids (PDOs), recapitulating the derived tumour. These PDOs were cocultured with immune cells (T- cells and Natural killer cells (NK cells)) and treated with T-Dxd and or trastuzumab in combination with durvalumab. Levels of cytotoxic markers were assessed using flow cytometry and cytokine assays.

Results

Our findings revealed synergistic effects in HER2-low BC patient-derived organoids when treated with T-Dxd and or trastuzumab in combination with durvalumab. We also observed antibody-dependent cellular cytotoxicity (ADCC) response with trastuzumab in combination with durvalumab. These results highlight the need to develop a combination treatment of PD-1/PD-L1 inhibitors with targeted therapies, and other immunotherapies to maximize clinical efficacy.

Conclusions

Altogether, despite preliminary, these findings support the rationale for combining anti-HER2 therapies with immunotherapy in HER2-low BC patients.

Legal entity responsible for the study

M. Arshad.

Funding

King's College London.

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster Display Poster Display session

90P - Long-term outcomes in melanoma patients achieving a complete response under immune checkpoint inhibition (ID 182)

Presentation Number
90P
Lecture Time
12:30 - 12:30
Speakers
  • Eftychia Chatziioannou (Tuebingen, Germany)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immune checkpoint inhibition (ICI) has changed the landscape of melanoma therapy. However, not much is known about the long-term outcomes and durability of response in patients who experienced a complete response (CR) under ICI. It is important to monitor this patient population due to the financial toxicity and the immune-related adverse events (irAEs) associated with prolonged treatment.

Methods

We evaluated unresectable stage IV melanoma patients treated with first-line ICI between 2013 and 2018 outside of a clinical trial. Overall survival (OS) and progression-free survival (PFS) were assessed. irAEs occurring after six months of treatment and response to ICI rechallenge were recorded. Additionally, the prognostic value of clinicopathologic features was investigated.

Results

43 (12.1%) patients experienced CR. 40 (93%) patients discontinued therapy. Three (7%) patients are still receiving treatment without having a progressive disease (PD). The median age at the time of CR was 74 [IQR; 66-77] years old. 26 patients (65%) received anti-PD-1 monotherapy and 14 patients (35%) received nivolumab plus ipilimumab. The median follow-up (FUP) after CR was 49 [IQR; 43-57] months. The median treatment duration was 22 [IQR; 17–24] months, and the median time from CR to therapy ending was 10 [IQR; 1–17] months. Median PFS and OS were not reached (95% CI; NR-NR). 5-year PFS and OS after CR was 79% (95% CI; 62%-89%) and 83% (95% CI; 60%-93%), respectively. Age below 77 years at CR (p=0.043) was linked to a better prognosis in the univariable Cox regression. Neither the total length of treatment (p=0.754) nor the treatment duration after CR were associated with prognosis (p=0.398). Second-line ICI was administered to eight patients, and disease control was seen in 5 (64%). 10 (25%) of the patients experienced late toxicities, with two of them being severe.

Conclusions

In our cohort, CR under ICI seems to be a surrogate marker for long-term survival. The duration of treatment was not associated with either PFS or OS, but the optimal duration of therapy in patients achieving CR is not established. Biomarkers are required to help identify patients at risk of relapse among complete responders but also patients who can safely stop therapy after CR.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

U. Leiter-Stoppke: Financial Interests, Personal, Advisory Role: MSD, Novartis, Roche, Sanofi, Sun Pharma; Financial Interests, Institutional, Funding: MSD. I. Thomas: Financial Interests, Personal, Advisory Board: BMS; Financial Interests, Institutional, Invited Speaker: BMS, Pfizer, MSD, Amgen, Novartis, Sanofi, 4SC, AstraZeneca, Biontech, Genentech, Roche, Biotech, Curevac, HUYA, Incyte, Idera, Iovance, Infla-Rx, Kartos, Nektar, Regeneron; Non-Financial Interests, Personal, Member: ADO, DKG, EADO. A. Forschner: Financial Interests, Institutional, Funding: Bristol Myers Squibb-Stiftung Immunonkologie; Financial Interests, Personal, Advisory Role: Bristol-Myers Squibb, MSD Oncology, Novartis, Pierre Fabre, Roche Pharma AG; Financial Interests, Personal, Other: Bristol-Myers Squibb, Novartis, Pierre Fabre, Roche Pharma AG; Financial Interests, Personal, Invited Speaker: CeGaT, MSD Oncology, Bristol-Myers Squibb, Novartis, Roche Pharma AG. L. Flatz: Financial Interests, Personal, Ownership Interest: Hookipa Pharma; Financial Interests, Personal, Invited Speaker: Bristol-Myers Squibb, Novartis, Sanofi; Financial Interests, Personal, Advisory Role: Bristol-Myers Squibb, Novartis, Sanofi; Financial Interests, Institutional, Funding: Hookipa Pharma; Financial Interests, Personal, Royalties: Hookipa Pharma. T.M.S. Amaral: Financial Interests, Personal, Invited Speaker: CeCaVa, BMS, Novartis, Pierre Fabre; Financial Interests, Institutional, Funding: Novartis, Neracare, Sanofi, Skyline-Dx; Financial Interests, Institutional, Research Grant: Novartis, iFIT; Non-Financial Interests, Personal, Member: Portuguese Society for Medical Oncology, Portuguese Society of Medical Oncology - Young Oncologists Group, ASCO; Other, Personal, Other, Clinical expert: Infarmed. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

91P - Association between weight variation and survival in patients treated with immune checkpoint inhibitors (ID 183)

Presentation Number
91P
Lecture Time
12:30 - 12:30
Speakers
  • Raquel M. Romao (Porto, Portugal)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immune checkpoint inhibitors (ICI) have revolutionized cancer treatment, showing impressive clinical benefit in some patients with multiple types of malignancies.

Some of the studies carried out to determine which patients could benefit the most of ICI therapy have indicated that patient nutritional status may impact immune response.

The aim of our study was to explore the associations between weight variation during ICI treatment and its clinical outcomes, as overall survival (OS) and progression free survival (PFS).

Methods

We conducted a single-center retrospective cohort study with advanced cancer patients, that received at least one cycle of ICI in monotherapy at an academic center. The patients were stratified according to their changes of weight during treatment with ICI (Decreasing vs Increasing/Stable). Overall survival (OS) was defined time between start of immunotherapy and death. Proportions were analyzed using Pearson Chi Square, and time to event data with the Kaplan-Meier curves and Cox Proportional Hazards.

Results

143 patients were included with a median age 64 (IQR 54-70) years and 76.9% were male. 35 (24.5%) patients had ECOG PS 0, 95 (66.5%) 1 and 13 (9.1%) 2. Lung cancer was the most common diagnosis (49%), followed by skin (16.1%) and kidney (12.6%) cancers. Median follow-up was 36 months (IQR 48-29). Most patients (134 – 93.2%) received anti-PD-1/anti-PD-L1; 44 (30.8%) were treated at 1st line and 87 (60.8%) at 2nd line. The group whose weight decreased (66-46.2%) experienced shorter median OS than the group whose weight increased/remain stable (77-53.8%) [11 (95% CI, 5.43- 16.57) vs 19 (95% CI, 9.82- 28.2) months, p=0.037]. Median PFS was also shorter for the group whose weight decreased [12 (95% CI, 6.52- 17.48) vs 18 (95% CI, 10.41- 25.59) months]. However, differences in PFS were not statistically significant (p=0.05).

Multivariate analysis revealed that weight loss, ECOG PS 1 or 2, second or further line treatment significantly and independently correlated with poor overall survival.

Conclusions

We observed lower survival in patients with weight loss during ICI treatment. Weight changes could help to detect progression and long-term benefit. The multidisciplinary care of patients and regular nutritional assessment is of utmost importance.

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 Poster Display session

92P - NEoadjuvant multimodality RX including immUnotherapy for highly Selective unresectable locally advanced esophageal squamous cell carcinoma (NEXUS): a prospective, single-arm, phase 2 trial (ID 184)

Presentation Number
92P
Lecture Time
12:30 - 12:30
Speakers
  • Xin Wang (Beijing, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

This study aimed to evaluate the efficacy and safety of preoperative PD-1 inhibitor tislelizumab combined with CRT in unresectable esophageal squamous cell carcinoma (ESCC).

Methods

This is a single-arm, phase II trial, planned to enroll 30 patients. Eligibility criteria include histologically confirmed unresectable thoracic ESCC stage at cT4bNxM0 (AJCC 8th). Radiotherapy was delivered to a total dose of 50Gy/25f, concurrently with cisplatin (25 mg/m2) and nab-paclitaxel (100 mg) (QW, at least 3 cycles). Then followed by two cycles of tislelizumab (200 mg, Q3W), cisplatin (75 mg/m2, Q3W) and nab-paclitaxel (150 mg/m2, Q3W). If curative resection was considered, esophagectomy was performed within 4 weeks. Patients with pathologically residual disease would receive tislelizumab (200 mg, Q3W) for 1 year. The primary endpoint is 1-year progression free survival rate.

Results

From December 2021 to July 2022, 21 patients were enrolled. 16 patients completed chemoradiotherapy and received immunochemotherapy. Reasons for 5 patients who did not receive immunochemotherapy included patient refusal (n=1), progressive disease (n=1), esophageal fistula (n=2), waiting for immunochemotherapy (n=1). Finally, 13 patients proceeded to surgery, with R0 resection rate of 100%. Reasons for not undergoing surgery after immunochemotherapy were esophageal mediastinal fistula (n=1), surgery delay due to COVID-19 epidemic (n=2). The pCR and MPR rate was 61.5% (8/13) and 76.9% (10/13). G1, G2, G3 immune related pneumonia occurred in 3 patients, respectively. ≥G3 AEs occurred in 9 (42.9%) patients. Postoperative complications included anastomotic fistula (2/13), pleural effusion (2/13), pneumonia (1/13), myocardial damage (1/13), delayed wound healing (1/13).

Conclusions

Chemoradiotherapy followed by immunochemotherapy might be a useful conversional treatment option for unresectable ESCC. Esophagectomy after this could be safe with acceptable complications for unresectable locally advanced ESCC.

Clinical trial identification

ChiCTR2100054327.

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 Poster Display session

93P - Cetuximab-based chemotherapy after progression on immune-checkpoint inhibitors in recurrent/metastatic head and neck squamous cell cancer (ID 185)

Presentation Number
93P
Lecture Time
12:30 - 12:30
Speakers
  • Santiago Cabezas-Camarero (Madrid, Spain)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

There is no standard therapy after progression to immune checkpoint inhibitor (ICI) therapy in patients with recurrent/metastatic (R/M) squamous cell carcinoma of the head and neck (SCCHN). Chemotherapy has been shown to increase objective response rates after ICIs. The aim of this study was to evaluate the efficacy and safety of a weekly cetuximab-paclitaxel (PC regimen) after checkpoint inhibitors in R/M SCCHN.

Methods

Retrospective study of patients with R/M SCCHN that received post-ICI therapy with weekly cetuximab [400 mg/m2 (load); 250 mg/m2 (follow)] plus paclitaxel (80 mg/m2) at Hospital Clinico Universitario San Carlos. Overall response rate (ORR), disease control rate (DCR), depth of response (DoR) and survival since the start of cetuximab-paclitaxel and since first-line therapy were evaluated. Safety according to CTCAE were recorded.

Results

Female = 24:8; median age: 67 (Min-Max: 54-95); primary tumor: oral cavity (n=17), oropharynx (n=6), hypopharynx (n=3), larynx (n=4), unknown primary (n=2). ORR (n=28 evaluable): was 71% (20/28) (PR: n=17, CR: n=3). DCR: 79% (22/28). Median PCBTL (n=27 available): -45% (-100% to +31%) SD and PD were. Median follow-up since first-line, since the start of ICI and since the start of PC were 23.5 months (0-71), 18 m (0-71) and 11 m (0-63), respectively. Median overall survival (OS) since first-line was 23 months (95%CI: 18.8-27.2) since the start of ICI was 19 months (95%CI 14-24) and since the start of PC was 12 months (95%CI 8.8-15.1). Median OS with PC in first line (n=21, OS= 12 months (95%CI 7.6-16.4) was no different (P=0.798) than OS with PC in second or further lines (n=11, OS= 12 months (95%CI 6.2 - 17.8). Grade 1-2 AEs occurred in 100% of patients. Grade 3 or 4 AEs developed in 65%, being grade 3 in all of them except in 2 patients (grade 4 neutropenia, grade 4 pneumonia). There were no treatment-related deaths.

Conclusions

Weekly, low-dose, cetuximab-based chemotherapy achieves high and deep response rates with a favorable toxicity profile. These findings as well as its potential impact in survival should be evaluated in larger, prospective studies.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

S. Cabezas-Camarero: Financial Interests, Personal, Invited Speaker: BMS, Merck KgGa, MSD; Financial Interests, Personal, Other, Travel arrangements: Merck KgGa, Janssen; Financial Interests, Institutional, Funding: Merck KgGa; Financial Interests, Personal, Advisory Board: BMS. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

94P - Does hyperprogressive disease predict poor survival? (ID 186)

Presentation Number
94P
Lecture Time
12:30 - 12:30
Speakers
  • Gulin Alkan (Istanbul, Turkey)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Hyperprogressive disease (HPD) is a new phenomenon developing in the era of immune checkpoint inhibitor (ICI) therapy. HPD is characterized by an unexpected and fast progression in tumor volume and poor survival.There is no standardized definition for HPD and clinicopathological variables associated with HPD are unclear. Herein, we assessed incidence and factors predictive of HPD in patients terated with ICIs.

Methods

We retrospectively analysed patients with advanced cancer treated with ICI at Cerrahpasa Medical Faculty Clinical Trial Unit between 2014-2021. We used the Lo Russo’s adopted criteria combined clinical and radiologic parameters for the definition of HPD. All patients who underwent their first tumor evaluation according to RECIST1.1 were included.

Results

Of 155 patients,147 were eligible for analysis. The median age was 61 and 83% were male. The cancer types were; 67,3% lung, 12,9% bladder, 9,5% gastric, 5,4% colon and 4,8% renal cell carcinoma. 59,9% patients were treatment naive and others had one or more lines of chemotherapy. There were 124 patients (84%) who received a single-agent ICI as PD-1 or PD-L1 inhibitors. 23 (16%) patients were treated with combination ICI with anti VEGF, TKI or other ICIs. The median duration of follow-up was 15,6 months. Incidence of HPD was 12,9%. Patients with progressive disease (PD) without HPD were the 20,4%. The median overall survival (mOS) was 3,0 months for HPD patients and 23,1 months for non-HPD patients (p <.001). The mOS for HPD and PD without HPD was 3,0 vs 7,7 months respectively (p <.001). In the univariate analysis (HPD vs non-HPD group) baseline sum of diameters of the lesions (p=0.040) and the baseline high LDH levels (p=0.040) were associated with HPD.

Conclusions

With increased use of ICIs, early recognition of HPD that is predicting very poor survival, will provide insight for the physicians in the management of cancer patients.

Legal entity responsible for the study

G. Alkan.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

95P - A phase II study of combination of H101 (a recombinant human adenovirus type 5) and nivolumab for advanced hepatocellular carcinoma (HCC) after systemic therapy failure (ID 187)

Presentation Number
95P
Lecture Time
12:30 - 12:30
Speakers
  • Jing Xie (Shanghai, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

HCC has a mortality/morbidity ratio of 0.98 and a five-year survival rate of only about 5%-6%, indicating a very poor prognosis. Nivolumab is a programmed death receptor-1 (PD-1) inhibitor. H101, an E1B gene deleted oncolytic adenovirus, is known to have significant antitumor activity. In addition, local injection of H101 might enhance the effect of antitumor therapies (chemotherapy and radiotherapy). We report on the safety and efficacy of H101 in combination with nivolumab in patients (pts) with advanced HCC.

Methods

This single-arm, phase II study enrolled pts with HCC who failed prior systemic therapy. The combined treatment period starts from day 8, which will be recorded as the first cycle. Eligible pts received i.v. H101 (2 vials, on day 8) and i.v. nivolumab (3mg/kg, on day 9) followed by every 2 weeks. While H101 could be injected once every 4 weeks or suspended, depending on the percentage of CD8+ lymphocytes. Treatment continued until disease progression, unacceptable toxicity, consent withdrawal, or the physician’s decision. . The primary endpoint was objective response rate (ORR) per RECIST v1.1. and 6-month OS% was the other co-primary endpoint. The secondary endpoints included progression-free survival (PFS), overall survival (OS), disease control rate (DCR), duration of response (DOR), safety and exploratory bioinformatics analysis.

Results

Up to Mar 2022, 21 pts were enrolled in cohorts. Of 18 evaluable pts, confirmed ORR and DCR were 11.1% and 38.9% respectively, with 2 partial responses (PR) and 5 stable diseases (SD), and 6-month OS% was 77.8%. Median PFS was 2.27 months [95% confidence interval (CI), 1.44-3.09], median OS was 15.04 months [95% CI, 8.33-21.76] and median DOR was 6.51 months. The most common treatment-emergent adverse events (TEAEs) in all pts were low-grade fever (90%) and pain related to centesis (60%). No grade 4/5 adverse events were reported.

Conclusions

H101 in combination with nivolumab showed promising activity with well-tolerated toxicities in pts with advanced HCC. Updated results will be presented.

Clinical trial identification

MIT-003 Institutional)/CA209-7CE (BMS).

Legal entity responsible for the study

Z. Meng.

Funding

Bristol Myers Squibb.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

96P - Long-term outcomes after initial disease progression with anti-PD-1 in melanoma (ID 188)

Presentation Number
96P
Lecture Time
12:30 - 12:30
Speakers
  • Kimberly Loo (New York, NY, United States of America)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Long-term melanoma survival is now attainable with anti-PD-1 regimens. Notably, a subset of patients experience disease progression (PD) during their immunotherapy treatment but still survive >5 years. Factors that contribute to this prolonged survival despite initial PD are of great interest for prognostication and treatment selection.

Methods

We conducted a retrospective study of patients who survived >5 years from initial treatment with anti-PD-1 with advanced non-uveal melanoma at two centers. Overall survival (OS) was calculated from 5-years post initial anti-PD-1 treatment. Time to second progression was calculated from the date of initial progression on anti-PD-1 to date of second progression.

Results

Of the 298 patients treated with anti-PD-1 who survived at least 5 years, 102 experienced disease progression. Median patient age was 62 years (range 22-90); 30 had unresectable stage III, 64 M1a, 75 M1b, 98 M1c, and 31 M1d disease. Patients received anti-PD(L)1 monotherapy (176) or nivolumab + ipilimumab (122). The best overall response to initial anti-PD-1 was complete response (150), partial response (92), stable disease (30), or PD (26). Median follow-up among survivors (274) was 79 months (range 60-143). OS at year 6 (1-year post 5-year landmark) was 94.9% (95% CI: 91.4-97.0%). 24 patients died after 5 years post initial anti-PD-1, 10 due to melanoma. In those who survived to 5 years without treatment failure, the probability of remaining treatment failure-free for an additional 2 years was 96.5% (95% CI: 93.4-99.6%). Among the 102 patients that progressed on initial anti-PD-1, 56 progressed at existing sites, 28 at new sites, and 18 at both. 6 patients experienced initial PD after 5 yrs. Probability of remaining free from progression at 5 and 10 years from initial anti-PD-1 was 67.8% (95% CI: 62.2-72.8%) and 64.4% (95% CI: 58.2-69.8%), respectively. 69 patients had a second progression; median time to second progression from initial progression was 36 (95% CI: 21-58) mos.

Conclusions

Patients who progress on initial anti-PD-1-based regimens but survive 5+ years have promising overall survival and low rates of second progression. These data help guide survivorship planning in patients who progress on anti-PD-1 and go on to respond to later lines of therapy.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

K. Panageas: Financial Interests, Personal, Stocks/Shares: AstraZeneca, Catalyst Biotech, Dynavax Technologies, Pfizer, Sunesis Pharmaceuticals, Viking Therapeutics. D. Johnson: Financial Interests, Personal, Advisory Board: BMS, Catalyst, Iovance, Oncosec, Merck, Mallinckrodt, Pfizer, Novartis, Mosaic, Targovax; Financial Interests, Institutional, Funding: BMS; Financial Interests, Institutional, Invited Speaker: Incyte; Non-Financial Interests, Personal, Other, Guidelines: SITC; Non-Financial Interests, Personal, Other, Guidelines: NCCN. A. Betof Warner: Financial Interests, Personal, Advisory Role: Iovance Biotherapeutics, Novartis, Shanghai Jo'Ann Medical Technology, BluePath Solutions, Pfizer, Instil Bio, Lyell Immunopharma, Bristol-Myers Squibb/Medarex. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

97P - Toxicity of bispecific antibody glofitamab in patients with aggressive B-cell non-Hodgkin lymphoma in real clinical practice (ID 189)

Presentation Number
97P
Lecture Time
12:30 - 12:30
Speakers
  • Liudmila Fedorova (Saint-Petersburg, Russian Federation)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Anti CD20/CD3 bispecific antibody glofitamab (G) demonstrated high efficacy and acceptable toxicity profile in patients with aggressive r/r B-NHL. Nevertheless, the number of patients and conditions in clinical trials is limited, which requires further study of the G safety in real clinical practice.

Methods

This study included 28 pts with r/r B-NHL who were treated with G from May 2021 to August 2022 within the Russian Named Patient Program. G was prescribed in escalated regimen: 2.5 mg D8C1, 10 mg D15C1, 30 mg D1C2-12. Anti-CD20 antibody was administrated in D1C1. Efficacy was analyzed by PET-CT (Lugano criteria). Adverse events (AEs) were graded according to NCI CTCAE 5.0.

Results

Median age at G initiation was 50 (21-83), male/female ratio - 11/17 (39/61%). Median number of therapy lines before G was 3 (2-8). ECOG>1 at G initiation was in 7 (25%), B symptoms in 6 (21%) and bulky disease in 8 (29%) pts. Median follow-up was 6 (1-16) mo. At analysis 22 (79%) pts discontinued therapy due to PD (n=10, 36%), severe COVID-19 (n=5, 18%), therapy completion (n=5, 18%), other reason (n=2, 7%). Median number of cycles was 6 (1-12). ORR was 67% (56% CR, 11% PR). Eight pts died during G therapy including 5 (18%) pts due to PD. AEs were present in 27 (96%) pts including gr 3-4 in 14 (43%) and gr 5 in 3 (11%) pts (Table). Any grade COVID-19 was revealed in 9 (32%) pts. Three (11%) pts died due to severe COVID-19. Anti-SARS-CoV-2 antibodies were introduced in 14 (50%) pts: in 6 (21%) pts after Covid-19 and in 8 (29%) pts as a Covid-19 prophylaxis. There were no cases of severe Covid-19 after prophylaxis with antibodies. Other viral infections have also been observed: gr 1-2 VZV in 3 (11%), gr 4 CMV pneumonia in 1 (4%) pts.

All AEs 27 (96%)
Any grade Grade 3-4 Grade 5
Hematologic AEs
Neutropenia 17 (61%) 8 (29%) no
Anemia 14 (50%) 3 (11%) no
Thrombocytopenia 7 (25%) 1 (4%) no
Infection
Covid-19 10 (36%) 4 (14%) 3 (11%)
VZV 3 (11%) no no
CMV pneumonia 1 (4%) 1 (4%) no
Immunological
CRS 15 (54%) 1 (4%) no
Tumor flare 6 (21%) 2 (8%) no
Other
Headache 1 (4%) 1 (4%) no

Conclusions

G demonstrated high efficacy in patients with r/r B-NHL. However, the wide range of toxicities has also been demonstrated including a high rate of viral infections. Anti-SARS-CoV-2 antibodies and standard viral prophylaxis should be considered in this patient group.

Legal entity responsible for the study

Liudmila Fedorova, Kirill Lepik, Olesya Smykova, Natalia Mikhailova, Marina Popova, Vladislav Markelov, Elena Kondakova, Ivan Moiseev, Alexander Kulagin.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

98P - First-line atezolizumab/durvalumab plus platinumÐetoposide combined with radiotherapy in extensive-stage small-cell lung cancer (ID 190)

Presentation Number
98P
Lecture Time
12:30 - 12:30
Speakers
  • Anhui Shi (Beijing, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immunotherapy has made significant advances in the treatment of extensive-stage small-cell lung cancer (ES-SCLC), but data in combination with radiotherapy are scarce. This study aims to assess the safety and efficacy of chemoimmunotherapy combined with thoracic radiotherapy in patients with ES-SCLC.

Methods

This single-center retrospective study analyzed patients with ES-SCLC who received standard platinum–etoposide chemotherapy combined with atezolizumab or durvalumab immunotherapy as induction treatment, followed by consolidative thoracic radiotherapy (CTRT) before disease progression in the first-line setting. Adverse events during radiotherapy with or without maintenance immunotherapy and survival outcomes were assessed.

Results

Between December 2019 and November 2021, 36 patients with ES-SCLC were identified to have received such treatment modality at one hospital. The number of metastatic sites at diagnosis was 1–4. The biological effective dose of CTRT ranged from 52Gy to 113Gy. Only two patients (6%) developed grade 3 toxic effect of thrombocytopenia, but none experienced grade 4 or 5 toxicity. Four patients developed immune-related pneumonitis during the induction treatment period but successfully completed later CTRT. The rate of radiation-related pneumonitis was 8% with grades 1–2 and well tolerated. The median progression-free survival (PFS) was 12.8 months, but the median overall survival (OS) was not determined. The estimated 1-year OS was 80.2% and 1-year PFS was 53.4%.

Conclusions

Immunotherapy combined with CTRT for ES-SCLC is safe and has ample survival benefit.

Legal entity responsible for the study

The authors.

Funding

CSCO-Linghang Cancer Research No. Y-2019AZMS-0519.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

99P - Sintilimab plus nab-paclitaxel in platinum-refractory head and neck squamous cell carcinoma: a phase 2 trial (ID 191)

Presentation Number
99P
Lecture Time
12:30 - 12:30
Speakers
  • Shengyu Zhou (Bejing, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The prognosis of patients with platinum-refractory head and neck squamous cell carcinoma (HNSCC) was poor. Sintilimab is an immune checkpoint inhibitor. Nab-paclitaxel is a nanoparticle taxane that showed activity in platin-resistant HNSCC. This study aimed to evaluate the efficacy and safety of sintilimab plus nab-paclitaxel in patients with platinum-refractory HNSCC.

Methods

This study is a single-arm, open-label, single-center phase II clinical study. Patients with R/M HNSCC who have failed platinum-based therapy were enrolled. All patients were treated with sintilimab(200mg iv d1) and nab-paclitaxel(120mg/m2 iv d1 and d8) every 3 weeks up to 6 cycles and then maintained with sintilimab until disease progression, death, or dose-limiting toxicities. The primary endpoint was the objective response rate (ORR). The secondary endpoints included progression-free survival (PFS), overall survival(OS), and safety.

Results

From April 2019 to April 2022, 15 patients were enrolled. The median age was 55 years (range34-67), and 11 patients were male. Primary site included the oropharynx (1, 6.7%), nasopharynx (6, 40.0%), larynx/hypopharynx (5, 33.3%) and oral cavity (3, 20.0%). Presence of distant metastases(11, 73.3%); prior platinum-based chemotherapy(15, 100%), radiation (11, 73.3%), EGFR targeted therapy (6, 40.0%); ECOG PS =1 (15, 100%); PD-L1 CPS≥10 (6, 40.0%), not applicable (7, 46.7%). The ORR was 20.0% (95% CI, 7.1-45.2%), DCR was 80.0% (95%CI 54.8-92.9%). With a median follow-up time of 12.1 months, 8 patients experienced disease progression, and 5 died. The median PFS was 14.4 (95% CI, 4.0-24.7) months. The median OS was not reached. Common treatment-related adverse events (TRAEs) were hypothyroidism(26.7%), leukopenia(20.0%), pneumonia (20.0%), and nausea (13.3%). Most of the TRAEs were grade 1 or 2. Only one patient experienced grade 3 hypothyroidism.

Conclusions

Sintilimab plus nab-paclitaxel is well-tolerated with very encouraging clinical activity in platinum-refractory HNSCC and warrant further exploration in this disease.

Clinical trial identification

NCT03975270, June 5, 2019.

Legal entity responsible for the study

The authors.

Funding

Innovent biologics Inc.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

100P - Immunotherapy around the clock: impact on stage IV melanoma (ID 192)

Presentation Number
100P
Lecture Time
12:30 - 12:30
Speakers
  • Lisa Goncalves (Lisbon, Portugal)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immunotherapy is currently the standard of care in the treatment of metastatic melanoma. Immune cells in the tumor microenvironment play a decisive role in tumor growth and response to therapy. NK and dendritic cells, monocytes, T and B lymphocytes exhibit circadian oscillations in the peripheral blood, as well as PD-L1 expression. Recent data from MEMOIR study suggests the effectiveness of immune checkpoint inhibitors in melanoma is lower when more than 20% of infusions are in the late afternoon.

Methods

Retrospective, unicentric, cohort study of stage IV melanoma patients under immunotherapy (ipilimumab, nivolumab or pembrolizumab) with PS 0-1, followed at our center between July 2016 and March 2022. Infusion times were obtained and dichotomized as morning (8am-2pm) or afternoon (2pm-8pm). Time to event outcomes were calculated using the Kaplan Meier method and tested using Cox regression model, using a 95% confidence interval (IC). Objective: To determine the impact of immunotherapy administration timing on the overall survival (OS) of patients with metastatic melanoma.

Results

In this time period, 73 patients were treated, and 37.0% of patients had at least three fourths (75%) of the immunotherapy infusions in the afternoon period. The median OS of the population was 24.2 months [CI 95%, 9.04 to 39.8), with a median follow-up time of 15.3 months. No significant demographic or tumor burden differences were found between the morning and afternoon groups. Having more than 75% of immunotherapy infusions in the afternoon results in a shorter median OS (13.8 vs 38.1 months; HR 1.94 [CI 95% 1.01 to 3.74]; p<0,01).

Conclusions

This study suggests that increasing the number of treatments in the afternoon period worsens the outcome of metastatic melanoma patients. Chrono-immunotherapy is a developing topic and could lead to higher survival rates in metastatic melanoma. Prospective randomized studies on immunotherapy timing should be performed.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

A.R. Teixeira Sousa: Financial Interests, Personal, Invited Speaker: Roche, Merck Sharp & Dome, Novartis, Pierre-Fabre, Bristol-Myers Squibb, AstraZeneca, Glaxosmith, Lilly, Pfizer, Tesaro. A.B. Mansinho: Financial Interests, Personal, Invited Speaker: Amgen, Astellas, Bayer, BBraun, Bristol Myers-Squibb, Janssen, Merck-Serono, Merck Sharp & Dohme, Novartis, OMPharma, Pfizer, Pierre Fabre, Roche, Servier; Financial Interests, Personal and Institutional, Funding: Bayer, BBraun. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

101P - Safety and effectiveness of pembrolizumab combined with albumin-bound paclitaxel and nedaplatin as first-line treatment in advanced esophageal squamous cell carcinoma patients (ID 193)

Presentation Number
101P
Lecture Time
12:30 - 12:30
Speakers
  • Fang Yan (Shanghai, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The value of immune checkpoint inhibitor (ICI) combined with chemotherapy in the first line treatment of locally advanced/metastatic esophageal cancer has been confirmed by several clinical studies with the regimens of 5-fluorouracil plus cisplatin(FP)or Paclitaxel plus cisplatin (TP) which were most commonly used in the protocol. However, retrospective study had shown that nedaplatin combined with nab-paclitaxel was more effective than other chemotherapy regimens with fewer adverse events. Here we report the efficacy and safety of ICI combined with nedaplatin and nab-paclitaxel in ESCC.

Methods

Clinical outcomes of 35 patients with metastatic ESCC in Changhai hospital from March 2020 to September 2021 were included in this study. All patients received pembrolizumab 200mg on day 1, albumin-bound paclitaxel 130 mg/m2 on day 1 and 8, and nedaplatin 70 mg/m2 on day 1. The treatment was repeated every 21 days. Evaluation of tumor response was performed according to Response Evaluation Criteria in Solid Tumors 1.1 (RECIST1.1). Toxicities were graded using version 5.0 of the National Cancer Institute Common Toxicity Criteria (NCI-CTC).

Results

All patients were available for evaluation. Of the 35 patients, 4 patients (11.4%) had complete response(CR), 21 patients (60.0%) were partial response (PR), 10 patients (28.6%) achieved stable disease (SD) and no patients had progression disease (PD). The objective response rate (ORR) and disease control rate (DCR) were 71.4% and 100% respectively. The median progression free survival (PFS) was 13.4 months. Main toxicities include hematological toxicity, thyroid dysfunction, rash, fever, arthralgia, myalgia and alopecia. Treatment-related adverse events of grade 3 or higher occurred in 3 patients (8.6%).

Conclusions

Pembrolizumab plus albumin-bound paclitaxel and nedaplatin as first-line treatment demonstrated promising anti-tumor activity and manageable safety in patients with advanced ESCC. Randomized trials to evaluate this new combination strategy 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 Poster Display session

102P - A Multicenter Retrospective Cohort Study Comparing the Efficacy and Safety of Lenvatinib in Combination with PD-1 Inhibitor with or without Transarterial Chemoembolization in Patients with unresectable Hepatocellular Carcinoma (ID 194)

Presentation Number
102P
Lecture Time
12:30 - 12:30
Speakers
  • Guang Tan (Dalian, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Through clinical practice, lenvatinib-based combination treatments are commonly used in patients with unresectable hepatocellular carcinoma (uHCC), but their curative effect warrants further investigation. This study compares the efficacy and safety of lenvatinib plus PD-1 inhibitor and TACE (LPT) vs. lenvatinib plus PD-1 inhibitor (LP) in uHCC.

Methods

Between January 2019 and June 2022, clinical data for patients with uHCC treated with LPT or LP were reviewed from multicenter in China. The treatment effects, overall survival (OS), progression-free survival (PFS), time to failure (TTF), and treatment-related advent events (TRAEs) are compared between the groups.

Results

A total of 63 patients were enrolled, including 27 in LPT and 36 in LP. The median follow-up time was 16.17 months and 13.93 months. Patients in the LPT group had a significantly higher ORR (66.70% vs. 37.00%, P=0.02*) and a considerably longer OS (HR=0.29, 95%CI 0.09-0.87, P=0.03*), PFS (HR=0.39, 95%CI 0.18-0.82, P=0.01*), and mTTF (14.47 months vs. 9.87 months, P=0.049*) than the LP group. The LPT group had significantly higher 1-year and 2-year OS rates (93.87% vs. 71.25%,82.09% vs. 42.22%). A comparable 1-year and 2-year PFS rates advantages were reported in the LPT group (74.14% vs. 47.17%, 52.66% vs. 8.85%). Among the subgroup analysis, we revealed that in non-surgery patients, the LPT group (n=26) had superior OS (HR=0.27, 95%CI 0.07-0.95, P=0.04*) and PFS (HR=0.35, 95%CI 0.14-0.85, P=0.01*) to the LP group (n=19) (1-year OS rate 91.09% vs. 63.66%,1-year PFS rate 65.84% vs. 34.63% ). The total incidence and severity of adverse events were similar in both groups (63.90% vs. 66.70%, P=0.82). Multivariate analysis showed that the choice of LP and hepatic vein thrombus were the independent risk factors for OS, whereas LP was the independent risk factor for PFS.

Conclusions

For uHCC, our research found that LPT therapy was safe and beneficial. In terms of effectiveness and survival time, the LPT therapy model outperforms the LP mode. The potential of the LPT should be evaluated in prospective cohort investigations.

Legal entity responsible for the study

The First Affiliated Hospital of Dalian Medical University.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

103P - Phase IIIb study of durvalumab plus platinumÐetoposide in first-line treatment of Chinese extensive-stage small-cell lung cancer (ORIENTAL): preliminary safety and efficacy results (ID 195)

Presentation Number
103P
Lecture Time
12:30 - 12:30
Speakers
  • Ying Cheng (Changchun, Ji, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

First-line durvalumab (D) plus platinum–etoposide (EP) significantly improved overall survival versus chemotherapy group in CASPIAN phase III study. We initiated a phase IIIb study to assess the safety and efficacy of D+EP in Chinese patients with ES-SCLC.

Methods

ORIENTAL is a single arm, multicenter, phase IIIb study. Treatment-naive ES-SCLC with ≥18 years of age and ECOG PS 0-2 were eligible. Durvalumab (1500 mg) was concurrently administered with first-line EP every 3 weeks for 4 to 6 cycles, followed by Durvalumab maintenance every 4 weeks until progressive disease or unacceptable toxicity. The primary endpoint was immune-mediated adverse events (imAE), the secondary endpoints included progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), overall survival (OS) and adverse events (AEs).

Results

Between Nov. 2020 and Aug. 2021, 151 eligible patients from 32 sites in China were enrolled and received study treatment. At the data cut-off (Feb. 28th, 2022), 38 (25%) patients were still on study treatment. The median age was 66 years (range 43-82). 86.1% of patients were males, 4% were ECOG PS 2, and 13.2% had brain metastasis. The median cycle number of durvalumab was 7 (1-18). Any-cause AEs occurred in 97.4% of patients, and the incidence of imAE was 21.9% [95%CI 15.5-29.3]. AEs ≥ grade 3 occurred in 46.4% of patients, the most common ones were anemia (17.2%), platelet count decreased (7.3%), myelosuppression (6.0%), neutrophil count decreased (4.0%), white blood cell count decreased (3.3%), and hypokalemia (2.6%). There were discontinuations of study treatment in 12 patients (7.9%) and 5 deaths (3.3%) due to treatment-related AEs. ORR was 75.5% [95%CI 67.8-82.1], including two CRs. The median PFS was 6.3 months [95%CI 5.8-6.5), with a median follow-up of 5.7 months. The median OS was not reached yet [95%CI 9.6-NA].

Conclusions

ORIENTAL is the largest study to evaluate the safety and efficacy of D+EP in Chinese ES-SCLC patients to date. The preliminary safety profile and efficacy data are consistent with the CASPIAN study and support D+EP as the standard of care in ES-SCLC.

Clinical trial identification

NCT04449861.

Legal entity responsible for the study

The authors.

Funding

AstraZeneca China.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

104P - Camrelizumab combined with chemotherapy and apatinib as first-line therapy for extensive-stage small cell lung cancer: a phase II, single-arm, exploratory research (ID 196)

Presentation Number
104P
Lecture Time
12:30 - 12:30
Speakers
  • Yanbin Zhao (Harbin, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Small-cell lung cancer is an aggressive tumor type with limited therapeutic options and poor prognosis. It is worthy to explore the new treatment pattern in consideration of the rapid disease progression. Therefore, this study aims to explore the effectiveness and safety of camrelizumab combined with chemotherapy and apatinib in the first-line treatment of extensive-stage small cell lung cancer (ES-SCLC).

Methods

In this Ⅱ study, 40 patients with pathological diagnosis of ES-SCLC who haven’t received prior systemetic therapy plan to be enrolled. The enrolled patients receive camrelizumab (200 mg, iv, q3w) combined with etoposide (80-100 mg/m2, iv, q3w, 4-6 cycles) and platinum drugs (selected by the researcher according to the patients, iv, q3w, 4-6 cycles) followed by maintenance with camrelizumab and apatinib (250 mg, QD). The primary endpoint is 6-month progress-free survival (6-month PFS) rate. Secondary endpoints are objective response rate (ORR), disease control rate (DCR), progression-free survival, overall survival and safety.

Results

Up to September 17, 2022, 13 patients with a median age of 63 years were enrolled. The median treatment duration was 105 days. Among them, 13 patients were available for efficacy analysis, of which 10 patients achieved partial response, and 1 had stable disease. The 6-month PFS rate in evaluable patients was 54.55% (6/11). The ORR was 76.92% and DCR was 84.62%. The adverse reactions included reduction of proteinuria (23.0%), hemoptysis (8.0%), hypothyroidism (8.0%), abnormal liver function (38.0%), skin-related adverse reactions (38.0%), nausea (92.0%), fatigue (8.0%), lymphopenia (15.0%), leukopenia (15.0%) and anemia (54%). During the course of therapy, the majority of reported adverse events were grade 1-2 in severity. Of the 13 patients, only 1 patient experienced grade 3 treatment-related adverse event (anemia). All the adverse events can be controlled and alleviated after symptomatic treatment.

Conclusions

Camrelizumab in combination with chemotherapy and apatinib provided significant benefit and controllable security, supporting this combination as a new first-line treatment option for this population.

Clinical trial identification

ChiCTR2100046355.

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 Poster Display session

105P - Neoadjuvant tislelizumab combined with chemoradiotherapy for resectable locally advanced esophageal squamous cell carcinoma (ESCC): single arm phase II study (ID 197)

Presentation Number
105P
Lecture Time
12:30 - 12:30
Speakers
  • Peng Jin (Jinan, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

This study aimed to evaluate the safety and efficacy of neoadjuvant tislelizumab combined with chemoradiotherapy in patients with resectable esophageal squamous cell cancer.

Methods

This is a prospective single-arm clinical trial. A total of 19 patients with newly diagnosed resectable esophageal cancer (cT1-2N+ / cT3-4aN0-3 M0) received 2 cycles of tislelizumab (200mg every 3 weeks for 2 cycles) concurrent with chemoradiotherapy (radiotherapy: 41.4Gy in 23 fractions; chemotherapy: Paclitaxel (Albumin bound) 100mg/m2, and Cisplatin 75 mg/m2 once every 3 weeks for 2 cycles.) Radical esophagectomy was performed within 4-6 weeks after neoadjuvant therapy. PET/CT was performed at baseline and before surgery. Primary endpoints included pathological response rate (pCR) and major pathological response rate (MPR), the secondary endpoints were disease free survival and safety. Exploratory endpoints include molecular imaging research to further explore the factors affecting the efficacy of neoadjuvant therapy for esophageal cancer.

Results

Nineteen patients enrolled, all of whom received neoadjuvant tislelizumab combined with chemoradiotherapy. Seventeen patients underwent radical esophagectomy. One patient did not undergo radical chemoradiotherapy due to lymph node metastases after neoadjuvant therapy. One patient died of pneumonia before surgery. Among 17 patients who underwent surgery, R0 was 100% (17/17), 8 patients achieved pCR (47.1%), and 12 patients achieved MPR (70.6%). Most of treatment-related adverse event (TRAE) were grade 1-2, and the most common TRAE was anemia (15, 78.9%). The grade 3 TRAE included 1 leukopenia (5.3%), 1 neutropenia (5.3%), 1 liver damage (5.3%), and 1 elevated cardiac troponin T (5.3%). A significant decrease in SUVmax was observed in both pCR and no-pCR patients after treatment. Baseline SUVmax in no-pCR patients tended to be higher than pCR patients(p=0.0642).

Conclusions

Neoadjuvant tislelizumab combined with chemoradiotherapy for locally advanced ESCC has promising efficacy and good safety.

Clinical trial identification

NCT05323890.

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 Poster Display session

106P - Zanubrutinib in combination with tislelizumab in patients with refractory diffuse large B-cell lymphoma (DLBCL): a phase II study (ID 198)

Presentation Number
106P
Lecture Time
12:30 - 12:30
Speakers
  • Lijie Zuo (Beijing, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Treatment of refractory DLBCL still represents a unique area of unmet need. Previous studies have shown synergistic antitumour effects between Bruton’s tyrosine kinase inhibitors (BTKi) and immune checkpoint inhibitors. However, it lacks data of zanubrutinib and tislelizumab. Herein, we present the premilitary results of an ongoing, multicenter, single-arm phase II study designed to assess the safety and efficacy of zanubrutinib in combination with tislelizumab in refractory DLBCL patients.

Methods

Adult patients diagnosed with DLBCL, with ECOG performance status 0-3 and adequate organ function, who were refractory to at least the first-line of systemic therapies containing rituximab were eligible. Patients who had been exposed to any PD-1/PD-L1 inhibitors or BTKi prior to enrollment were excluded. Each 21-day treatment cycle comprises daily zanubrutinib (160mg bid, d1-d21) in combination with tislelizumab (200mg once, d1). Treatment repeats in the absence of disease progression or unacceptable toxicity, up to 12 months. The primary endpoint is the overall objective response rate (ORR). Secondary endpoints include disease control rate (DCR), duration of response (DoR), time to relapse (TTR), progression-free survival (PFS), overall survival (OS), and safety outcomes assessed by the CTCAE 5.0.

Results

From Nov 1, 2020 to Sept 5, 2022, 10 patients (70% of male) were enrolled in the study, with a median (range) age of 60.5 (54-76) years. During a median follow up time of 11.1 months, 3 patients achieved CR, of whom all were of non-germinal center origin, two of primary lymph nodes and one of primary testis. This combination resulted in an ORR of 30% with a DCR of 50%. Median PFS was 4.885 (range, 0.76-22.3) months. Other outcomes were immature and not formally tested. TRAEs observed were neutropenia (1 of 10 patients), thrombocytopenia (1 of 10 patients), and elevated creatine phosphokinase (1 of 10 patients). All TRAEs were grade 1 or 2.

Conclusions

This combination of zanubrutinib and tislelizumab is well-tolerated with promising clinical response in refractory DLBCL patients. Further exploration is still warranted, especially in patients with non-GCB and specific refractory DLBCL at immune-privileged sites.

Legal entity responsible for the study

S. Zhou.

Funding

Shanghai Lianxiang Public Welfare Foundation.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

107P - Outcomes of patients with metastatic non-small-cell lung cancer (mNSCLC) receiving first-line (1L) immunotherapy (IO) with or without chemotherapy (CT): real-world (RW) evidence vs clinical trial results: CORRELATE (ID 199)

Presentation Number
107P
Lecture Time
12:30 - 12:30
Speakers
  • Solange Peters (Lausanne, Switzerland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Recent data suggest that the overall survival (OS) and progression-free survival (PFS) observed in RW patients with mNSCLC receiving IO regimens may be shorter than that seen in randomised clinical trials (RCTs). This retrospective, observational study describes rwOS and rwPFS (overall and by PD-L1 expression) compared with the outcomes observed in RCTs in patients with mNSCLC.

Methods

Eligible RW patients from the US Flatiron Enhanced Data-Mart database were those who developed stage IV mNSCLC, initiated 1L treatment with IO ± CT between 1 Nov 2016 and 31 May 2021, and met select eligibility criteria of 6 RCTs for IO regimens with a US approval in mNSCLC: KEYNOTE (KN) -024, KN-189, KN-407, IMpower150, CheckMate (CM) 9LA, and CM 227. Patients with brain metastases at diagnosis were excluded. Efficacy-effectiveness factors (EEFs) allowed evaluation of the gaps between RW and RCT outcomes.

Results

Most patient baseline characteristics (e.g., % male, ECOG/WHO performance status [PS], PD-L1 expression, and smoking status) differed by <10% between the RW and RCT datasets. Among patients with ECOG PS 0–1, rwOS and rwPFS were considerably shorter vs RCTs, with EEF ratios between 42–73% and 53–78%, respectively (Table). Similar results were observed by PD-L1 status, as applicable (not shown). Where sample size allowed, expanding the analysis to cohorts of PS 2 and PS 3–4, separately, showed even greater disparities in outcomes (not shown).

RCT DatasetAnalytic N mOS 95% CI OS EEF, % mPFS 95% CI PFS EEF, %
KN-024 pembro RCT 154 30.0 18.3–NE 54.0 10.3 6.7–NE 56.3
RW 796 16.2 13.9–18.7 5.8 4.9–6.7
KN-189 pembro+CT RCT 410 22.0 19.5–25.2 55.9 9.0 8.1–9.9 65.6
RW 1836 12.3 11.3–13.3 5.9 5.6–6.2
KN-407 pembro+CT RCT 278 17.2 14.4–19.7 72.7 8.0 6.3–8.5 77.5
RW 412 12.5 10.1–14.9 6.2 5.4–7.3
IMpower150 atezo+bev+CT RCT400 19.5 17.0–22.2 60.0 8.3 7.7–9.8 67.4
RW 31 11.7 8.1–27.2 5.6 3.6–9.8
CM 9LA nivo+ipi+CT RCT361 15.8 13.9–19.7 69.0 6.4 5.5–7.8 53.1
RW 17 10.9 2.0–NE 3.4 1.2–5.9
CM 227 nivo+ipi RCT396 17.1 15.0–20.2 42.1 5.1 4.1–6.3 76.5
RW 35 7.2 3.0–NE 3.9 1.4–6.9

EEF: estimated as median from RW/median from RCT ×100 m, median (months); NE, not estimable.

Conclusions

IO has been established as the standard of care in mNSCLC; however, RW survival outcomes are considerably shorter than those reported in pivotal RCTs, even for indicated populations in the RW. Despite some limitations of the dataset and the US-only population, our RW findings are broadly consistent with other RW studies, highlighting the unmet need for more effective treatment options for RW patients with mNSCLC.

Editorial acknowledgement

Medical writing support for this abstract, under the direction of the authors, was provided by Gauri Saal, MA Economics, of INIZIO Medical, and was funded by AstraZeneca.

Legal entity responsible for the study

AstraZeneca.

Funding

AstraZeneca.

Disclosure

S. Peters: Financial Interests, Institutional, Advisory Board: Vaccibody, Takeda, Seattle Genetics, Sanofi, Roche/Genentech, Regeneron, Phosplatin Therapeutics, PharmaMar, Pfizer, Novartis, Mirati, Merck Serono, MSD, Janssen, Incyte, Illumina, IQVIA, GlaxoSmithKline, Gilhead, Genzyme, Foundation Medicine, F-Star, Eli Lilly, Debiopharm, Daiichi Sankyo, Boehringer Ingelheim, Blueprint Medicines, Biocartis, Bio Invent, BeiGene, Bayer, BMS, AstraZeneca, Arcus, Amgen, AbbVie, iTheos, Novocure; Financial Interests, Institutional, Invited Speaker: Takeda, Sanofi, Roche/Genentech, RTP, Pfizer, PRIME, PER, Novartis, Medscape, MSD, Imedex, Illumina, Fishawack, Eli Lilly, Ecancer, Boehringer Ingelheim, BMS, AstraZeneca, OncologyEducation, RMEI, Mirati; Financial Interests, Personal, Other, Associate Editor Annals of Oncology: Elsevier; Financial Interests, Institutional, Invited Speaker, MERMAID-1: AstraZeneca; Financial Interests, Institutional, Invited Speaker, MERMAID-2, POSEIDON, MYSTIC: AstraZeneca; Financial Interests, Institutional, Invited Speaker, Clinical Trial Steering committee CheckMate 743, CheckMate 73L, CheckMate 331 and 451: BMS; Financial Interests, Institutional, Invited Speaker, RELATIVITY 095: BMS; Financial Interests, Institutional, Invited Speaker, BGB-A317-A1217-301/AdvanTIG-301: Beigene; Financial Interests, Institutional, Invited Speaker, Clinical Trial Chair ZEAL-1: GSK; Financial Interests, Institutional, Invited Speaker, Clinical Trial steering Committee PEARLS, MK-7684A: MSD; Financial Interests, Institutional, Invited Speaker, Clinical Trial Steering Committee SAPPHIRE: Mirati; Financial Interests, Institutional, Invited Speaker, LAGOON: Pharma Mar; Financial Interests, Institutional, Invited Speaker, phase 1/2 trials: Phosplatin Therapeutics; Financial Interests, Institutional, Invited Speaker, Clinical Trial Chair Skyscraper-01; chair ALEX; steering committee BFAST; steering committee BEAT-Meso; steering committee ImPower-030, IMforte: Roche/Genentech; Financial Interests, Institutional, Invited Speaker, Phase 2 Inupadenant with chemo: iTeos; Non-Financial Interests, Personal, Officer, ESMO President 2020-2022: ESMO; Non-Financial Interests, Personal, Officer, Council Member & Scientific Committee Chair: ETOP/IBCSG Partners; Non-Financial Interests, Personal, Officer, Vice-President Lung Group: SAKK; Non-Financial Interests, Personal, Other, Involved in Swiss politics: Swiss Political Activities; Non-Financial Interests, Personal, Officer, President and Council Member: Ballet Béjart Lausanne Foundation; Non-Financial Interests, Personal, Principal Investigator, Involved in academic trials: ETOP / EORTC / SAKK; Non-Financial Interests, Personal, Member: Association of Swiss Physicians FMH (CH), ASCO, AACR, IASLC; Non-Financial Interests, Personal, Leadership Role, ESMO President: ESMO; Non-Financial Interests, Personal, Member, Vice-President Lung Group: SAKK; Non-Financial Interests, Personal, Leadership Role, Vice -President: SAMO; Non-Financial Interests, Personal, Member, Association of Swiss interns and residents: ASMAC/VSAO. R.J. Salomonsen: Financial Interests, Personal, Full or part-time Employment: AstraZeneca. R. Tattersfield: Financial Interests, Personal, Full or part-time Employment: AstraZeneca. A. Wang: Financial Interests, Personal, Full or part-time Employment, Contractor (PHASTAR): AstraZeneca. Y. Xiao: Financial Interests, Personal, Full or part-time Employment: AstraZeneca. L. Cai: Financial Interests, Personal, Full or part-time Employment: AstraZeneca. S. Sadow: Financial Interests, Personal, Full or part-time Employment: AstraZeneca. R. Jassim: Financial Interests, Personal, Full or part-time Employment: AstraZeneca. S.V. Liu: Financial Interests, Personal, Advisory Board: Amgen, AstraZeneca, Bayer, Beigene, Blueprint, Boehringer Ingelheim, Bristol-Myers Squibb, Catalyst, Daiichi Sankyo, Eisai, Elevation Oncology, Genentech/Roche, Gilead, Guardant Health, Janssen, Jazz Pharmaceuticals, Lilly, Merck/MSD, Novartis, Regeneron, Sanofi, Takeda, Turning Point Therapeutics; Financial Interests, Personal, Principal Investigator: Alkermes, Blueprint, Bristol-Myers Squibb, Elevation Oncology, Genentech, Gilead, Merck, Merus, Nuvalent, Pfizer, RAPT, Turning Point Therapeutics; Financial Interests, Personal, Invited Speaker: Merck, Genentech, AstraZeneca; Financial Interests, Personal, Other, Travel, accommodations, expenses: Merck, Genentech, AstraZeneca.

Collapse
Poster Display Poster Display session

108P - Role of the prognostic nutritional index in predicting survival in advanced hepatocellular carcinoma treated with atezolizumab plus bevacizumab. (ID 200)

Presentation Number
108P
Lecture Time
12:30 - 12:30
Speakers
  • MARA PERSANO (Cagliari, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The prognostic nutritional index (PNI) is a multiparametric score introduced by Onodera based on the blood levels of lymphocytes and albumin in patients with gastrointestinal neoplasms. Regarding hepatocellular carcinoma (HCC), its prognostic role has been demonstrated in patients treated with sorafenib and lenvatinib. The aim of this real-world study is to investigate the association between clinical outcomes and PNI in patients being treated with atezolizumab plus bevacizumab.

Methods

The overall cohort of this multicentric study included 871 consecutive HCC patients treated with atezolizumab plus bevacizumab in first-line therapy. The PNI was calculated as follows: 10 × serum albumin concentration (g/dL) + 0.005 × peripheral lymphocyte count (number/mm3).

Results

For only 773 patients, data regarding lymphocyte counts and albumin levels were available, so only these patients were included in the final analysis. The cut-off point of the PNI was determined to be 41 by ROC) analysis. 268 patients (34.7%) were categorized as the PNI-low group, while the remaining 505 (65.3%) patients as the PNI-high group. At the univariate analysis, high PNI was associated with longer overall survival (OS) (22.5 vs. 10.1 months, HR 0.34, p < 0.01) and progression-free survival (PFS) (8.7 vs. 5.8 months, HR 0.63, p < 0.01) compared to patients with low PNI. At the multivariate analysis, high versus low PNI resulted as an independent prognostic factor for OS (HR 0.49, p < 0.01) and PFS (HR 0.82, p = 0.01). There was no difference in objective response rate (ORR) between the two groups (high 26.1% vs. low 19.8%, p = 0.09), while disease control rate (DCR) was significantly higher in the PNI-high group (76.8% vs. 66.4%, p = 0.01).

Conclusions

PNI is an independent prognostic factor for OS and PFS in HCC patients on first-line treatment with atezolizumab plus bevacizumab.

Legal entity responsible for the study

M. Persano.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

109P - Impact of anti-drug antibody (ADA) on naxitamab efficacy and safety (ID 201)

Presentation Number
109P
Lecture Time
12:30 - 12:30
Speakers
  • Brian Kushner (New York, NY, United States of America)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Monoclonal antibody (mAb) immunogenicity manifests as ADA generation that may impact mAb efficacy and safety. Naxitamab (NAX) is a humanized GD2-binding mAb. We explored ADA formation to NAX in the ongoing phase II clinical trial 201 (NCT03363373).

Methods

NAX was administered with granulocyte-macrophage colony-stimulating factor (GM-CSF) on 4-week cycles. ADA positivity and neutralizing potential were assessed at baseline and post-baseline (Cycle 1 Day 12; pre NAX infusion for subsequent cycles) by validated assay following a multitiered approach, and effect on efficacy and safety was evaluated.

Results

At data cutoff (Dec 31, 2021) of planned interim analysis 74 patients (pts, safety population) contributed ADA data; 29 ps (39%) had positive ADA titers at any time during trial, 33% had neutralizing ADA post-baseline. In efficacy population (N=52) 17 pts had neutralizing ADA: 4 pts achieved complete response (CR), 3 pts had minor response, 6 had neutralizing ADA at time of response, 1 had non-neutralizing ADA, later became neutralizing with ongoing response. In all CR pts (n=4) the neutralizing ADA became undetectable after continued NAX treatment. The highest titers were found in patients with CR. Progressive disease was confirmed in 5 pts at first response assessment (study days 38-45) coinciding with first detection of neutralizing ADA (study day 29-57). All subjects with neutralizing ADA reported CTCAE Grade ≥2 pain AEs and pain as per Wong Baker/FLACC scales, indicating that NAX may bind to GD2 in the presence of neutralizing ADA. The NAX safety profile was similar between pts with or without neutralizing ADA. Notably, 4 SAEs of anaphylactic reaction occurred in 3 ADA-negative pts.

Efficacy by ADA status

Response ≥ one positive neutralizing ADA titer (n=17*) No ADA positive titers (n=34)
ORR, % (number of responders) (95% CI) 24 % (4) (6.8 – 49.9) 65% (22) (46.5 – 80.3)
CR, % (number of responders) (95% CI) 24% (4) (6.8 – 49.9) 47% (16) (29.8 -64.9)

* One patient in the efficacy population had non-neutralizing ADA

Conclusions

The presence of ADA during NAX+GM-CSF treatment did not impact the NAX safety profile. Presence of neutralizing ADA in CR patients warrants further exploration to better understand the impact of ADA on efficacy.

Clinical trial identification

NCT03363373.

Legal entity responsible for the study

Y-mabs Therapeutics.

Funding

Y-mabs Therapeutics.

Disclosure

D.A. Morgenstern: Financial Interests, Personal, Advisory Board: Boehringer Ingelheim, Clarity Pharmaceuticals, EUSA Pharma, Roche, Y-mAbs Therapeutics, Oncoheros Biosciences. K. Nysom: Financial Interests, Personal, Advisory Board: Y-mAbs, EUSA Pharma; Financial Interests, Personal, Invited Speaker: Y-mAbs, Bayer; Financial Interests, Personal, Advisory Role: Bayer; Financial Interests, Personal, Other, Data Monitoring Committee: Lilly. J. Faber: Financial Interests, Institutional, Invited Speaker: Y-mAbs Therapeutics Inc.; Non-Financial Interests, Personal, Principal Investigator: Y-mAbs Therapeutics Inc.; Non-Financial Interests, Institutional, Other, Participation in compassionate use program: Y-mAbs Therapeutics Inc. M. Bear: Financial Interests, Personal, Advisory Role: Y-mAbs. K. Tornøe: Financial Interests, Personal, Full or part-time Employment: Y-mAbs; Financial Interests, Personal, Stocks/Shares: Y-mAbs. P.S. Sørensen: Financial Interests, Personal, Full or part-time Employment: Y-mAbs; Financial Interests, Personal, Stocks/Shares: Y-mAbs. J. Mora: Financial Interests, Personal, Advisory Role: Y-mAbs. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

110P - Fecal Microbiota Transplantation in Combination with Fruquintinib and Tislelizumab in Refractory Microsatellite Stable Metastatic Colorectal Cancer: A single center phase II trial (ID 202)

Presentation Number
110P
Lecture Time
12:30 - 12:30
Speakers
  • Wensi Zhao (Wuhan, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Microsatellite stable (MSS) metastatic colorectal cancer (mCRC) is one of the tumor type in which PD-1 inhibitor as monotherapy has proven less effective. Studies have shown that anti-VEGF therapies may enhance anti-PD-1 efficacy and microbiome-based therapies can overcome resistance to anti-PD-1. This study was to explore the efficacy and safety of fecal microbiota transplantation (FMT) combined with anti-VEGF and anti-PD-1 therapy in advanced mCRC with MSS.

Methods

This open-label, phase II trial (ChiCTR2100046768) was conducted at Renmin Hospital of Wuhan University. Patients with MSS mCRC progressed after at least 2-lines prior systemic treatments were administered custom-made fecal microbiota capsule (10#, 3 times per day, day1-3) plus fruquintinib (5-3mg; once per day, 2 weeks on/1 week off) and tislelizumab (200mg, day 4) every 3 weeks. Stool and peripheral blood were collected to detect the dynamic changes of intestinal microbiota diversity and immune metabolism indexes. The primary endpoint was progression-free survival (PFS). Secondary endpoints were overall survival (OS), objective response rate (ORR), disease control rate (DCR), and safety.

Results

From May 10, 2021 to January 17, 2022, 20 patients were assigned to treatment. As of data cutoff on September 25, 2022, with a median follow-up time of 10.7 months (IQR 7.9-13.7), median PFS was 9.6 months (95% CI 6.8-12.5) and median OS was 13.5 months (95% CI 9.0-NR). The median treatment duration was 7 cycles. A reduction in the size of target lesions was achieved by 60% (12/20). ORR was 20% (4/20; 95%CI, 6.6-44.3). DCR was 95% (19/20; 95%CI, 73.1-99.7). Quality of life (QoL) improved in 55% of patients. The most common treatment-related adverse events of grade 3 or worse were stool occult blood positive (15%), hand-foot skin reaction (10%), proteinuria (10%), increased ALT (5%), hypertension (10%), hypothyroidism (5%). No treatment-related deaths occurred.

Conclusions

Ttislelizumab combined with fruquintinib and FMT significantly improved PFS and OS in advanced MSS mCRC compared with historical controls, with a manageable safety profile and good QoL. It deserves to be validated in a larger trial.

Clinical trial identification

ChiCTR2100046768, 2021-5-28.

Legal entity responsible for the study

The authors.

Funding

National Natural Science Foundation of China (grant no.82102954) Central Leading Local Science and Technology Development Special Foundation (grant no. ZYYD2020000169).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

111P - Identification of Atezolizumab plus BEvacizumab prognostic index via recursive partitioning analysis in advanced hepatocellular carcinoma: the ABE index. (ID 203)

Presentation Number
111P
Lecture Time
12:30 - 12:30
Speakers
  • MARA PERSANO (Cagliari, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

This study aimed to identify a new prognostic index by applying recursive partitioning analysis (RPA) in hepatocellular carcinoma (HCC) patients treated with atezolizumab plus bevacizumab (AB).

Methods

RPA was applied on 784 consecutive HCC patients treated with AB.

Results

RPA allowed the identification of the atezolizumab bevacizumab prognostic (ABE) index, comprising three groups of patients: low risk, [(i) Child-Pugh A (CPA) patients without macrovascular invasion (MVI) but with albumin-bilirubin (ALBI) 1, aspartate aminotransferase (AST) normal value (NV), and alpha-fetoprotein (AFP) < 400 ng/mL, (ii) CPA patients without MVI but with ALBI 1, AST increased value (IV), and neutrophil-lymphocyte ratio (NLR) < 3, and (iii) CPA patients with MVI, ALBI 1, and AFP < 400 ng/mL]; intermediate risk, [(i) CPA patients without MVI but with ALBI 1, AST NV, and AFP ≥ 400 ng/mL, and (ii) CPA patients without MVI but with ALBI 1, AST IV, and NLR ≥ 3]; high risk [(i) CPA patients with ALBI 2, (ii) CPA patients with ALBI 1, MVI, and AFP ≥ 400 ng/mL, and (iii) CPB patients]. Overall survival was 22.5 months (95% CI 17.0-22.5 months) in patients with low risk (60.1%), 14.2 months (95% CI 12.4-15.7 months) in intermediate risk (19.1%), and 7.0 months (95% CI 6.0-8.7 months) in high risk (20.8%); low risk HR 1, intermediate risk HR 1.76 (95% CI 1.26-2.46), high risk HR 3.99 (95% CI 2.76-5.77); P < 0.01. Progression-free survival was 9.4 months (95% CI 8.4-10.8 months) in patients with low risk, 6.1 months (95% CI 5.5-8.1 months) in intermediate risk, and 5.3 months (95% CI 3.7-5.8 months) in high risk; low risk HR 1 (reference group), intermediate risk HR 1.47 (95% CI 1.14-1.89), high risk HR 1.79 (95% CI 1.37-2.35); P < 0.01. In the three groups, differing profiles of toxicity have been highlighted, notably in terms of hypertension (low risk 27.4%; intermediate risk 22.7%; high risk 17.2%, P = 0.03), proteinuria (low risk 28.7%; intermediate risk 35.3%; high risk 22.7%, P < 0.05), and hypothyroidism (low risk 6.1%; intermediate risk 2.7%; high risk 1.8%; P = 0.03).

Conclusions

The ABE index is an easy-to-use tool able to stratify HCC patients undergoing first-line therapy with AB.

Legal entity responsible for the study

M. Persano.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

112P - Pseudoprogression predicts better prognosis in advanced non small cell lung cancer treated with immune-check point inhibitors (ID 204)

Presentation Number
112P
Lecture Time
12:30 - 12:30
Speakers
  • Nihan Senturk Oztas (Istanbul, Turkey)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Pseudoprogression (PSP) is an initial increase in the size of the primary tumor or the appearance of the new lesions followed by a distinct decrease in tumor burden. Herein, we aimed to evaluate the effect of PSP on treatment response in patients with advanced non-small cell lung cancer (NSCLC).

Methods

We retrospectively analyzed patients with advanced NSCLC treated with immune checkpoint inhibitors in Cerrahpasa Medical Faculty clinical trial unit between 2014-2021. We defined PSP as a perceptible increase in tumor volume followed by subsequent clinical benefit beyond progression. All treatment responses were evaluated according to RECIST 1.1 criteria.

Results

Demographics of the entire study population are depicted in the table. Sixteen (17%) of the 90 patients were evaluated to have PSP. The median overall survival (OS) was 25 months in patients without PSP and 48,9 months in patients with PSP (p=0.008). Median progression-free survival (PFS) for PSP and non-PSP group was 13,5 months and 36,9 months, respectively (p=0.0036). The duration of response was 11,8 months in the non-PSP group and 36,6 months in the PSP group (p=0.0016). The results showed a statistically significant improvement in PFS and OS in favor of the PSP group.

Demographics of the entire study population

Variable PSP Group Non-PSP Group
Number of patients 16 74
Age, mean, years 61 61,5
Female, n (%) 61 (82,4) 15 (93.7)
Mono ICI,n(%) 14 (87,5) 63 (85,1)
Dual ICI,n(%) 0 4 (5,4)
Chemotherapy + ICI, n (%) 2 (12,5) 7 (9,5)

Conclusions

Our study shows that PSP is not rare and related to good clinical outcomes. Recognizing pseudoprogression in clinical practice is critical to prevent premature discontinuation of ICI.

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 Poster Display session

113P - The prognostic and predictive roles of serum C-reactive protein and PD-L1 in non-small cell lung cancer (ID 205)

Presentation Number
113P
Lecture Time
12:30 - 12:30
Speakers
  • Saara V. Kuusisalo (Oulu, Finland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

PD-(L)1 agents have revolutionized the treatment paradigms of non-small cell lung cancer (NSCLC). In addition to PD-L1 score, predictive biomarkers for PD-(L)1 therapies are limited. Us and others have previously shown that systemic inflammation, indicated by elevated C-reactive protein (CRP) level, is associated with a poor prognosis in PD-(L)1 treated.

Methods

We collected all NSCLC patients (n=314) who had undergone tumor PD-L1 tumor proportion score (TPS) analysis at Oulu University Hospital between 2015-21. CRP levels, treatment history, immune checkpoint inhibitor (ICI) therapy details, and survival were collected from electronic patient records. The patients were divided into two categories based on plasma CRP levels (≤10 vs >10) and PD-L1 TPS (<50 vs ≥50).

Results

In the whole cohort (n=314), CRP level of ≤10mg/l was associated with improved overall survival in univariate (HR 0.30, Cl 95% 0.22-0.41) and multivariate analyzes (HR 0.42, CI 95% 0.28-0.66). Among the ICI treated (n=55), both CRP of ≤10 and PD-L1 TPS of ≥50 were associated with improved progression-free survival (PFS) in univariate (HR 0.45, CI 95% 0.23-0.89; HR 0.49, CI 95% 0.25-0.94) and multivariate (HR 0.44, CI 95% 0.22-0.86; HR 0.46, CI 95% 0.24-0.92) analyzes. The combination of high PD-L1 TPS (≥50) and CRP (>10) carried a high negative predictive value among the ICI treated with a median PFS of 3.22 months (CI 95% 0.67-5.77) which was very similar to patients with low PD-L1 (3.35 months, CI 95% 1.33-5.37). Univariate and multivariate analysis for PFS (IO) and OS (IO).

Univariate Multivariate
HR CI (95%) HR CI (95%)
PFS (IO)
CRP
≤10 vs. >10 0.449 0.227-0.886 0.435 0.220-0.862
PD-L1
≥50 vs. <50 0.485 0.250-0.942 0.464 0.235-0.917
OS (IO)
CRP
≤10 vs. >10 0.479 0.225-1.021 0.536 0.248-1.159
PD-L1
≥50 vs. <50 0.385 0.180-0.825 0.387 0.175-0.875

Conclusions

Adding plasma CRP levels to PD-L1 TPS significantly increased the predictive value of sole PD-L1 and patients with high CRP bared little benefit from PD-(L)1 therapies regardless of PD-L1 score. The study highlights combined evaluation of plasma CRP and PD-L1 TPS as negative predictive marker for ICI therapies.

Legal entity responsible for the study

The authors.

Funding

Finnish Cancer Institute.

Disclosure

S.M.E. Iivanainen: Financial Interests, Personal, Invited Speaker: Roche, MSD, Boehringer Ingelheim, Takeda, BMS, Novartis, Pierre-Fabre, AstraZeneca; Financial Interests, Institutional, Research Grant: Roche, AstraZeneca; Financial Interests, Personal, Research Grant: Boehringer Ingelheim. J.P. Koivunen: Financial Interests, Personal, Advisory Board: Merck Sharp Dome, BMS, AstraZeneca, Pfizer, Roche, Boehringer Ingelheim, Takeda, Amgen; Financial Interests, Personal, Full or part-time Employment: Faron Pharmaceuticals; Financial Interests, Personal, Stocks/Shares: Faron Pharmaceuticals; Financial Interests, Institutional, Funding: AstraZeneca; Financial Interests, Institutional, Invited Speaker: Boehringer Ingelheim, Roche. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

114P - Systemic Inflammatory Index as a prognostic biomarker in metastatic melanoma patients under immune checkpoint inhibitors (ID 206)

Presentation Number
114P
Lecture Time
12:30 - 12:30
Speakers
  • Mariana Rebordão Pires (Coimbra, Portugal)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of melanoma. Systemic Inflammatory Index (SII) has been evaluated as a prognostic biomarker in different solid neoplasms, such as melanoma.

The aim of this work was to evaluate the SII prognostic value in metastatic melanoma patients under first line ICIs.

Methods

Retrospective, single-center study of metastatic melanoma patients who received first- line ICIs between January/2017 and August/2021.

The SII [Platelets x neutrophil/lymphocyte ratio (NLR)] was calculated before ICIs start and was considered high if > 572 (Zhong JH et al. Prognostic role of systemic immune-inflammation index in solid tumors: a systematic review and meta-analysis. Oncotarget. 2017 Jun 29;8(43):75381-75388.)

Results

Fifty-one patients enrolled, 32 male, median age at diagnosis 67 years [30-84]. Twenty were stage IV at diagnosis. In patients not stage IV at diagnosis, the median time to metastasis was 11 months [0-279]. Median follow-up time was 7 months [0-54]. The median progression-free survival (PFS) and overall survival (OS) was, respectively, 7 months [1.7-12.2] and 19 months [4.9-33.0]. A total of 24 patients were included in the high SII group. The median PFS and OS were, respectively, 10 months [95%CI, 4.0-15.9, p=0.008] and 29 months [95% CI, 13.8-44.1, p=0.022] in the low SII group and 1 month [95% CI, 0.0-3.7, p=0.008] and 7 months [95% CI, 5.1-8.9, p=0.022] in the high IIS group. Multivariate analysis, showed as independent predictors of survival the NLR [p=0.001, HR 0.520, 95%CI 0.349-0.69] and thrombocytosis [p=0.034, HR 0.160, 95%CI 0.055- 0.265].

Conclusions

This paper shows the prognostic value of SII in metastatic malignant melanoma patients. It may become an important low-cost tool in the management and multidisciplinary approach of these 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 Poster Display session

115P - Reflectance Confocal Microscopy - An In Vivo Method Of Visualization Of NMSC Under Anti-PD-1 Therapy In Correlation To Histological Findings - Preliminary Report (ID 207)

Presentation Number
115P
Lecture Time
12:30 - 12:30
Speakers
  • Iwona Lugowska (Warsaw, Poland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Tumour microenvironment is crucial for skin cancer progression, metastasizing and response to immunotherapy. Reflectance confocal microscopy (RCM) is a non-invasive in vivo method enabling skin cancer diagnostics and observation of inflammatory infiltrates in near histological resolution. The aim of the study is to describe the changes of microscopic criteria (by RCM and optical histology) in NMSC, stroma, and inflammatory infiltrates during the balstilimab (AGEN2034) therapy to evaluate the practical prediction attitude and repeatability of RCM.

Methods

Thirteen patients with NMSC (BCC, SCC, Adnexal carcinoma) enrolled in an open-label phase-II study (AGENONMELA; ABM_01_00004_03). RCM was performed in 8/13 patients in 2 parts of neoplasm on screening and/or week12 visits (2 patients had 2 RCM and 6 patients had a single RCM examination). In 13 patients were performed biopsies of corresponding areas for HP and translational tests were on the same visits.

Results

The inflammatory infiltrates were visualized in 12/12 lesions during the SCR and in 6/6 lesions on W12 visit in peripheral (epidermis, DEJ), perivascular, and/or peri/intra-tumour locations. Its mixed, neutrophilic or lymphocytic composition was confirmed in both methods. In complete response, an excellent RCM-HP correlation was observed. The partial regression of cancer was observed with concomitant differences in TILs and stroma with correspondence to HP.

Conclusions

The penetration limits RCM utility up to a depth of 200-300um. Further studies on the larger group of patients are needed to confirm the RCM accuracy in the prediction of histological findings, demonstrating its possible role in the observation of NMSC changes under immunotherapy. RCM is valuable in vivo method to monitor the response of NMSCs to immunotherapy, which may have applications in clinical practice.

Clinical trial identification

AGENONMELA; ABM_01_00004_03.

Legal entity responsible for the study

Early Phase Research Center, Maria Skłodowska-Curie National Research Institute, National Institute of Oncology, Warsaw, Poland.

Funding

Medical Research Agency, Poland Agenus Inc.

Disclosure

M.A. Slowinska: Financial Interests, Personal, Advisory Board: Takeda, Novartis, BMS; Financial Interests, Personal, Invited Speaker: Takeda, Novartis, Roche, Medac, BMS. A. Szumera-Cieckiewicz: Financial Interests, Personal, Research Grant: Agenus. M. Chelstowska: Financial Interests, Personal, Research Grant: Agenus. A. Dawidowska: Financial Interests, Personal, Research Grant: Agenus. S. Jaczewska: Financial Interests, Personal, Research Grant: Agenus. P. Rutkowski: Financial Interests, Personal, Invited Speaker, honoraria for lectures: MSD, BMS, Pierre Fabre; Financial Interests, Personal, Advisory Board: MSD, BMS, Pierre Fabre, Merck, Sanofi, Blueprint Medicines, Philogen; Financial Interests, Personal, Invited Speaker: Merck, Sanofi, Novartis; Financial Interests, Institutional, Research Grant, research grant for ISS: Pfzer; Financial Interests, Institutional, Funding, research grant for institution: BMS; Non-Financial Interests, Personal, Invited Speaker: Polish Society of Surgical Oncology; Non-Financial Interests, Personal, Officer: ASCO; Non-Financial Interests, Personal, Invited Speaker, President Elect: Polish Oncological Society. I. Lugowska: Financial Interests, Personal, Invited Speaker, The reports of clinical trials: Roche, BMS, Macrogenics, Amgen; Financial Interests, Institutional, Other, Research grants: Roche; Financial Interests, Institutional, Other, Research grant: Agenus; Financial Interests, Personal and Institutional, Invited Speaker: Agenus, Roche, BMS, Janssen, Astra, Incyte, Macrogenics, Checkpoint Inhibitors, Celon, Pfizer, MSD, Debio; Non-Financial Interests, Personal, Project Lead: MSCI; Non-Financial Interests, Personal, Advisory Role, Board Member: EORTC. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

116P - Clinical Significance of Serum-derived Exosomal PD-L1 Expression in Patients with Advanced Pancreatic Cancer (ID 208)

Presentation Number
116P
Lecture Time
12:30 - 12:30
Speakers
  • Se Jun Park (Seoul, Korea, Republic of)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

PD-1 and PD-L1 interaction leads to immune evasion of various tumors and is associated with poor prognosis in patients with pancreatic cancer. Although immune check point inhibitor has yielded clinical benefits in several types of solid tumor, the efficacy was modest for pancreatic cancer. PD-L1 expressing exosomes can diminish immune responses against tumor, however, the roles of PD-L1 containing exosomes in pancreatic cancer are poorly understood.

Methods

Pre-treatment serum samples were collected from patients with advanced pancreatic cancer. Exosomes derived from the serum were isolated using Exoquick kit. Exosomal PD-L1 (exoPD-L1) was detected by enzyme-linked immuno-sorbent assay, and the concentration was further normalized to per milligram exosomal protein. PD-L1 expression in matched tissues were evaluated by PD-L1 immunohistochemistry (22C3) assay, described with combined positive score. Cutoff value of exoPD-L1 for survival was assessed with ROC curve analysis. Kaplan-Meier analysis was performed to obtain median overall survival.

Results

In total, 77 samples of patients with advanced pancreatic cancer were analyzed for exoPD-L1 level. The median values of exoPD-L1 in plasma was 0.16 pg/mg (IQR, 0.12-0.20), and exoPD-L1 was not detected in 7 (9.0%) patients. ExoPD-L1 was measured significantly higher in patients with systemic disease than in locally advanced disease (p=0.023). Significant higher proportion of elevated exoPD-L1 was observed in patients with positive tissue PD-L1 expression versus those who were negative tissue PD-L1 expression (p=0.001). Patients were classified as two groups with low and high levels of exoPD-L1 using cutoff determined with ROC curves (0.165 pg/mg, AUC area 0.617, p=0.078). At a median follow-up of 7.97 months, the median OS was 11.2 months (95% CI, 6.33-16.1) in the low ExoPD-L1 group, as compared with 7.01 months (95% CI, 3.27-9.45) in the high ExoPD-L1 group (HR=0.65; 95% CI, 0.38-1.14; p=0.116).

Conclusions

The serum-derived exoPD-L1 levels were higher in metastatic pancreatic cancer than locally advanced disease. Patients with high exoPD-L1 levels tended to have a worse survival outcome than patients with low exoPD-L1 levels, though statistically not significant.

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 Poster Display session

117P - Beta-Blockers in Lung Cancer Patients Receiving Immunotherapy (ID 209)

Presentation Number
117P
Lecture Time
12:30 - 12:30
Speakers
  • Ana Mendes (Amadora, Portugal)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The treatment standard of cancer has dramatically diverted with immune checkpoint inhibitors (ICI). Despite the proven clinical advantage, some tumors do not respond to ICI. The expression of PD-L1, the tumor microenvironment (TME), and the tumor mutational burden are essential to the success of ICI and are currently considered biomarkers predictive of response. Increased Beta-adrenergic receptor signaling has been shown to promote the creation of an immunosuppressive TME. The annulment of this pathway provides a more responsive TME and may enhance the activity of ICI, and the use of beta-blockade for this purpose has shown conflicting results. We investigated patients with lung cancer who concomitantly used beta-blockers (BB) and ICI, hypothesizing that blocking the beta-adrenergic pathway would impact the outcome.

Methods

We retrospectively reviewed 51 patients treated at our institution for 18 months with ICIs for non-small-cell lung cancer (NSCLC). Comparisons of overall survival and progression-free survival (PFS) were performed using Kaplan-Meier analysis with log-rank test, and a univariate regression analysis was performed with a Cox proportional hazards model.

Results

Among the 51 patients, 11 concomitantly used beta blockers. There was no significant increase in overall survival among patients who took beta-blockers (p=0.83; 95% confidence interval, 0.33-2.47), and BB was not predictive of PFS. Although well established, our study confirmed that elevated levels of lactate dehydrogenase were associated with poorer overall survival (p=0.034, 95% confidence interval 1.000-1.005), and weight loss was predictive of PFS (p=0.019, 95% confidence interval 1.097-2.839).

Conclusions

In summary, this study found no evidence that BBs enhance immunotherapy effectiveness. Despite that, this was a small study, and these results should be validated in prospective clinical studies.

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 Poster Display session

118P - Immunosenescence and response to immunotherapy in elderly patients: A possible prognostic tool (ID 210)

Presentation Number
118P
Lecture Time
12:30 - 12:30
Speakers
  • Andrea Pretta (Cagliari, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Aging leads to several changes concerning the immune system activity too. This process is known as “immunosenescence” and it can alter the immune response, especially T-cells response. However, there is lack of data about the response to immunotherapy regimen in elderly patients as well as a predictive factor of response. This retrospective study aims to evaluate the association between the lymphocytes/monocytes ratio (LMr) as a factor of better outcomes.

Methods

Data were collected in a single center, at University Hospital of Cagliari, from 2016 to 2022. Overall, 65 patients were enrolled. The median age was 77 (±5,6). 51 out of 65 (78%) were males, 14 were females (22%). As for pathology: 31 out 65 (47.7%) had lung cancer, 16/65 (24.6%) had melanoma, 11/65 (16,9%) had kidney cancer, and 7/65 (10,8%) had gastrointestinal carcinoma. All of patients underwent anti PD-1 therapy. ECOG PS were 0 and 1. Statistical analysis were performed through MedCalc package.

Results

ROC curves were performed evaluating LMr in patients alive at 6, 12, and 24 months. All of them were significant for the cut-off of > 2.5 (p < 0.0001). Subsequently, we split patients into two subgroups: one harboring an LMr ≤ 2.5 and the other one an LMr > 2.5 (32 out 65 patients and 33 out of 65 patients had a ratio ≤ and > 2.5, respectively). Patients with ratio > 2.5 showed a statistically significant higher median OS (36 months, 95% CI 31 – 47) than patients with a ratio ≤ 2.5 (16 months, 95% CI 10-44) (p = 0.03).

Conclusions

The results showed a correlation between baseline higher LMr to better outcomes in patients undergoing treatment with Immune Checkpoint Inhibitors. LMr could be a simple efficient tool to predict a higher T-cells activity and a better response to immunotherapy. Further studies are needed to validate it.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

E. Lai: Financial Interests, Personal, Advisory Board: AstraZeneca, MSD. M. Scartozzi: Financial Interests, Personal, Advisory Board: Amgen, Sanofi, MSD, EISAI, Merck, Bayer; Financial Interests, Personal, Speaker’s Bureau: Amgen, Sanofi, MSD, EISAI, Merck, Bayer. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

119TiP - An observational study to assess the effectiveness and safety of cemiplimab in patients with advanced non small cell lung cancer (NSCLC) in routine clinical practice within Europe (CEMI-LUNG) (ID 211)

Presentation Number
119TiP
Lecture Time
12:30 - 12:30
Speakers
  • Christian Gessner (Leipzig, Germany)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Blockade of the PD-1 pathway is the mainstay for the first-line (1L) treatment of patients (pts) with advanced NSCLC without targetable oncogenic alterations. Cemiplimab is a PD-1 inhibitor and has improved overall survival (OS) and progression-free survival (PFS) vs chemotherapy in 1L advanced NSCLC patients with high PD-L1 expression (PD-L1 ≥50%) who are without driver mutations based on the EMPOWER-Lung 1 study. Cemiplimab was subsequently licensed as monotherapy in this front line pt population by the FDA (Feb 2021) and EMA (May 2021) based on results from EMPOWER-Lung 1. Currently, there are no prospective data on the effectiveness and safety of cemiplimab in advanced NSCLC within routine clinical practice setting. Such data could provide additional evidence to guide treatment and optimal use as per the licensed indication, including in patients that could have been excluded or not typically included in clinical trials. Therefore, CEMI-LUNG (NCT05363319) is a pragmatic, prospective, noninterventional, observational cohort study that aims to address these data gaps.

Trial Design

Pts with advanced NSCLC who are initiating a licensed cemiplimab-based regimen as part of their routine clinical practice are eligible. Study visits will follow the standard-of-care schedule and data will be collected every 3 months while on treatment and every 6 months for 36 months following treatment discontinuation (maximum study duration, 72 months) until death, loss to follow-up, study withdrawal or end of study period, whichever occurs first. The study plans to enrol ∼300 pts across 30 European sites. Decisions regarding treatment will be made by the treating physician in accordance with local clinical practice. The primary objective is to assess overall survival. Secondary objectives are to describe objective response rate, time to response, time to first subsequent anti-NSCLC treatment, duration of response, PFS, and the incidence and severity of adverse events. Three interim analyses are planned in addition to the final analysis. This study is open for enrolment.

Clinical trial identification

NCT05363319.

Editorial acknowledgement

Editorial writing support was provided by Sameen Yousaf, PhD, of Prime, Knutsford, UK, funded by Regeneron Pharmaceuticals, Inc., and Sanofi.

Legal entity responsible for the study

Sanofi and Regeneron Pharmaceuticals, Inc.

Funding

Sanofi and Regeneron Pharmaceuticals, Inc.

Disclosure

C. Gessner: Financial Interests, Personal, Advisory Board: GlaxoSmithKline, Pfizer, AstraZeneca, Roche, Novartis, Bristol-Myers Squibb, Merck Sharp & Dohme, Berlin-Chemie, Chiesi, Boehringer Ingelheim and Sanofi. F. Griesinger: Financial Interests, Personal, Other, Scientific support: Astra, Boehringer, BMS, Celgene, Lilly, MSD, Novartis, Pfizer, Roche, Takeda, Siemens, Sanofi; Financial Interests, Personal, Speaker’s Bureau: Astra, Boehringer, BMS, Celgene, Lilly, MSD, Novartis, Pfizer, Roche, Takeda, Ariad, AbbVie, Siemens, Tesaro/GSK, Amgen, Sanofi; Financial Interests, Personal, Advisory Board: Astra, Boehringer, BMS, Celgene, Lilly, MSD, Novartis, Pfizer, Roche, Takeda, Ariad, AbbVie, Tesaro/GSK, Siemens, Tesaro, Amgen, Sanofi. R. Shah: Financial Interests, Personal, Advisory Board: Boehringer Ingelheim, AstraZeneca, Roche, BMS, MSD, Pfizer, Lilly, Novartis, Takeda, Bayer, Beigene, Guardant, Sanofi. T. Talbot: Financial Interests, Personal, Advisory Board: AbbVie, Amgen Boerhinger Ingelheim, Bristol-Myers Squibb, Lilly, GlaxoSmithKline, Merck Sharp & Dohme, Novartis, Pfizer, Roche and Sanofi-Genzyme. S. Venkateshan: Financial Interests, Personal, Full or part-time Employment: Regeneron Pharmaceuticals, Inc.; Financial Interests, Personal, Stocks/Shares: Regeneron Pharmaceuticals, Inc. A.K. Vadanahalli Shankar: Financial Interests, Personal, Stocks/Shares: Sanofi; Financial Interests, Personal, Full or part-time Employment: Sanofi. A. Seluzhytsky: Financial Interests, Personal, Stocks/Shares: Sanofi; Financial Interests, Personal, Full or part-time Employment: Sanofi. D.M. Marsden: Financial Interests, Personal, Stocks/Shares: Sanofi; Financial Interests, Personal, Full or part-time Employment: Sanofi.

Collapse
Poster Display Poster Display session

120TiP - Conversion of Response to Immune Checkpoint Inhibition by Fecal Microbiota Transplantation in Patients With Metastatic Melanoma: a Randomized Phase Ib/IIa Trial (ID 212)

Presentation Number
120TiP
Lecture Time
12:30 - 12:30
Speakers
  • Jessica S. Borgers (Amsterdam, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The gut microbiome plays an important role in immune modulation. Specifically, the presence and function of certain gut microbial taxa has been associated with better anti-tumor immune responses. In clinical trials using fecal microbiota transplantation (FMT) to treat immune checkpoint inhibitor (ICI) refractory metastatic melanoma patients, complete responses (CR), partial responses (PR) and durable stable disease (SD) have been observed, with prolonged overall survival. However, the underlying mechanism determining which patients will or will not respond and what the optimal FMT composition is, is not fully elucidated. Furthermore, it is unknown whether a change in microbiome irrespective of the origin of the FMT (from ICI-responding (R) or non-responding (NR) metastatic melanoma donors) is able to revert ICI unresponsiveness. To address this, we will transfer the microbiota of either ICI-R or ICI-NR melanoma patients via FMT.

Trial Design

In this randomized, double-blinded phase Ib/IIa trial 24 αPD1-refractory patients with advanced stage cutaneous melanoma, will receive an FMT of ICI-R or ICI-NR donors. Anti-PD-1 treatment will be continued according to the patient’s regular treatment schedule. Donors will be selected from patients with metastatic melanoma treated with anti-PD-1 therapy. Two patients with a good response (≥30% decrease within the past 24 months) and two patients with progression (≥20% increase within the past 3 months) will be selected as ICI responding or non-responding donor, respectively. The primary endpoint is to investigate the efficacy, defined as clinical benefit (SD, PR, CR) at 12 weeks, confirmed on a second scan at week 16. The secondary endpoint is safety. The study will be considered safe if less than seven patients have experienced a grade 3-4 toxicity. To assess changes in the gut microbiome and metabolome, feces samples will be collected at baseline, prior to FMT and after FMT. Tumor biopsies and blood samples will be collected to analyze local and systemic immune changes. Patient recruitment started in April 2022.

Clinical trial identification

NCT05251389.

Legal entity responsible for the study

Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital.

Funding

AVL Donation Investment Fund.

Disclosure

E.M. Terveer: Financial Interests, Personal, Advisory Board: Finch Therapeutics . E.J. Kuijper: Financial Interests, Personal, Research Grant: Vedanta Biosciences. 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, Third Rock Venture, Iovance Biotherapeutics; 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, Stocks/Shares: Neogene Therapeutics; Financial Interests, Institutional, Research Grant: Bristol Myers Squibb, BioNTech US, Merck Sharpe & Dohme, Amgen, Novartis, Asher Bio; 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. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

121TiP - Prospective, single arm, phase II clinical trial of pembrolizumab combined with neoadjuvant chemoradiotherapy and surgery for locally advanced upper esophageal squamous cell carcinoma (ID 213)

Presentation Number
121TiP
Lecture Time
12:30 - 12:30
Speakers
  • Yong Li (Bejing, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Definitive chemoradiotherapy (dCRT) is recommended as the standard treatment for locally advanced cervical esophageal cancer (EC). However, the recurrence rate of cervical or upper thoracic EC patients (pts) who only received high-dose dCRT was still as high as about 60% within 3 years, and the 5-year survival rate was about 30%. Therefore, these pts need more effective treatment to prevent recurrence and prolong survival.

Trial Design

The study is a prospective, single center, single arm phase II trial, aims to explore the safety and feasibility of pembrolizumab (Pem) combined with neoadjuvant chemoradiotherapy and surgery for locally advanced upper esophageal squamous cell carcinoma (ESCC). Pts are eligible with cT1-3N1-2M0 or cT2-3N0M0 (according to UICC/AJCC 8th and JES) high (the distance from the upper edge of the tumor to the esophageal entrance ≤ 5cm) ESCC. 5 subjects are first enrolled as safety run-in part. After comprehensive risk assessment, continued to include 40 subjects. The subjects first received the 2 cycles of induction treatment (Pem 200mg, IV, D1, q3w +albumin paclitaxel, 125mg/m2, IV, D1/D8, q3w +cisplatin, 70mg/m2 in total, IV, given in three times, q3w) for 6 weeks, then received the sequential treatment (Pem 200mg, IV, D1, q3w, 2 cycles +albumin paclitaxel, 100mg, IV, D1, QW, 4 cycles+ cisplatin, 20mg/m2, IV, D1, QW, 4 cycles + PTV, 44gy/2gy/22fx, 5 days a week, 5 weeks in total) for 6 weeks, and then received surgery with 4-8 weeks. For pts with non-pCR after surgery, continue to receive maintenance therapy of Pem after surgery (until 1 year or disease progression or intolerable toxicity); Follow up for pts with pCR. Safety follow-up will be conducted after the first medication or within 30 days after surgery (whichever occurs first). The primary endpoint is major pathological response, which is defined as ≤ 10% of residual viable tumor at the time of resection.

Clinical trial identification

NCT05541445.

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 Poster Display session

201P - Phase II study of taminadenant (A2AR antagonist) + spartalizumab (anti PD-1 antibody) in patients with triple negative breast cancer (TNBC) (ID 291)

Presentation Number
201P
Lecture Time
12:30 - 12:30
Speakers
  • Guy Jerusalem (Liège, Belgium)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Anti–PD-(L)1 agents have limited activity in relapsed/refractory (r/r) TNBC. Blockade of immune suppression by antagonism of the adenosine pathway may increase the antitumor activity of PD-1 inhibitors in r/r TNBC. Here, we present data on the A2AR antagonist TAM + the anti–PD-1 antibody spartalizumab in TNBC.

Methods

In part 1 of this phase II study (NCT03207867), pts with relapsed TNBC were given 160 mg oral TAM twice daily, continuously, plus 400 mg intravenous spartalizumab every 4 weeks. The primary objective was to evaluate the overall response rate (ORR) per RECIST v1.1. Secondary objectives were to assess the safety and tolerability of the combination treatment, and to characterize changes in the immune infiltrates in tumors upon treatment. Tumor biopsies were taken prior to and on treatment for biomarker analyses.

Results

As of Dec 3, 2021, 30 pts with PD-(L)1–naive TNBC were enrolled in part 1. The majority of pts had received ≥2 lines of therapy for their disease and had an ECOG status ≤1 at study entry. Based on data from 27 pts with ≥1 post-baseline scan, the ORR was 11%, the disease control rate was 37%, and median progression-free survival was 1.8 months. In all 30 pts, common, all-grade, treatment-related adverse events (TRAEs) were observed in 23 pts (77%); Grade ≥3 TRAEs were reported in seven pts (including fatigue [n=2]; increased aspartate aminotransferase [n=2]; increased alanine aminotransferase [n=2]; dyspnea [n=1]; interstitial lung disease [n=1]; and duodenal ulcer [n=1]). There was evidence of a robust induction of CD8 infiltration in the tumors upon treatment (n=13), unlike the changes observed during a phase I trial of spartalizumab in a similar TNBC population. In addition, a trend was seen between CD8 percent marker area change and TAM exposure. Pharmacokinetics data will be presented.

Conclusions

TAM + spartalizumab showed clinical benefit in a fraction of pts with relapsed TNBC. Biomarker data support the role of TAM in shifting the immune microenvironment toward a “T-cell–inflamed” tumor, and possibly in increasing the efficacy of anti–PD-1 agents.

Clinical trial identification

NCT03207867.

Editorial acknowledgement

Editorial assistance was provided by Manoshi Nath, MSc of Articulate Science Ltd, and was funded by Novartis Pharmaceuticals Corporation.

Legal entity responsible for the study

Novartis Pharmaceuticals Corporation.

Funding

Novartis Pharmaceuticals Corporation.

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, Novartis, AZ/Daiichi Sankyo, BMS; Financial Interests, Institutional, Invited Speaker: Novartis, Roche, Syneos Health, Iqvia, TRIO, Bayer, MSD, IBCSG, PRA, Eli Lilly, PPD, Theradex, ABCSG, Modra, AstraZeneca, Odonate Therapeutics, BMS, ICON, Boehringer Ingelheim, 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. F.G.M. De Braud: Financial Interests, Personal, Invited Speaker: BMS, Healthcare Research & Pharmacoepidemiology, Merck Group, MSD, Pfizer, Servier, Sanofi, Roche, Amgen, Incyte, Dephaforum, Seagen, Novartis, F.Hoffmann-LaRoche Ltd., BMS, Ignyta Operating Inc., Merck Sharp & Dohme Spa, Kymab, Pfizer, Tesaro, MSD, MedImmune LCC, Exelixis Inc., Loxo Oncology Incorporated, Daiichi Sankyo Dev. Limited, Basilea Pharmaceutica International AG, Janssen-Cilag International NV, Merck KGAA; Financial Interests, Personal, Other, Consultant Advisory Board: Roche, EMD Serono, NMS Nerviano Medical Science, Sanofi, MSD, Novartis, Incyte, BMS, Menarini, AstraZeneca, Pierre Fabre. P. Grell: Financial Interests, Personal, Invited Speaker: Servier; Financial Interests, Personal, Advisory Board: Roche. A. Gonçalves: Financial Interests, Institutional, Invited Speaker: Novartis, Roche, MSD, AstraZeneca, Daiichy Sankyo. T.J.Y. Tan: Financial Interests, Personal, Advisory Board: AstraZeneca, MSD, Everest Medicines (Singapore) Pte Ltd., DKSH, Pfizer; Financial Interests, Personal, Invited Speaker: DKSH, AstraZeneca, Novartis, Roche, Pfizer, MSD, DHPL Malaysia SDN BHD; Financial Interests, Institutional, Research Grant: AstraZeneca; Financial Interests, Institutional, Invited Speaker: Roche, Novartis, AstraZeneca, Odonate, Daiichi Sankyo, Genentech, Sanofi; Non-Financial Interests, Personal, Member: ASCO. R. Greil: Financial Interests, Personal, Advisory Role: Celgene, Novartis, Amgen, Bristol Myers Squibb, Takeda, AbbVie, AstraZeneca, Janssen, Gilead, Merck, Daiichi Sankyo, Sanofi, Sandoz; Financial Interests, Personal, Sponsor/Funding: Celgene, Roche, Merck, Takeda, AstraZeneca, Novartis, Amgen, Bristol Myers Squibb, Merck, Sandoz, AbbVie, Gilead, Daiichi Sankyo; Financial Interests, Personal, Other, Travel, Accomodations, Expenses: Roche, Amgen, Janssen, AstraZeneca, Novartis, Merck, Celgene, Gilead, Bristol Myers Squibb, AbbVie, Daiichi Sankyo. T.A. Yap: Financial Interests, Personal, Other, Consultant: Almac, Aduro, AstraZeneca, Atrin, Axiom, Bayer, Bristol Myers Squibb, Clovis, Cybrexa, EMD Serono, Guidepoint, Ignyta, I-Mab, Jansen, Merck, Pfizer, Repare, Roche, Schrodinger, Varian, Zai Labs, AbbVie, Acrivon, Adagene, Amphista, Artios, Athena, Avoro, Baptist Health Systems, Beigene, Boxer, C4 Therapeutics, Calithera, Cancer Research UK, Diffusion, F-Star, Genmab, Glenmark, GLG, Globe Life Sciences, GSK, Idience, ImmuneSensor, Institut Gustave Roussy, Intellisphere, Kyn, MEI Pharma, Mereo, Natera, Nexys, Novocure, OHSU, OncoSec, Ono Pharma, Pegascy, PER, Piper-Sandler, Prolynx, resTORbio, Theragnostics, Versant, Vibliome, Xinthera, ZielBio; Financial Interests, Personal, Other, University of Texas MD Anderson Cancer Center, where I am Medical Director of the Institute for Applied Cancer Science, which has a commercial interest in DDR and other inhibitors (IACS30380/ART0380 was licensed to Artios): MD Anderson Cancer Center, Institute for Applied Cancer Sciences; Financial Interests, Personal, Stocks/Shares: Seagan; Financial Interests, Institutional, Other, Grant/Research support: Bayer, Cyteir, EMD Serono, GlaxoSmithKline, Karyopharm, Pfizer, Repare, Sanofi, Artios, AstraZeneca, Beigene, BioNTech, Blueprint, BMS, Clovis, Constellation, Eli Lilly, Forbius, F-Star, Genentech, Haihe, ImmuneSensor, Ionis, Ipsen, Jounce, KSQ, Kyowa, Merck, Mirati, Novartis, Ribon Therapeutics, Regeneron, Rubius, Scholar Rock, Seattle Genetics, Tesaro, Vivace, Acrivon, Zenith. C. Lin: Financial Interests, Personal, Other, Travel support: BeiGene, Daiichi Sankyo; Financial Interests, Personal, Advisory Board: Blueprint Medicines, Bristol Myers Squibb, Daiichi Sankyo, Novartis, AbbVie, PharmaEngine, Merck KGaA; Financial Interests, Personal, Invited Speaker: Eli Lilly, Novartis, Roche. K.A. Kornbluth: Financial Interests, Personal, Full or part-time Employment, I work full-time as a Clinical Trial Leader at Novartis: Novartis Institutes for BioMedical Research. J. Mataraza: Financial Interests, Institutional, Full or part-time Employment: Novartis; Financial Interests, Personal, Stocks/Shares: Novartis. L.H. Lee: Financial Interests, Personal, Full or part-time Employment: Novartis. J.A. Otero: Financial Interests, Personal, Full or part-time Employment, I am a full time employee (Senior Clinical Program Leader) in the translational clinical oncology group: Novartis; Financial Interests, Personal, Stocks/Shares: Novartis; Non-Financial Interests, Personal, Member: AACR, ASCO, ASHPO, ASH, American Academy of Pediatrics (AAP). E. Garralda: Financial Interests, Personal, Advisory Board: Genentech, F.Hoffmann/La Roche, Neomed Therapeutics1 Inc, Boehringer Ingelheim, Janssen Global Services, Alkermes, Thermo Fisher, MabDiscovery, Anaveon, Lilly, Hengrui; Financial Interests, Personal, Invited Speaker: Ellipses Pharma, Seattle Genetics, Bristol Mayers Squibb, MSD, F-Star Therapeutics; Financial Interests, Institutional, Funding: Novartis, Roche, Thermo Fisher, AstraZeneca, Taiho. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

202P - The therapeutic cancer vaccine PDC*lung01 induces immune responses with or without anti- PD-1 treatment in patients with non-small cell lung cancer (ID 292)

Presentation Number
202P
Lecture Time
12:30 - 12:30
Speakers
  • Anne Sibille (Liège, Belgium)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

PDC*lung01 (IMP) is a therapeutic cancer vaccine based on an irradiated plasmacytoid dendritic cell line loaded with HLA-A*02:01 restricted peptides (NY-ESO-1, MAGE-A3, MAGE-A4, Multi-MAGE-A, MUC1, Survivin and Melan-A) able to prime and expand peptide-specific CD8+ T cells in vitro and in vivo, and is synergistic with anti-PD-1 (Charles, 2020; Lenogue 2021). Stage II/IIIA (cohort A) or stage IV, PD-L1≥50% (cohort B; +pembrolizumab) non-small cell lung cancer (NSCLC) patients received weekly intravenous and subcutaneous IMP for 6 times at two dose levels. Early safety and clinical activity findings with IMP were presented at ESMO Paris 2022. We report here the analysis of immune response of the first 3 cohorts of patients.

Methods

Several circulating immune parameters were monitored at different times before and after vaccination (W1, W4, W10) using specific assays developed by the sponsor: leukocyte count and determination of peptide-specific CD8+ T cells, for which a limit of quantification (LOQ) was defined to better assess the fold changes of the cell expansion. In addition, tumor microenvironment (TME) was analyzed by multifluorescent immunochemistry.

Results

23 of the 25 patients included received at least 4 doses and were evaluable. No major changes in circulating lymphocyte frequencies (B cells, NK cells, CD4+, CD8+, or Treg T cells) were observed during treatment. In addition, no major cell activation (CD25+, CD54, or DR+) was noted. In contrast, PDC*lung01 induced peptide-specific CD8+ T cell expansion in all 3 cohorts at different levels. An immune response was induced against lung antigens in 33%, 45% and 67% in A1, A2 and B1 cohort, respectively. The intensity of the immune response was proportional to the IMP dose and to the combination with pembrolizumab. When possible, CD8+ T cells were sorted for TCR repertoire analysis, illustrating the modelling and dynamics of the immune response. Detailed findings will be graphically presented at the meeting.

Conclusions

Treatment with PDC*lung01 induces an anti-tumour immune response in a significant number of patients which appears to be enhanced by the combination with pembrolizumab.

Clinical trial identification

NCT03970746.

Legal entity responsible for the study

PDC*line Pharma SAS.

Funding

PDC*line Pharma SAS.

Disclosure

A. Sibille: Financial Interests, Institutional, Advisory Board: Merck Sharp & Dome, AstraZeneca, Bristol Myers Squibb, Roche, Pfizer. J. Plumas: Financial Interests, Personal, Full or part-time Employment: CSO of PDC*line Pharma; Financial Interests, Personal, Stocks/Shares: PDC*line Pharma. I. Demedts: Financial Interests, Personal, Other, invited speaker and participation in Advisory Board meetings: AstraZeneca, BMS, Boehringer, GSK, Lilly, MSD, Novartis, Pfizer, Roche, Takeda. E. Pons-Tostivint: Financial Interests, Institutional, Invited Speaker: AstraZeneca, BMS, Daiichi Sankyo, Sanofi, PDC line, Takeda. M. Collodoro: Financial Interests, Personal, Full or part-time Employment: PDC*line; Financial Interests, Personal, Stocks/Shares: PDC*line. K. Al Badawy: Financial Interests, Personal, Full or part-time Employment: QC Department, PDC*line Pharma SA. C. Duchayne: Financial Interests, Personal, Full or part-time Employment: QC Department, PDC*line Pharma SA. C. Debruyne: Financial Interests, Institutional, Other, Consultant as Medical Director: PDC*line Pharma SAS; Financial Interests, Institutional, Stocks/Shares: PDC*line Pharma SAS. M. Pérol: Financial Interests, Personal, Advisory Board: Roche, AstraZeneca, MSD, BMS, Lilly, Novartis, Takeda, Gritstone, Sanofi, Pfizer, Amgen, Janssen, GlaxoSmithKline; Financial Interests, Personal, Invited Speaker: Roche, AstraZeneca, MSD, BMS, Boehringer Ingelheim, Takeda, Illumina, Pfizer, Roche; Financial Interests, Institutional, Research Grant: AstraZeneca, Roche, Takeda, Boehringer Ingelheim. E.L. Buchmeier: Financial Interests, Personal, Invited Speaker, and congress sponsoring: Takeda; Financial Interests, Personal, Advisory Board: Ipsen; Financial Interests, Institutional, Invited Speaker: BMS, AstraZeneca, Novartis, MSD, Janssen Cilag, PDC Line Pharma, Roche, GSK, Novartis; Non-Financial Interests, Personal, Member: Political Party FDP. K. Cuppens: Financial Interests, Personal, Advisory Board: F. Hoffmann-La Roche, AstraZeneca, Merck Sharp Dohme, Bristol Myers Squibb, Boehringer Ingelheim, Bayer; Financial Interests, Personal, Invited Speaker: Pfizer, Bristol Myers Squibb, Merck Sharp & Dohme, F. Hoffmann-La Roche; Financial Interests, Personal, Expert Testimony: Merck Sharp & Dohme, AstraZeneca. J.F. Vansteenkiste: Financial Interests, Institutional, Advisory Board: AstraZeneca, BMS, Daiichi Sankyo, MSD, Pfizer, Roche, Janssen, Merck, Novartis, PDC*line, Sanofi; Financial Interests, Institutional, Invited Speaker: AstraZeneca, BMS, Novartis, Janssen, Eli Lilly, Roche; Financial Interests, Institutional, Research Grant: MSD. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

203P - Early remodeling of systemic antitumor T cell immunity in head and neck cancer patients treated by chemoradiation (ID 293)

Presentation Number
203P
Lecture Time
12:30 - 12:30
Speakers
  • Benoît Lecoester (Besancon, France)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

A combination of radiotherapy and chemotherapy (CRT: chemoradiation) is particularly used as a cytotoxic therapy in locally advanced cancers. CRT also gained great interest as a combination approach with immune checkpoint inhibitors (ICI). In this regard, our recent report showed in the murine tumor microenvironment that CRT synergistically improves the antitumor T cell immunity suitable for ICI action (Lauret et al, JITC, 2021). Here, we performed a high throughput blood-based immune analysis in head and neck squamous cell carcinoma (HNSCC) treated by CRT.

Methods

Forty-eight HNSCC patients were enrolled in the i-CRT cohort (NCT03117946). Blood samples were collected before (BSL), on day 15, and 3 months after platinum-based CRT. Tumor-specific T cell responses were measured by IFN-γ ELISpot using a mixture of peptides derived from telomerase and NY-ESO-1. Flow cytometry was used for phenotypic and functional characterization (exhaustion, polarization, function) of circulating T cells and for immune suppressive cell monitoring. Bulk RNA sequencing was performed to analyze transcriptomic signatures from blood immune cells at different times.

Results

Twenty-five out of 40 (62%) evaluable patients had pre-treatment circulating anti-tumor T cell responses. A transient decrease in this response occurred two weeks after CRT and most patients (85%) recovered their immune responses 3 months after CRT. These circulating T cells induced after CRT displayed a Th1-polarized profile with upregulation of IFN-γ, TNF-α, and IL-2 expression. Furthermore, early blood expansion of exhausted phenotype T cells co-expressing PD-1+ TIM-3+ TIGIT+ was detected after CRT together with MDSC expansion. Patients with high adaptive antitumor T cell response at baseline showed improved clinical outcomes. Transcriptomic analysis from PBMC support that CRT responders displayed upregulation of inflammatory-, M1 macrophages-, Th1 polarization-, and T cell memory-associated gene signatures while Treg, M2, neutrophils, and T cell exhaustion were downregulated.

Conclusions

These results provide insight into the systemic immunological changes that occurred during CRT which should be taken into account for combining CRT with immunotherapy.

Clinical trial identification

NCT03117946.

Legal entity responsible for the study

UMR1098.

Funding

Ligue Contre le Cancer, the Bourgogne Franche Comte Regional Council, the ARC and the Canceropole EST.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

204P - Tumor immune microenvironment and immunotherapy efficacy in BRAF mutation non-small-cell lung cancer (ID 294)

Presentation Number
204P
Lecture Time
12:30 - 12:30
Speakers
  • Yun Fan (Hangzhou, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Previous small size studies reported BRAF mutated NSCLC patients have comparable sensitivity to immune checkpoint inhibitors (ICIs). However, how BRAF mutation affects the tumor immune microenvironment (TIME) is unknown.

Methods

We performed Nanostring-panel RNA sequencing to evaluate TIME in 57 BRAF mutated and wild-type (WT) NSCLC specimens (cohort A). The efficacy of ICI monotherapy or combined therapies was determined in 417 patients with WT and BRAF mutated NSCLC (cohort B).

Results

We found that BRAF-mutant tumors had similar ratios of CD8+ T cells to Tregs, the balance of a cytotoxicity gene expression signatures and immune suppressive features, and similar ICI-response-related biomarkers to WT NSCLC. A similar pattern of TIME was observed between the BRAF V600E and non-V600E subgroups of NSCLC. Further retrospective study confirmed that treatment with ICI monotherapy or combined therapies resulted in similar overall survival (OS) ( (HR: 0.85; 95% CI, 0.56 to 1.30; p=0.47) and progress-free survival (PFS) (HR: 1.02; 95% CI, 0.72 to 1.44; p=0.91) of patients with WT (n = 358) and BRAF mutant (n=59) NSCLC. Similarly, both patients with BRAF V600E or non-V600E NSCLC had similar responses to immunotherapy.

Conclusions

Our findings indicated that BRAF mutation, including V600 E, did not modulate TIME in NSCLC and therapeutic responses to ICIs. Keywords: BRAF; NSCLC; TIME; immunotherapy.

Legal entity responsible for the study

The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital).

Funding

This work was supported by Beijing Xisike Clinical Oncology Research Foundation (Grant No. Y-XD202002-0251), the National Natural Science Foundation of China (Grant No. 81972718), the Natural Science Foundation of Zhejiang Province (Grant No. LY22H160037) and the Science and Technology Program for Health and Medicine in Zhejiang Province, China (Gran No. 2021KY541).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

205P - Pre-existing tumor host immunity characterization in resected Non-Small Cell Lung Cancer (ID 295)

Presentation Number
205P
Lecture Time
12:30 - 12:30
Speakers
  • Laura M. Pinto (Barcelona, Spain)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Neoadjuvant and adjuvant immune checkpoint blockade (ICB) have recently become standard of care in resectable NSCLC. Yet, biomarkers that inform patient benefit with this approach remain largely unknown. Here, we interrogated the tumor immune microenvironment (TIME) in early-stage NSCLC patients that underwent up-front surgery.

Methods

A total of 185 treatment-naive early-stage NSCLC patients, that underwent upfront surgical treatment between 2006 and 2018 at Hospital del Mar were included. Core biopsies from the surgical specimens were included in a tissue microarray. Immunohistochemistry for CD3, CD4, CD8, CD68, CD80, CD103, FOXP3, PD-1, PD-L1, PDL2 and HLA class II were evaluated by digital image analysis (QuPath software). TIME was categorized into four groups using PD-L1 expression in tumor cells (<1% or ≥1%) and tumor infiltrating resident memory (CD103+) immune cells (using the median as cut-off): 1) PD-L1- /CD103-; 2) PD-L1-/CD103+; 3) PD-L1+/CD103-; 4) PD-L1+/CD103+. TIME characteristics and immune markers were statistically compared based on clinicopathological and molecular features and survival outcomes.

Results

We found elevated levels of T cell markers (CD3+, CD4+, CD8+ cells), functional immune markers (FOXP3+ cells) as well as, higher HLA-II tumor membrane expression in LUADs (p<0.05 for all). In contrast, LUSCs displayed higher percentage of intratumor macrophages (CD68+ cells) as well as, higher PD-L1 and PD-L2 tumor membrane expression (p<0.05 for all). PD-L1 positive (≥1%) LUADs exhibited an augmented infiltration of T cells (CD3+, CD4+, CD8+ cells) along with increase of FOXP3+ cells, resident memory cells (CD103+) and macrophages (CD68+) (p<0.05 for all). Enrichment of T cells (CD3+, CD8+ cells), regulatory T cells (FOXP3+ cells) and macrophages (CD68+ cells) was observed in the CD103+/PD-L1+ group (p<0.05 for all), while T helper cells (CD4+), antigen experienced immune cells (PD-1+) and CD80+ immune cells were higher in the CD103+/PD-L1- (p<0.05 for all).

Conclusions

TIME analysis in resected NSCLC highlighted differences by histology, PD-L1 expression and molecular subgroups. Biomarker studies might aid to individually tailor adjuvant treatment in early-stage NSCLC.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

A. Taus Garcia: Non-Financial Interests, Personal, Other: Roche, BMS, MSD, GSK, AstraZeneca, Pfizer. B. Bellosillo Paricio: Non-Financial Interests, Personal, Other: Amgen, AstraZeneca, Biocartis, Janssen, Merck-Serono, Novartis, Qiagen, F. Hoffman-La Roche, ThermoFisher, Pfizer, BMS. E. Arriola: Non-Financial Interests, Personal, Other: Roche, BMS, MSD, AstraZeneca, Pfizer, Boehringer Ingelheim, Lilly, Takeda. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

206P - Characterization of the immune microenvironment of recurrent and/or metastatic (R/M) squamous cell carcinoma of the head and neck (SCCHN) that progress on or after platinum and anti-PD(L)1 therapies Ð An EORTC IMMUcan sub-project (ID 296)

Presentation Number
206P
Lecture Time
12:30 - 12:30
Speakers
  • Athénaïs Van Der Elst (Brussels, Belgium)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

R/M SCCHN patients that progress after platinum therapy and PD-1 inhibitors represent an unmet medical need. A better characterization of the tumor molecular landscape and immune micro-environment is needed to guide the rational development of new therapeutic approaches.

Methods

EORTC1559 is a biomarker-driven study including patients with R/M SCCHN. Tumor biopsies are collected at time of patient enrolment to perform WES, RNAseq, multiplex immunofluorescence (mIF) and Imaging Mass Cytometry (IMC).

Results

95 R/M SCCHN patients were included (oropharynx 50% (HPV+ 13%), oral cavity 23%, hypopharynx 19%, larynx 8%). All patients progressed on/after platinum therapy and 80% of them (n= 76) progressed on/after anti-PD(L)1. Preliminary transcriptomic analyses on 83 patients indicated that the last regimen given before tumor biopsy had a significant impact on the immune infiltrate. For instance, patients treated with anti-PD(L)1 as last treatment before biopsy (n=38) had a significantly (p= 0.001) higher tumor immune score (ESTIMATE immune score) compared to patients that received other systemic therapies such as taxanes as last treatment. LAG3 expression was significantly higher in the tumor of patients treated with anti-PD(L)1 as last treatment compared to patients never exposed to anti-PD(L)1 (n=15) (padj = 0.001) and to patients pre-treated with anti-PD(L)1 in previous line (n=30) (padj = 0.002). Genomic analyses on n=95 showed median TMB was 4.6mut/MB (range 0.8-46.1). Correlation between TMB and tumor immune score was weak (r= 0.25, p= 0.02). The most frequent oncogenic non-synonymous mutations were found in genes TP53, LRP1B, PIK3CA, FAT1, and CDKN2A. Interestingly, 2 and 3 patients progressing on anti-PD(L)1 had mutations in TAP2 and STK11, respectively, which were not found in anti-PD(L)1-naive patients.

Conclusions

Preliminary results suggest that patients progressing on anti-PD(L)1 administered as last treatment have a higher tumor immune score and LAG3 expression. We will validate our findings on 85 additional patients and integrate our RNAseq and WES analyses with mIF and IMC analyses.

Clinical trial identification

SPECTA: NCT02834884 - 15/07/2016; UPSTREAM (EORTC-HNCG-1559): NCT03088059 - 23/03/2017.

Legal entity responsible for the study

EORTC.

Funding

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

Disclosure

J. Machiels: Financial Interests, Institutional, Advisory Board: Novartis, Pfizer, Roche, Debio, AstraZeneca, Innate, Nanobiotix, Bayer, Merck Serono, Boerhinger Ingelheim, BMS, Pfizer, Cue Pharma, Incyte, Janssen, Johnson & Johnson, ALX Oncology, F-star, Nektar; Financial Interests, Institutional, Advisory Board, uncompensated: MSD; Financial Interests, Institutional, Other, Travel expense: Gilead; Financial Interests, Institutional, Invited Speaker: AstraZeneca, MSD, MSD, iTeos, eTheRNA, Pfizer, Ceylad, MSD, Novartis, KURA, Roche, Lilly, Boehringer, Sanofi Aventis, Incyte, Bayer, Merck - Serono, Janssen, Johnson & Johnson, Amgen, AbbVie, GalxoSmithKline; Non-Financial Interests, Personal, Leadership Role, Chair: EORTC Head and Neck Group. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

207P - Impact of Lymphatic Vessel Derived Oxysterol on anti-tumor immunity (ID 297)

Presentation Number
207P
Lecture Time
12:30 - 12:30
Speakers
  • Mengzhu Sun (Geneva, Switzerland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Bioactive derivatives of cholesterol have been demonstrated to regulate immune cell function and migration, the tumor microenvironment (TME), and consequently tumor progression. Ch25h, a key enzyme involved in cholesterol metabolism and 25- hydroxycholesterol(25-HC) production, was positively correlated with overall survival in melanoma, breast cancer and colon cancer patients. High Ch25h expression is correlated with good response to immunotherapy in melanoma and glioblastoma patients. Expression levels of Ch25h in human tumors are also positively correlated with lymphatic signature. However, the function and mechanism of lymphatic vessel expressed Ch25h in tumor microenvironment remain unknown.

Methods

Mouse B16-OVA-VEGFC melanoma, MC38 colon cancer, and E0771 breast cancer models are used on Ch25h conditional knockout mice with C57/bl6 and immunodeficient background to study the impact of lymphatic endothelial cells (LECs) derived oxysterol on anti-tumor immunity. CpG-based tumor vaccination and activated CD8 T cell adoptive transfer are used to study the impact of LEC-derived Ch25h under immunotherapy.

Results

Our RNA sequencing data on lymphangiogenic murine melanoma showed an increase of Ch25h expression in tumor-associated LECs. Using conditional-knockout mice, we found that Ch25h deficiency in LECs leads to a significant decrease of tumor interstitial 25HC, as well as increased tumor aggressiveness and suppressed immune cell infiltration. Specific deletion of Ch25h in LECs impedes intra-tumoral effector T cell accumulation upon T cell adoptive transfer. Further, mice with LEC-specific Ch25h deficiency show earlier relapse in tumor vaccination and T cell adoptive transfer experiments, whereas littermates maintain the control of tumor growth. Mechanistically, we found that anti-CSFR1 administration upon tumor vaccination promotes tumor relapse in control groups, whereas in contrast, introducing 25-HC intratumorally can prevent tumor relapse in LEC-Ch25h conditional-knockout mice.

Conclusions

Our results suggest a novel active way of lymphatic vessels participating in anti-tumor immunity by regulating anti-tumor immune response through oxysterol.

Legal entity responsible for the study

The authors.

Funding

Swiss Cancer League.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

208P - Sex hormones impact the response to immunotherapy in obese male mice. (ID 298)

Presentation Number
208P
Lecture Time
12:30 - 12:30
Speakers
  • Aurélien Pommier (Geneva, Switzerland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Recent clinical studies suggest that obese cancer patients, especially men, have a better outcome when treated with immune checkpoint inhibitors (ICI) compared to non-obese patients. However, whether obesity and sex are determinant factors influencing the antitumor immune response in the context of ICI is unknown. We hypothesized that the estrogen/androgen balance may play a role in the response to ICI as their level is known to be sex- and BMI-dependent.

Methods

Male and female mice fed with a high-fat diet to induce obesity or with a control diet were subcutaneously injected with B16-F10 melanoma cells. Obese males were treated with the aromatase inhibitor letrozole in order to decrease the estrogen/androgen ratio. ICI efficacy was assessed upon anti-PD-1 treatment by measuring tumor growth and immune cell activation markers using FACS and RNA sequencing analyses. The direct effect of sex hormones on the anti-tumor immunity was tested in vitro using murine bone marrow-derived dendritic cells and CD8+ T cells.

Results

In non-obese mice, ICI decreased tumor growth in females but not in males. In obese mice, males and females showed the similar response to non-obese female to ICI treatment indicating that obesity sensitized to ICI in male mice only. Tumor gene expression analyses prior to ICI therapy revealed that obesity induced a female-like tumor gene expression profile in males characterized by the enrichment of differentially expressed genes involved in response to estrogen signaling. Treatment with letrozole abolished the efficacy of ICI in obese males, demonstrating the pivotal role of estrogens in the ICI sensitivity of obese males. In vitro experiments demonstrated that estrogen treatment enhanced the tumor immune response mainly by stimulating the antigen presentation capacity of dendritic cells.

Conclusions

These results demonstrate that the estrogen/androgen ratio can determine the response to ICI in obese mice and support the hypothesis that sex hormones may contribute to ICI sensitivity in obese male patients with melanoma. This concept opens the path to investigate whether serum sex hormones levels could be used as biomarkers for response to ICI and whether targeting the hormonal balance may be a relevant therapeutic strategy to improve the response to ICI.

Legal entity responsible for the study

The authors.

Funding

Ligue suisse contre le cancer.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

209P - Single-cell profiling of tumor-associated neutrophils in advanced non-small cell lung cancer (ID 299)

Presentation Number
209P
Lecture Time
12:30 - 12:30
Speakers
  • Jinpeng Shi (Shanghai, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Neutrophils act as a non-negligible regulator in the initiation and progression of malignancies, playing bifacial roles in the process. Thus, to understand the heterogeneity of tumor-associated neutrophils (TANs) comprehensively in advanced non-small cell lung cancer (NSCLC) at single-cell resolution is necessary and urgent.

Methods

We applied single-cell RNA-sequencing (scRNA-seq) to portray the subtype-specific transcriptome landscape of TANs in advanced NSCLC using nine freshly obtained specimens. The scRNA-seq data were further processed for pseudo-time analysis to depict the developmental trajectory of TANs. Meanwhile, the interplay between TANs and other cell types within tumor microenvironment (TME) was revealed by intercellular interaction analysis.

Results

Seven distinct TAN subtypes were defined, of which, the N7 cluster was the most distinctive one exhibiting unique and independent characteristics of gene signatures and potential functions. N1 and N5 clusters were considered well differentiated and mature neutrophils based on CXCR2 expression and pseudo-time patterns, and both accounted for relatively high proportions in lung adenocarcinoma. Besides, certain genes related to neutrophil differentiation were discovered, and we also found that TAN subtypes interacted most closely with macrophages through chemokine signaling pathways within TME.

Conclusions

Our study refined TAN subtypes and mapped the transcriptome landscape of TANs at single-cell resolution in advanced NSCLC, collectively indicating the heterogeneity of TANs in NSCLC. Neutrophil differentiation- and maturation-related genes were also disclosed, which shed light on different functions of subclones in tumor immune escape, and may further provide novel targets for immunotherapy.

Legal entity responsible for the study

The Ethical Committee of Shanghai Pulmonary Hospital Affiliated to Tongji University.

Funding

This work was supported by grants from National Natural Science Foundation of China (No. 81902314), Natural Science Foundation of Shanghai (No. 20ZR1447100), and Clinical Research Plan of Shanghai Hospital Development Center (No. SHDC2020CR4001).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

210P - Anti-CTLA-4 overcomes inhibitory effect of PD1high regulatory T cell on circulating antitumor CD4+ Th1 response in patients treated by anti-PD(L)-1 (ID 300)

Presentation Number
210P
Lecture Time
12:30 - 12:30
Speakers
  • Amélie Marguier (Besancon, France)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Evidence supports the crucial involvement of CD4+ T cells in anti-tumor immunity and immunotherapy (IO). In this study, we studied the systemic antitumor CD4 Th1 response in patients receiving anti-PD(L)-1 therapy.

Methods

Patients with metastatic cancers receiving anti-PD(L)-1 therapy (n=117) were enrolled in an immunomonitoring cohort (ITHER study, NCT02840058). Blood samples were collected at baseline, one month, and three months after IO. Tumor-reactive CD4+ Th1 response was measured by IFN-y ELISPOT, after lymphocytes culture with pan-HLA class II binding peptides derived from telomerase, as previously described (Laheurte et al., BJC 2019, Nardin et al., J Inves Dermatol 2022). Phenotypic analysis of circulating immune cells was performed by flow cytometry.

Results

While an expansion of circulating antitumor CD4+ Th1 response was associated with favorable clinical outcomes, its impairment after IO appeared deleterious. Searching for an explanation for this unexpected loss of tumor-specific CD4+ T cells, we ruled out a potential apoptotic effect resulting from activation of the PD1 signaling. However, we identified a role of Foxp3+Treg subset overexpressing PD1 (PD1high Treg). Indeed, a high level of circulating PD1high Treg was preferentially found in these patients. PD1high Treg displayed overexpression of inhibitor receptors such as CTLA-4, ICOS, and GITR, which are upregulated after IO. The addition of anti-CTLA-4 mAb in vitro restored the function of antitumor CD4+ T cells, supporting the suppressive effect exerted by circulating PD1high Treg during IO.

Conclusions

Thus, activation of PD1high Treg inhibits antitumor T cell response in patients treated by anti-PD(L)-1 therapy. These results support blood-based monitoring of tumor-reactive CD4+ Th1 response together with PD1High Treg for patient management and treatment decision.

Clinical trial identification

NCT02840058.

Legal entity responsible for the study

UMR1098.

Funding

Ligue contre le cancer, the Bourgogne Franche Comté regional council, the ARC and the Cancéropôle EST.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

211P - A new role for CXCL3 in shaping the metastatic tumor microenvironment (ID 301)

Presentation Number
211P
Lecture Time
12:30 - 12:30
Speakers
  • Camilla Wolowczyk (Trondheim, Norway)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The type and activity of immune cells infiltrating solid tumors influence patient prognosis and survival. Cancer cells secrete molecules such as chemoattractants and immunomodulatory mediators that attract and polarize the infiltrating immune cells. The combination of such compounds contributes to forming an immunosuppressive tumor microenvironment that promotes tumor growth. Therefore, we hypothesized that non-metastatic and metastatic cancer cells will recruit and polarize different types of immune cells. Our aim was to investigate the potential differences in immune cell recruitment to tumors with different metastatic potential and to uncover signaling compounds that contribute to making an immunosuppressive environment.

Methods

Using RNA sequencing, we compared non-metastatic 67NR and metastatic 66cl4 cells and primary tumors, originating from the 4T1 mammary tumor model. We used several molecular biological methods, including flow cytometry and immunohistochemistry to determine the immune cell heterogeneity within the tumors. Further, we performed qPCR, ELISA and shRNA-mediated knockdown to elucidate the role of our proteins of interest.

Results

Analysis of primary tumor transcriptomes and flow cytometry of dissociated tumors formed by 67NR cells and 66cl4 cells revealed clear differences in tumor landscapes of the two tumors. The most striking difference was the high infiltration of myeloid cells in the metastatic tumors. Consistent with this result, we found that 66cl4 cells produce and secrete high levels of the chemokine CXCL3, a potent chemoattractant of neutrophils. We used CXCL3 knock-down to determine its role in tumors. The loss of CXCL3 expression resulted in a significant decrease in infiltrating neutrophils and a shift in the macrophage polarization in the primary tumors.

Conclusions

Our results highlight the essential role of CXCL3 in shaping the tumor microenvironment. When CXCL3 is knocked-down it shifts the tumor microenvironment towards a less immunosuppressive state, making CXCL3 a potential new therapeutic target.

Legal entity responsible for the study

The authors.

Funding

Norges Forskningsråd, Norske Kvinners Sanitetsforening, Helse Midt-Norge.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

212P - Augmenting Cancer Immunotherapy by Targeting the Fes Kinase (ID 302)

Presentation Number
212P
Lecture Time
12:30 - 12:30
Speakers
  • Brian J. Laight (Kingston, ON, Canada)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immunogenic cell death (ICD) causes release of tumour antigens and damage associated molecular patterns which recruit and activate antigen presenting cells (APCs) by binding to their pattern recognition receptors (PRRs). This leads to their production of the pro-inflammatory cytokines required for adaptive immune cell activation (e.g., Cytotoxic T Lymphocytes [CTLs]). The non-receptor tyrosine kinase Fes, which is abundantly expressed in innate immune cells, dampens innate immune responses by inhibiting PRR signaling. We hypothesize that this same Fes-dependent mechanism which serves to limit the consequences of overactive innate immunity, such as septic shock, causes Fes to inhibit successful anti-cancer immunotherapy by preventing efficient priming of cancer specific CTLs by APCs.

Methods

Immunoblot analysis assessed PRR signaling in wildtype (WT) or fes-/- bone marrow derived macrophages (BMDMs). C57BL/6 WT or fes-/- mice orthotopically engrafted with E0771 triple-negative breast cancer or B16-F10 melanoma cells were treated with vehicle or doxorubicin and assessed for tumour growth and survival. Tumours and spleens were harvested to analyze immune profiles by flow cytometry. In separate experiments, E0771 and B16-F10 tumor bearing WT and fes-/- mice were treated with doxorubicin and either IgG control or anti-PD-1 antibody.

Results

Fes-/- BMDMs display stronger PRR signaling in vitro compared to WT following LPS stimulation. In vivo, we show increased tumour control and survival in Fes-/- mice compared to WT, which was further enhanced by stimulating ICD with doxorubicin. Fes-/- mice demonstrated increased CTL and NK cell activation and PD-1 positivity, which was enhanced by doxorubicin, indicating a novel role of Fes in regulating CTL and NK cell activation. Additionally, we found a shift from M2- to M1-polarized tumour associated macrophages in Fes-/- versus WT mice. Finally, when treated with anti-PD-1 antibody, Fes-/- mice demonstrated greater tumour control and survival than WT.

Conclusions

Consistent with improved overall- and disease-free survival observed in low Fes-expressing cancer patients, our results identify Fes as a potential novel therapeutic target to enhance anti-cancer immunotherapy.

Legal entity responsible for the study

The authors.

Funding

Canadian Cancer Society, Canadian Breast Cancer Foundation, Canadian Institutes of Health Research.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

213P - Type-1 diabetes restricts melanoma growth by reprogramming intra-tumoral T cell metabolism (ID 303)

Presentation Number
213P
Lecture Time
12:30 - 12:30
Speakers
  • Anirban Sarkar (Kolkata, India)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Epidemiological studies from Bendix et al. (2016) with 5 countries and 9000 type-I diabetes mellitus (T1DM) patients with cancers showed reduced risk in melanoma, breast and prostate cancer and increased risk of oesophagus, stomach, colon and liver cancer, suggested differential consequences of T1DM with cancers of different etiopathology. However, prospective studies to decipher the possible mechanism are not well documented. In both T1DM and cancer, CD8+ T cells plays crucial role and faces functional as well as metabolic alterations. Objective of this study was to evaluate the possible modulatory effect of pre-existing T1DM in melanoma growth, systemic immune landscape and T cell metabolism.

Methods

Murine T1DM model was established by using intra-peritoneal injection of streptozotocin (STZ) to C57BL/6J mice. B16F10 cells were inoculated to T1DM and non-diabetic control mice. Tumor progression and host survival was closely monitored following establishment of B16 melanoma. RT-PCR, Western-blot, Flow-cytometry and LDH release assay were used to study different immune cells, metabolic pathways etc. Athymic nude mice were used to examine the possible involvement of immune system in T1DM associated cancer progression.

Results

Pre-existence of T1DM showed restricted melanoma growth and survival benefits in murine host, however, such effect was found to be mitigated in immune-compromised mice. Significant intra-tumoral infiltration of IFNg+PerforinhighGranzymeBhigh CD8+ T cells were observed with reduced Tregs and MDSCs in T1DM host compared to control. Moreover, pre-existence of T1DM modulates extracellular acidification rate (ECAR) and expression of enzymes associated with glucose-metabolism like PCX1, LDH, PKM2 in tumor infiltrated CD8+T cells. Obtained results also pointed out the involvement of IGF1-mTOR signalling axis within CD8+-effector T cells in regulation of T1DM associated tumor growth restriction.

Conclusions

Pre-existing T1DM promotes CD8+ T cell dependent murine melanoma growth restriction which significantly increases tumor host survival. IGF1-mTOR signalling axis could be exploited in cancer patients with or without T1DM for therapeutic benefit.

Legal entity responsible for the study

R. Baral.

Funding

University Grants Commission, New Delhi, India. Indian Council of Medical Research, New Delhi, India.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

214P - Location matters: oral cancer cells at the tumor invasive border that express GARP exclude immune cells (ID 304)

Presentation Number
214P
Lecture Time
12:30 - 12:30
Speakers
  • Rieneke Van de Ven (Amsterdam, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Failure to respond to anti-Programmed Death receptor-1 (PD-1) treatment has been linked to high levels of transforming growth factor (TGF)-β in the tumor microenvironment (TME). Since TGF-β requires activation to be functional, we evaluated the expression of the receptor glycoprotein-A repetition predominant (GARP), which facilitates TGF-β activation, on oral squamous cell carcinoma (OSCC) surgical resections.

Methods

Immunohistochemistry (IHC) for GARP and CD45 was performed on sixty-eight FFPE OSCC resection specimens. Presence (negative vs. positive) and expression pattern (diffuse or marginal) were determined. Expression patterns were linked to patient overall survival. Gene expression profiling was performed, using a Tumor-signaling (TS)360 panel, on RNA isolated from GARP negative, -diffuse and –marginal OSCC specimens (n=4 each), matched for clinicopathological features.

Results

We observed poor clinical outcome when OSCC expressed GARP on the outer rim of tumor islands located at the invasive tumor border (marginal localization) compared to tumors that lacked GARP expression or diffusely expressed GARP throughout the tumor. Gene expression analysis revealed that GARP-marginal tumors beside expressing more TGF-β1 and GARP, had enhanced expression of genes regulating “tumor-promoting inflammation”, “NF-κB signaling” and “Epithelial-to-Mesenchymal transition”. Based on the expression data, GARP-marginal tumors displayed increased CD45 immune cell infiltration, with myeloid cells having the most abundant cell scores. Quantifying CD45 in consecutive sections from GARP IHC revealed that while CD45 was present in the TME of GARP-marginal tumors in similar levels as GARP-negative tumors, significantly fewer CD45+ cells were able to penetrate GARP-marginal tumor islands compared to GARP-negative tumor islands within the same tumor specimen.

Conclusions

Our data suggest that infiltrating oral cancers utilize the GARP/TGF-β axis to support a pro-tumor TME and exclude infiltration of immune cells within tumor fields.

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 Poster Display session

215P - Proteogenomics to guide neoantigen discovery in non-small cell lung cancer (ID 305)

Presentation Number
215P
Lecture Time
12:30 - 12:30
Speakers
  • Yaroslav Kaminskiy (Stockholm, Sweden)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Targeted Immunotherapy is entirely dependent on the identification of suitable TAAs or even better, of TSAs, also referred to as neoantigens. This is currently an extremely challenging task, mainly based on using experimental data from DNA and RNA sequencing technologies to generate large lists of neoantigen candidates, followed by heavy bioinformatics to predict how well each neoantigen candidate would progress through the MHC processing and presentation pathway. Up till now, most of the candidates generated in this way fail to elicit any relevant immunogenicity. The vast majority of candidates likely fail because, being based on single point mutations (as most of them are with the current state-of-the-art), and thus single amino acid variants (SAAVs), they are not that different from the respective non-mutated self-antigens tolerated by the immune system. Thus, very likely there is no T cell clone available capable of recognizing the putative neoantigen. Other problems could be due to intricacies of the MHC processing and presentation pathway, such as absence of a suitable HLA type able to present the putative neoantigen.

Methods

HiRIEF LC-MS, RNA-seq, DNA-seq (+ gene panel seq), bioinformatics pipeline (python and R).

Results

We used our proteogenomics-based pipeline to find potential neoantigens in a cohort of 141 NSCLC patients. Identified putative neoantigens were next in silico validated in 3 public NSCLC proteomic datasets and filtered based on 30 normal-tissue proteomic datasets to exclude unannotated normal peptides. After MHC-I binding prediction, our analysis revealed a list of high-confidence neoantigen candidates which will be subsequently validated in vitro and in vivo.

Conclusions

We identified a list of high-confidence non-canonical peptides in the cohort of 141 NSCLC patients which will be further validated to choose most promising candidates to advance into clinic.

Legal entity responsible for the study

Lehtio group (ONKPAT, Karolinska Institutet).

Funding

Karolinska Institutet, Scilifelab.

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster Display Poster Display session

216P - Relationship between regulatory T lymphocytes (Treg) - related genes and pathological response to neoadjuvant docetaxel - carboplatin in early-stage Triple Negative Breast Cancer (TNBC) (ID 306)

Presentation Number
216P
Lecture Time
12:30 - 12:30
Speakers
  • Rocío Martín Lozano (Madrid, Spain)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

An immune infiltrate rich in Treg is associated with worse prognosis in breast cancer, although there is controversy regarding their role and clinical relevance among the different tumor subtypes. This project aims to analyze the expression of Treg-related genes and its relationship with pathological response to neoadjuvant docetaxel-carboplatin in a cohort of patients with early-stage TNBC.

Methods

Within a prospective multicenter study, we analyzed 221 pre-treatment FFPE tumor samples from patients with TNBC who had received neoadjuvant docetaxel-carboplatin. By RNA sequencing, we analyzed 23 genes related to Treg. By IHC, we analyzed the presence of tumor-infiltrating lymphocytes (TILs). We performed univariate logistic regressions of the correlation of the expression of these genes with the response and multivariate analysis of the genes whose expression showed a statistically significant correlation with response.

Results

5 genes presented a higher expression in responders vs. non-responders in multivariable analysis: FOXP3 (p = 0.04), CTLA4 (p = 0.005), CD4 (p = 0.013), CD274 (p = 0.03), and FCRL1 (p = 0.019). The differences in CD8 expression (RNAseq) and the presence of TILs according to response were also studied. Responders had more TILs and higher CD8 expression. Ratios between FOXP3 with CD4, CD8, and TILs were calculated. In univariate analysis, responders had lower FOXP3/CD8A ratio (p = 0.102), FOXP3/CD8B ratio (p = 0.0352), and higher sTILs/FOXP3 ratio (p = 0.0104). These differences were not significant in the multivariate analysis.

Conclusions

In our cohort, the overexpression of genes related to Treg is associated with a better pathological response to treatment. This is observed in context of a greater immune infiltrate. A higher FOXP3/CD8 ratio is associated with a worse response in the univariate analysis, without reaching statistical significance in the multivariate analysis. Additional studies are needed to identify the mechanisms by which Treg modulate response in TNBC, whether their action is always immunosuppressive, or why they exert a different effect depending on the breast cancer subtype.

Legal entity responsible for the study

Fundación para la Investigación Biomédica del Hospital Gregorio Marañón.

Funding

This study has been funded by Instituto de Salud Carlos III (ISCIII) through the projects \"PI 15/00117\" and \"PI18/01775\" and co-funded by the European Union and by Centro de Investigación Biomédica en Red de Salud Carlos III CiberONC-ISCIII.

Disclosure

R. Martín Lozano: Financial Interests, Personal, Training: Sanofi, Vifor Pharma, Lilly. T. Massarrah: Financial Interests, Personal, Advisory Board: AstraZeneca, Novartis, Roche, GSK; Financial Interests, Personal, Training: Novartis, AstraZeneca. C. Bueno Muiño: Financial Interests, Personal, Invited Speaker: Roche, Novartis, Lilly, MSD, AstraZeneca, GSK; Financial Interests, Personal, Training: Roche, Novartis , Clovis, Pfizer. S. Lopez-Tarruella Cobo: Financial Interests, Personal, Advisory Board: Celgene, Novartis, Pierre Fabre, Pfizer, Roche, AstraZeneca, Daiichi Sankyo, MSD, Seagen, Gilead, Lilly; Financial Interests, Personal, Invited Speaker: Novartis, Roche, Lilly. M. Martin Jimenez: Financial Interests, Personal, Advisory Board: AstraZeneca, Lilly, Roche/Genentech, Daiichi Sankyo; Financial Interests, Personal, Invited Speaker: Pfizer, AstraZeneca, Lilly, Novartis, Roche/Genentech; Financial Interests, Institutional, Research Grant: Novartis, Roche, Puma; Non-Financial Interests, Personal, Invited Speaker: TRIO; Non-Financial Interests, Personal, Leadership Role: GEICAM. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

217P - Characterization of immune cells in the tumor microenvironment of advanced bladder cancer (ID 307)

Presentation Number
217P
Lecture Time
12:30 - 12:30
Speakers
  • Emilie Flaberg (Stockholm, Sweden)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Cisplatin-based combination chemotherapy (CHT) improves survival in patients with muscle-invasive (MIBC) and metastatic urothelial cancer (mUC). Here we map the cellular landscape in MIBC and mUC and explore its associations to outcome.

Methods

We have investigated the amount, proportions and the spatial relationship of tumor and immune cells in MIBC patients receiving neoadjuvant chemotherapy (NAC, n=100) and mUC patients treated with CHT (n=87). We performed multiplex immunofluorescence (mIF) of treatment naive samples on a tissue microarray (TMA), stained with antibodies for CD3, CD163, PD-L1, pSTAT1, CD25, Ki-67, epithelial cells and nuclei. We have developed an image analysis pipeline including machine learning for nuclei cell segmentation and downstream single cells analysis.

Results

Our data using an 8-plex mIF panel confirms that tumor infiltrating lymphocytes is linked to improved prognosis in MIBC. T-cell counts can separate patients into subgroups showing better and worse survival. A higher number of total CD3+ T cells and a higher number of CD3+ T-cells infiltrated into the tumor area associates with better survival in NAC-patients, while this pattern cannot be seen in the mUC patient group. Similarly, a higher number of stroma-infiltrating CD163+ macrophages seems to be a good prognostic factor for the NAC cohort. This is in contrast to the mUC group where a significant worse survival was observed for higher CD163+ cell counts in stroma. We also observed a contrasting survival trend in NAC vs mUC patients with respect to PD-L1-positivity when assessing PDL1-status by a mIF-score mimicking the combined-positive score used for immunohistochemical PD-L1 testing in clinical routine (DAKO 22C3).

Conclusions

mIF enables the exploration of the cellular landscape in advanced urothelial cancer towards clinical endpoints. The spatial distribution of CD3+ and CD163+ cells in tumor and stroma have different prognostic value in mUC and NAC, with possible implications on both treatment and follow-up. Moreover, our results illustrate the dynamic interplay between immune cells and tumor cells during tumor progression.

Anders Ullén and Päivi Östling have equally contributed to this work and share last authorship.

Legal entity responsible for the study

Karolinska Institutet.

Funding

Knut och Alice Wallenberg foundation, Swedish Cancer Society, Swedish research council, Cancer society Stockholm, King Gustaf V Jubilee fund.

Disclosure

R.M.P. Turkki: Financial Interests, Personal, Full or part-time Employment: AstraZeneca. L. Paavolainen: Financial Interests, Institutional, Invited Speaker: Orion Pharma. F. Costa Svedman: Non-Financial Interests, Personal, Principal Investigator, Principal investigator för clinical trials in our clinic: MSD; Non-Financial Interests, Personal, Member, SFUO Svensk Förening för Urologisk Onkologi: Swedish Union for Uro-oncology; Non-Financial Interests, Personal, Member, SOF Svensk Oncologisk Förening: Swedish Society of Ocology. O.P. Kallioniemi: Financial Interests, Personal, Invited Speaker: AstraZeneca. A. Ullén: Financial Interests, Personal and Institutional, Funding, Research funding for RWD study on avelumab. Participation in advisory board: Merck. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

218P - A symbiotic relationship between persistent and emerging resistant oligo-clones dictates resistance to immunotherapy in pancreatic cancer (ID 308)

Presentation Number
218P
Lecture Time
12:30 - 12:30
Speakers
  • Chanthirika Ragulan (London, United Kingdom)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immunotherapy has had a limited clinical benefit in pancreatic ductal adenocarcinoma (PDAC). In a pre-clinical mouse model of PDAC treated with glucocorticoid-induced TNFR-related protein (GITR) agonist, the tumours showed nondurable and heterogeneous responses to the GITR agonist. We hypothesised that resistance to GITR agonist is associated with a unique symbiotic interaction between persistent and emergent tumour clones resistant to therapy response.

Methods

Cutting-edge DNA-barcoding technology was applied to study heterogeneity in vivo using 3275 KPC (LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx-1-Cre) cell line. Two distinct oligo-clones representing different morphologies and molecular characteristics (assessed using Western blot and gene expression profiling) were selected. Response to GITR agonist and vehicle was assessed after orthotopic injection of cells into immunocompetent mice in vivo.

Results

DNA-barcoding experiment showed reduced clone size and heterogeneity in 3275 orthotopic tumours treated with GITR agonist compared with the control. This clonal heterogeneity was partly associated with two oligo-clones: A) an epithelial-like and B) a mesenchymal-like oligo-clones from parent 3275 cell line. The epithelial-like in vivo mouse tumours had increased viral-mimicry pathways and immune infiltrations, whereas the mesenchymal-like tumours showed immunodesert and epithelial-mesenchymal transition gene profiles. When these oligo-clones were mixed and co-cultured in vitro or injected in vivo in mice, we observed a unique symbiotic relationship between them. Moreover, treatment of the mixed tumours showed that while mesenchymal-like cells persisted with intrinsic resistance, epithelial-like cells emerged as adaptive resistance after treatment.

Conclusions

This study showed symbiotic interaction between persisted and emerging oligo-clones supporting each other leading to heterogenous and non-durable responses after treatment with GITR agonist. Further studies are required to understand the interaction between these oligo-clones to identify additional potential targets to perturb the symbiotic interaction for durable immunotherapy responses.

Legal entity responsible for the study

The authors.

Funding

Ian Harty Charitable Trust.

Disclosure

A. Sadanandam: Financial Interests, Institutional, Research Grant: BMS. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

219P - Spatial sequencing of T cell repertoire in breast cancer (ID 309)

Presentation Number
219P
Lecture Time
12:30 - 12:30
Speakers
  • Ahmad Merhi (Gosselies, Belgium)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Analysis of T-cell repertoire (TCR) has been proposed as a new approach to better understand the adaptive immune response in cancer diseases. Next generation sequencing of the TCR has proven to be a powerful approach to characterize this repertoire, with potential applications to select and follow patients who benefit from immunomodulatory therapies. However, current TCR sequencing in tumor tissues is based on bulk RNA or DNA extraction, which does not provide neither the functional information at the clonotype level nor the spatial context. Here, we explore a new method which provides the spatial localization of individual T cell clonotypes in tumor samples.

Methods

Frozen tumor sections (12 μm) were positioned on a 10x Visium spatial gene expression slide, and mRNA was captured in spatial spots of 55 μm diameter. Each transcript was tagged with a Unique Molecular Identifier (UMI) and a spatial barcode during an in situ reverse transcription. Starting from the generated cDNA, the TRB genes were amplified using a multiplex PCR with a set of primers positioned on the TRBV genes. TRB libraries were sequenced on the Illumina MiSeq platform. Analyzed samples were 3 mm diameter Tru-Cut tumor biopsies collected before and during neoadjuvant treatment of locally advanced breast cancer patients with informed consent.

Results

With our spatial TCR sequencing protocol we identified polyclonal T cell repertoires and localized individual T cell clonotypes on breast tumor slides. We observed distinct predominant clonotypes and accurately determined their spatial distribution before and during treatment.

Conclusions

This proof of concept work demonstrates the feasibility of spatial TCR sequencing in tumor biopsies, providing a new method to explore the spatial organization of T cell clonotypes in the tumor microenvironment, explore T cell functional profiles at the clonal level and follow modifications during tumor development and treatments.

Legal entity responsible for the study

The authors.

Funding

Seqalis.

Disclosure

F.P. Duhoux: Non-Financial Interests, Institutional, Advisory Role: Roche, Pfizer, AstraZeneca, Lilly, Novartis, Amgen, Daiichi Sankyo, Pierre Fabre, Gilead sciences, Seattle Genetics, MSD Oncology; Non-Financial Interests, Personal, Other: Amgen, Roche, Teva, Pfizer, Daiichi Sankyo/AstraZeneca. J. Carrasco: Non-Financial Interests, Institutional, Research Grant: AstraZeneca, Roche; Non-Financial Interests, Personal, Advisory Board: Seqalis. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

220P - HDAC1 inhibition sensitizes refractory tumor cells to anti-PD-1 therapy by upregulation chemokine expression and stimulating T cells infiltration into tumor beds. (ID 310)

Presentation Number
220P
Lecture Time
12:30 - 12:30
Speakers
  • Kyungjin Boo (Seoul, Korea, Republic of)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immune checkpoint inhibitors (ICIs), exemplified by anti-PD-1 are promising treatments for many tumors. However, the majority of patients lack effective responses because of the emergence of immune-refractory tumors that disrupt the amplification of antitumor immunity. In many cases, one of the major causes of resistance to these agents is the limited tumor penetrance of effector T cells. Durable clinical responses using anti–PD-1 have been associated with T cell–inflamed tumor microenvironment (TME) favoring the infiltration of functional cytotoxic T lymphocytes (CTLs). Efforts to identify therapeutic approaches able to turn immune cold tumors hot by enhancing immune cell infiltration into the tumor bed are currently undertaken.

Methods

We analyzed transcriptomic data on patients with cancer treated with anti-PD-1and newly established preclinical mouse models refractory to anti-PD-1. The effect of genetic ablation or pharmacological inhibition of HDAC1 were approached by transwell T cell migration assay, Immunostaining, flow cytometry and RT-qPCR. We confirmed the enhanced therapeutic effect against tumors when combining anti-PD-1 with HDAC1 inhibitor in refractory tumor mouse models.

Results

HDAC1 is a key factor for insufficient T cell infiltration and resistance to CTL-mediated killing in immune-refractory tumors. Ablation of HDAC1 significantly upregulates the expression of CXCL10 and MCL-1 in the refractory tumor cells, enforcing T cell infiltration and CTL-mediated apoptosis of the tumor cells. In immune-refractory tumor models, tumors were substantially smaller in size and tumor burden in the mice treated with both anti-PD-1 and HDAC1 inhibitor compared with those treated with either agent alone. The overall numbers of CD8 T cells and tumor-reactive CD8 T cells expressing granzyme B were significantly higher in the combination treatment group than in the other treatment groups.

Conclusions

Preliminary data suggest HDAC1 inhibition switched the immune phenotypes from an immune-refractory to an immune-stimulatory feature by simultaneously reversing NANOG-mediated immune-refractory states, thereby overcoming the resistance to PD-1 blockade.

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 Poster Display session

221P - The role of TNF-alpha in thymus dysfunction during Acute myeloid leukemia (ID 311)

Presentation Number
221P
Lecture Time
12:30 - 12:30
Speakers
  • Meriem BEN KHOUD (Villeneuve D'ascq, France)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Acute myeloid leukemia (AML) is characterized by an increased proliferation of hematopoietic progenitors or precursors (blasts) of the different myeloid lineages. Studies performed in AML-affected patients revealed a T-cell immunodeficiency, characterized by a decreased number of peripheral T lymphocytes' TRECs and a restricted repertoire.

Methods

To study thymus dysfunction during AML, we used an AML mouse model in which we previously showed a thymic atrophy notably due to an increased cell death among double-positive (CD4+CD8+) thymocytes. To better understand this massive thymocytes’ loss, we collected ex vivo thymi from control and leukemic mice and immunophenotyped them for cell death. In parallel, we also assessed for the expression of different actors of cell death signaling pathways by RT-qPCR or Western Blotting.

Results

When comparing leukemic to control mice, there was a significant increase in the expression of Mlkl gene, phosphorylated MLKL and RIPK3 proteins and TNF-alpha receptors on double-positive (CD4+CD8+) thymocytes. These findings revealed an abnormal cell death of double-positive (CD4+CD8+) thymocytes by necroptosis (in addition to apoptosis) during AML. Such cell death was also observed in vitro using cultured wild-type thymocytes and recombinant TNF-alpha protein in the presence or absence of apoptosis inhibitors.

Conclusions

Thus, we demonstrated that TNF-alpha plays a deleterious role in thymic function during AML by contributing to extensive thymocytes’ loss. Further investigations will help to better characterize its impact on the peripheral T-cell repertoire and antigens recognition.

Legal entity responsible for the study

Meriem Ben Khoud, Nathalie Jouy, Bruno Quesnel and Carine Brinster.

Funding

Canther UMR 9020 CNRS UMR 1277 INSERM.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

222P - Platinum-based chemotherapy attenuates the CD8 T cell proliferative response to concomitant PD-1 blockade (ID 312)

Presentation Number
222P
Lecture Time
12:30 - 12:30
Speakers
  • Annapaola Mariniello (Orbassano, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Combination of chemotherapy (CT) with PD-1 blockade is a front-line treatment for lung cancer. Our aim was to explore CT effects on CD8 T cells that proliferate in response to concomitant PD-1 blockade.

Methods

To reproduce the exhausted immune landscape of cancer, we used a well-established murine model of chronic lymphocytic choriomeningitis virus (LCMV) infection. At day 45 post-infection, mice were assigned to 4 treatment groups (untreated, CT, aPD-L1, combo). The CT regimen consisted of cisplatin (2.5 mg/kg) and pemetrexed (300 mg/kg). CT and aPDL1 were given every 3 days for 2 weeks. This regimen was tested also in the syngeneic CT26 tumor model and T cell phenotyping was performed after 4 doses (day 20 post-tumor implantation).

Results

In the LCMV model, LCMV-sp CD8 T cells in lymphoid and non-lymphoid tissues were lower in the combo vs aPD-L1. Proliferation of LCMV-sp CD8 T cell was higher in combo than in chemo and untreated, but lower than in aPD-L1. Frequency of stem-like LCMV-sp CD8 was higher in chemo group vs the others, with no differences in absolute numbers. The effector subsets, both the transitory and terminally differentiated, were more vulnerable, resulting in decreased IFN-g production and viral control in the combo compared to the aPD-L1 group. In the tumor model, CT and/or aPD-L1 delayed tumor growth. Compared to the untreated group, tumor infiltrating Ag-sp and PD-1+ CD8 T cells were significantly higher not only in the aPD-L1 and combo groups, but also in the chemo group. Though non-significant, frequency of proliferating Ag-sp CD8 T cells was lower in combo vs aPD-L1 group. Frequency of stem-like cells was low, without significant differences across groups.

Conclusions

Chemotherapy attenuates the proliferation of effector cells mediated by concomitant PD-1 blockade. The preservation of the stem-like subset, that provide the proliferative burst needed for an effector T cell response, suggests that chemotherapy toxicity on CD8 T cells is likely transient and reversible after discontinuation of chemotherapy.

Legal entity responsible for the study

The authors.

Funding

Emory University, Emory Vaccine Center.

Disclosure

S. Novello: Financial Interests, Personal, Invited Speaker: AstraZeneca, MSD, Eli Lilly, Novartis, Beigene, Amgen; Financial Interests, Personal, Advisory Board: BI, BMS, Pfizer, Takeda, Roche, Sanofi, Amgen; Financial Interests, Institutional, Invited Speaker, IIT: MSD, BI; Non-Financial Interests, Personal, Leadership Role, president of this european advocacy: WALCE. G. Scagliotti: Financial Interests, Personal, Other, Honoraria/Consultant: AstraZeneca, Eli Lilly, MSD, Pfizer, Roche, J&J, Takeda, BeiGene, Bayer. S.S. Ramalingam: Financial Interests, Institutional, Other, Consultant: Amgen, BMS, Merck, AstraZeneca, Takeda, Eisai, Daiichi Sankyo, GSK; Financial Interests, Personal, Other, Editor in Chief, Cancer journal: American Cancer Society; Financial Interests, Personal, Research Grant: Merck, AstraZeneca, Advaxis, BMS, Amgen, Takeda, Genmab, GSK. R. Ahmed: Other, Personal, Other, patent: Anti-PD-1 monoclonal antibodies; Other, Personal and Institutional, Funding, research fundings: Merck. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

223P - Single-cell RNA-seq dissecting the stemness of tumor cells and the tumor microenvironment of liver metastasis in lung cancer (ID 313)

Presentation Number
223P
Lecture Time
12:30 - 12:30
Speakers
  • Shu-yue Zheng (Hong Kong, Hong Kong PRC)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Liver metastasis (LM) frequently occurs in patients with advanced lung cancer; yet our understanding of the underlying salient biology is preliminary. Here, we performed single-cell RNA-seq in three patients with LM of lung cancer and compared them with the single-cell RNA-seq of hepatocellular carcinoma and primary lung cancer in public database.

Methods

Single cell RNA sequencing was performed on tissues from LM of lung cancer. Also, we downloaded the single-cell RNA-seq of hepatocellular carcinoma and primary lung cancer from TCGA and GEO database. We applied Seurat to sort single-cell RNA-seq of LM in lung cancer and primary lung cancer into different clusters via feature dimension reduction and then investigated their expression profiling, stemness initiating and enrichment pathways. Furthermore, CellChat was used to compare the cellular communication and regulatory network of tumor microenvironment among LM of lung cancer, hepatocellular carcinoma, and primary lung cancer.

Results

33820, 23427, 21750 tumor cells among LM in lung cancer, hepatocellular carcinoma, and primary lung cancer were extracted respectively. There were 30 clusters divided into LM of lung cancer and primary lung cancer and cluster 0, 5, 9 had much more epithelial cells in LM of lung cancer than primary lung cancer. 152 genes were only expressed in cluster 0, 5, 9 of LM in lung cancer, which might be regarded as the initiating stem genes for LM of lung cancer. And they were enriched in response to dexamethasone and epithelial cell differentiation. 27 clusters have been divided among LM of lung cancer, hepatocellular carcinoma, and primary lung cancer. Cell communication showed that macrophage and cancer associated fibroblasts (CAFs) were the main components for tumor microenvironment, of which migration inhibitor (MIF) (CD74+CXCR4) might be the key signaling pathway for LM of lung cancer.

Conclusions

The initiating stem genes, cellular communication, and regulatory network of tumor microenvironment for LM of lung cancer have been investigated in this study, which provided a further understanding of the potential biological mechanisms of LM in lung cancer.

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 Poster Display session

224P - Immune characterization of de novo metastatic breast cancer (ID 314)

Presentation Number
224P
Lecture Time
12:30 - 12:30
Speakers
  • Mathilde Yernaux (Brussels, Belgium)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Breast cancer (BC) is mostly detected at an early stage but can be diagnosed with de novo (dn) metastases. Oligometastatic BC (OMBC) is distinguished from polymetastatic BC (PMBC) based on a limited disease burden usually defined by a cut-off of 5 metastases and is associated with a better prognosis. The immune system could be implicated in the control of the OMBC state by limiting metastatic dissemination. Here we compared tumour immune infiltrates in a cohort of dn OMBC and dn PMBC and correlated them with survival.

Methods

Clinicopathological characteristics at diagnosis of 115 dn OMBC and 117 dn PMBC patients were retrospectively collected. Tumour-infiltrating lymphocytes (TILs) were quantified on standard H&E staining. Immune infiltrates of 31 dn OMBC and 42 dn PMBC primary tumours were further characterized using multiplex multispectral immunochemistry (mIHC) allowing detection of CD4, CD8, FoxP3 T and CD20 B cells classified as intra-tumoral (i) or stromal (s) according to their localization. Epithelial cells were marked with pancytokeratin. Survival analyses were assessed using Cox’s proportional hazards models.

Results

In both cohorts, BC were mostly staged cT2 with lymph node involvement. Patients with dn OMBC had lower neutrophil/lymphocyte ratios, LDH and CA15-3 levels and showed a better outcome. No statistical differences were noted regarding TIL levels (median 5% in OMBC, 0% in PMBC) or their immune spatial distribution and most tumours were classified as “cold”. mIHC revealed that CD4 and CD8 T cells were the most represented TILs. Very few FoxP3 T and CD20 B cells were present. We noted a significantly higher infiltration of iCD4, iCD8 and iFoxP3 T cells in dn OMBC. Similar findings were found for sCD4 T cells (p<0.001). Median CD4/CD8 ratios in both compartments were significantly higher in dn OMBC (p=0.001). Of interest, each 10% increment of sCD8 T cells were associated with better OS (HR: 0.48, p=0.034) and PFS (HR: 0.57, p=0.047) only in dn OMBC. sCD4/CD8 and sCD8/FoxP3 ratios were also correlated with survival in dn OMBC. In both cohorts iCD20 B cells were associated with worse survival.

Conclusions

Here we show that CD8 T cells have prognostic value in dn OMBC but not in dn PMBC suggesting a potential role of immunotherapy in this setting, and it should be further investigated.

Legal entity responsible for the study

Institute Jules Bordet.

Funding

Association Jules Bordet.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

225P - Ovarian cancer-cell glucocorticoid receptor activity modulates cytokine secretion promoting infiltration of immunosuppressive cells into the tumor microenvironment (ID 315)

Presentation Number
225P
Lecture Time
12:30 - 12:30
Speakers
  • Manisha Taya (Dallas, TX, United States of America)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immunosuppressive myeloid-derived suppressor cells (MDSCs) have recently been identified as a heterogeneous cell population that expand during tumor-associated inflammation and are significantly increased in the tumor, peripheral blood, as well as ascites of ovarian cancer patients. MDSCs contribute to tumor immune tolerance via inhibition of T-cell proliferation and activation. Signals from the tumor microenvironment in the form of soluble cytokines lead to MDSC infiltration. It is also known that high tumor cell glucocorticoid receptor (GR) expression is associated with a relatively poor prognosis in ovarian cancer patients. We hypothesized that ovarian cancer cell GR expression and activity may modulate tumor cell cytokine secretion thereby leading to increased MDSC recruitment to the tumor, thereby creating an immunosuppressive environment.

Methods

Our preliminary data confirm that GR activation in high-grade ovarian cancer cell lines upregulates secretion of several immunosuppressive proteins including GCSF, TGFb, and CXCL5 in ovarian cancer models. We also observed downregulation of cytokines required for anti-tumor T-cell function including IFN-g. Importantly, treatment of cells with a selective GR modulator (SGRM) reverses this immunosuppressive secretome effect.

Results

Upon culturing healthy peripheral blood mononuclear cells (PBMCs) with tumor conditioned media from GR activated ovarian cancer cells, we show that secreted immunomodulatory factors have the capacity to promote differentiation of human MDSCs from PBMCs. The resulting MDSC population was characterized via expression of activation markers including Arginase-1 and NOS2. Most importantly, the suppressive function of these MDSCs is being evaluated by determining their inhibitory potential on cytotoxic T-cell proliferation and/or activation. Finally, ovarian cancer xenograft models treated with a SGRM demonstrated reduced tumor infiltrated MDSCs.

Conclusions

We, hence, propose to determine whether targeting tumor cell-intrinsic GR activation and resulting MDSC generation has potential to improve patient outcomes in this subset of ovarian cancers.

Legal entity responsible for the study

The authors.

Funding

CPRIT.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

226P - "LncRNA DILC and PD-L1 Inhibitors; opposing metastatic and anti-oxidative roles in TME of locally advanced Triple Negative Breast Cancer" (ID 316)

Presentation Number
226P
Lecture Time
12:30 - 12:30
Speakers
  • Amany Samir (New Cairo, Egypt)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Recently, lncRNA DILC has been reported as a regulatory element in carcinogenesis of various tumors. Nevertheless, its impact on cancer immunity remains limited in breast cancer. The rationale of this study was to identify and correlate the impact of DILC and the PD-L1 inhibitor; Atezolizumab on the metastatic and oxidative function of PBMCs in tumor microenvironment (TME) of TNBC patients.

Methods

32 locally advanced TNBC peripheral blood samples were withdrawn. PBMCs isolation was performed using ficoll separation. A group of PBMCs was transfected with DILC siRNAs (using Hiperfect transfection reagent) and other group was treated with 100 nm Atezolizumab. Supernatants were collected after 48 hours and the metastatic and oxiditive profiles were analyzed by ELISA (TNF-α, MMP-9, nitric oxide, ROS and superoxide dismutase levels). One-way ANOVA statistical analysis was performed for multiple group comparison and Student's unpaired T-test for two group comparison.

Results

MMP-9 was significantly downregulated, while TNF- α was remarkably upregulated, upon siDILC and ATE treatment in comparison to naïve PBMCs (P <0.0001 and P<0.0001, respectively). Additionally, NO was increased with ATE treatment (P <0.0001), however, it remained unaffected in siDILC compared to naïve PBMCs. Moreover, ROS levels were increased in siDILC nevertheless, it was decreased in ATE treated PBMCs (P <0.0001 and P<0.0001, respectively) compared to naïve PBMCs. Surprisingly, SOD levels remained unaffected with no significant change in siDILC and ATE treated PBMCs compared to naïve cells.

Conclusions

This study introduces DILC as an oncogenic lncRNA that is able to modulate immune response in TME of TNBC compared to the PD-L1 inhbitors. These data suggest DILC as a promising target that may contribute to resistance to immunotherapy in locally advanced TNBC.

Legal entity responsible for the study

German University in Cairo, Egypt.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

227P - The elusive Luminal B breast tumor and the mysterious chemokines (ID 317)

Presentation Number
227P
Lecture Time
12:30 - 12:30
Speakers
  • Rogerio A. De Araujo (Uberlandia, Brazil)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Invasive ductal breast cancer (IDC) is a heterogeneous disease. Staging and immunohistochemistry allow adequate treatment but it is not yet ideal. Women with Luminal B tumors show an erratic response to treatment. The aim of this study is to improve the prognostic stratification of Luminal B patients.

Methods

A prospective study with 234 women with IDC, grouped by TNM and immunohistochemistry in subtypes Luminal A and B, HER2 and Triple Negative, for analysis of survival and its correlations with neutrophils/lymphocytes by hemogram. An equitable selection of 1/3 of these patients, between stages and subtypes, was analyzed for correlations of serum expressions of 7 CC-chemokines, 6 CXC-chemokines, and 3 cytokines; and analysis of TCD8+ and TCD4/FOXP3+ lymphocytes in the tumor microenvironment.

Results

Overall survival was significantly dependent on staging and tumor subtypes. There was correlation of age with IL-6 (r=+0.243;p=0.037), IL-10 (r=+0.304;p=0.009) and IP10/CXCL10 (r=+0.412;p=0.011). There was a correlation between BMI and the chemokine Rantes/CCL5 (r =-0.334; p=0.009). Kaplan-Meier curves showed that Luminal B patients with neutrophil/lymphocyte ratio >2 (Log-Rank p=0.005), or lower expression of ENA78/CXCL5 (≤239.69pg/ml) (Log-Rank p=0.016) and high expression of MIP1β/CCL4 (>34.53pg/ml) (Log-Rank p=0.017) in the serum, or TCD4 lymphocytes >30% (Log-Rank p=0.027) and TCD4+TCD8 lymphocytes >75% (Log-Rank p=0.033) infiltrators in the tumor microenvironment had higher risk of metastasis/death.

Conclusions

Luminal B patients can be better stratified both by blood count and/or serum chemokine analysis and by infiltration of T lymphocytes into the tumor, opening new target-specific therapeutic approaches, in addition to chemotherapy and hormone therapy.

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 Poster Display session

228P - Modelling T Cell-Vasculature interaction in a high-throughput microfluidic platform (ID 318)

Presentation Number
228P
Lecture Time
12:30 - 12:30
Speakers
  • Luuk De Haan (Oegstgeest, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Recent clinical success of immune checkpoint inhibitors and chimeric antigen receptor T cells has highly increased the attention for the field of immunotherapy. However, identifying responders to these therapies is challenging underscoring the necessity for translational models that increase understanding of tumor-immune responses.

Methods

In the present study, a co-culture system containing immune cells and vasculature was established. Both are essential components of the tumor microenvironment and very often lacking in in vitro tumor models, highlighting the added value of our co-culture platform. We focused on optimizing endothelial and CD8+ T cell co-cultures and subsequently assessing T cell migration from the endothelial tubes via endothelial sprouts towards various chemo attractants. In order to generate stratified 3D co-cultures, the Mimetas OrganoPlate Graft containing 64 microfluidic culture units was used. The two parallel microfluidic channels were used for generating endothelial tubules, whilst angiogenic factors (S1P, VEGF, bFGF and PMA) were added to the central chamber of the culture unit resulting in a generation of a gradient and sprouting of the endothelial tubes towards the central chamber.

Results

Angiogenic endothelial tubules formed vascular beds in presence of added factors within 3-5 days. Once vascular beds were formed, activated and fluorescently labeled CD8+ T cells were loaded in the endothelial tubules and followed in culture for 48 hours. CD8+ T cell migration was observed both via the sprouts as well as by crossing the endothelial barrier, and increased in presence of gradients of CCL2, CCXl12 and CCL9.

Conclusions

Here we present a high throughput co-culture system containing angiogenic endothelial tubules and CD8+ T cells. These co-cultures are highly suitable for studying T cell migration, event which precedes the detection and recognition of antigens at the surface of antigen-presenting cells and for interactions with other cells involved in the immune response. Furthermore, we envision that this model will evolve into an immunocompetent patient-derived tumor model that can be used to study immune responses to tumors.

Legal entity responsible for the study

Mimetas.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

229P - Identify the impact of SARS-CoV-2 on Lung Cancer tumorigenesis using host-pathogen interaction network analysis (ID 319)

Presentation Number
229P
Lecture Time
12:30 - 12:30
Speakers
  • Ravindra Kumar (Kozhikode, India)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the coronavirus variant that started the global pandemic and infected more than 12 million victims around the world. SARS-CoV-2, similar to other viruses, interacts with the host proteins to reach the host cells and replicate its genome. Consequently, viral-host protein-protein interaction (PPI) identification could help predict the virus's behavior and its overlap with other pathogenic pathways. Several viruses have been known to closely trigger cancer pathways, serving as the starting point for tumorigenic mutations and even metastatic relapse. This cancer-causing nature of viruses encourages us to investigate the differentially expressed genes (DEGs) and associated enriched pathways in patients during and after SARS-CoV-2 infection.

Methods

RNA microarray data of Covid-19 infected patients, available in the GEO database through the studies done by Gordon et al. and Blanco Melo et al., were used as Covid-19 differential expression dataset. Lung cancer differential expression datasets obtained from the GEPIA database and individual networks for both diseases were created using Cytoscape, keeping the entire human gene-gene interactions as the background (HIPPIE Database).

Results

In this study, we determined 287 DEGs for SARS-CoV-2, 4242 DEGs for lung adenocarcinoma (LUAD) and 5958 for lung squamous cell carcinoma (LUSC) through network analysis. Clustering of these networks was done using the Leiden clustering algorithm, and clusters with overlapping genes in both the diseases were chosen for pathway enrichment using CytoKEGG and Gene Ontology analysis using the GOC tool (based on PANTHER). These analyses revealed 71 genes involved in 6 significant pathways involved in tumor immune evasion, cancer proliferation and immune signaling, namely the TLR Pathway, cAMP signaling pathway, the VEGF signaling pathway, the G-protein Coupled Receptor signaling pathway, the IL-6 amplification Pathway, and the MAPK Signalling Pathway.

Conclusions

The findings suggest that SARS-CoV-2 can play a significant role in cancer relapse by supporting tumorigenic conditions in the tumor microenvironment and facilitating tumor cell migration.

Legal entity responsible for the study

National Institute of Technology Calicut, Kerala, India.

Funding

Has not received any funding.

Disclosure

The author has declared no conflicts of interest.

Collapse
Poster Display Poster Display session

230P - A novel cross-platform concordance analysis using MultiOmyx and PhenoImager multiplexed immunofluorescence (mIF) (ID 320)

Presentation Number
230P
Lecture Time
12:30 - 12:30
Speakers
  • Qingyan Au (Aliso Viejo, CA, United States of America)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Novel immunotherapy has revolutionized the landscape of cancer therapy. Growing evidence has revealed the importance of tumor microenvironment (TME) and how it may impact the response to immunotherapy. Emerging data has suggested immune biomarkers based on co-expression patterns and spatial distribution will improve predictive performance to the efficacy of immunotherapy. mIF approach is well suited to such a need. The deep profiling and spatial characterization provided by a high-plex mIF assay is a powerful tool to identify predictive biomarkers. And low-plex mIF technology offers a faster turn-around time and could potentially be translated into clinical practice.

Methods

In this study, 40 CRC samples were analyzed using MultiOmyxTM and PhenoImagerTM assays. For MultiOmyx analysis, the samples were stained with a comprehensive immunoncology (IO) panel including 17 biomarkers. The expression and spatial distribution of each biomarker was studied with proprietary deep-learning based image analysis. Adjacent sections of each sample were stained by two PhenoImager panels: MOTiF kit and a 5-plex custom assay for identification of mature tertiary lymphoid structures (TLS). Biomarker classification was performed using Indica Halo analytics algorithm.

Results

PhenoImager panels successfully identified different subtypes of tumor infiltrating lymphocytes (TILs) and mature TLS within the TME. MultiOmyx analysis was able to provide a comprehensive characterization of immune markers and further classification of TLS into different maturation stages based on biomarker expression and spatial organization of immune cells. To assess cross-platform concordance, correlation coefficient was calculated using cell density data generated by each platform. Direct correlation was observed for the markers used in this study.

Conclusions

This study provides a use case on complementary mIF platforms to support translational studies at different stages. The data indicates that it can effect a practical path with use of a high-plex mIF assay for discovery of novel biomarkers and then bridge into a low-plex mIF assay to further clinical understanding and practice.

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 Poster Display session

231P - Characterization of the tumor microenvironment in a cohort of KRAS- and EGFR mutant non-small cell lung cancer (ID 321)

Presentation Number
231P
Lecture Time
12:30 - 12:30
Speakers
  • Oliver A. Kindler (Graz, Austria)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immune Checkpoint Blockade (ICB) led to better outcomes in non-small cell lung cancer (NSCLC) but only a subset of patients benefits from current treatment regimens. Different molecular subtypes show diverse treatment responses. It was reported that patients with KRAS-mutant tumors respond better to ICB therapy, in contrast, EGFR-mutant tumors show higher resistance to this type of therapy. In order to evaluate distinctions between these subtypes, a thorough characterization of the immune environment was performed in patients with untreated NSCLC.

Methods

To characterize the immune environment, flow cytometry and multiplex immunohistochemistry was used. Additionally, TCR sequencing and RNA sequencing was performed. The findings were validated in public datasets. Therapeutic blockage of CCL20 was tested in a murine in-vivo flank tumor model comparing CCL20-neutralizing antibody to its isotype control.

Results

No apparent difference of immune cell composition was found between the molecular subgroups. Tumor mutational burden (TMB) and PDL1 expression on cancer cells was higher in KRAS-mutant NSCLC. Additionally, expression of TIGIT and CCL20 were upregulated in the same subgroup. CCL20 upregulation, but not TIGIT expression could be validated in the TCGA lung adenocarcinoma cohort. Additionally, higher CCL20 expression was associated with lower survival probability. Upregulation of CCL20 in KRAS-mutated NSCLC-derived cell lines in comparison to wild type and EGFR-mutated cell lines hints at a possible role of KRAS-mediated CCL20 expression. Additionally, in-vivo experiments with a CCL20-blocking antibody showed reduced tumour growth in the treatment group.

Conclusions

Higher TMB and PD-L1 expression, both suggested biomarkers for the prognosis of ICB response, could explain better results in KRAS-mutant NSCLC. Targeting of CCL20 may be a new option for therapy in this subgroup. In-vivo experiments, using CCL20 blocking antibodies, show promising therapeutic opportunities, although current scientific literature is conflicting and a thorough investigation is warranted. In EGFR-mutated lung cancer, additional research is needed to assist therapy decisions.

Legal entity responsible for the study

J. Kargl.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

232P - The efficacy of vasohibin 2 gene expression knockdown and biological relevance in pancreatic cancer cells and tumor associated macrophages. (ID 322)

Presentation Number
232P
Lecture Time
12:30 - 12:30
Speakers
  • Malgorzata Kubczak (Lodz, Poland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Vasohibin 2 expression correlates with malignant behavior of pancreatic cancer. We aimed to study the biological effect of VASH2 gene knockdown. Due to the fact, that vasohibin 2 in engaged in tumor microenvironment remodeling, the biological effect of VASH2 gene expression knockdown was evaluated in tumor associated macrophages.

Methods

Pancreatic cancer ductal adenocarcinoma cell line (PANC-1) and THP-1 derived M2-typr tumor associated macrophages were used in the study. Uptake of siRNA with tyrosine-modified PEIs as well as lysosomal escape was confirmed by confocal microscopy analysis. Gene knockdown and biological effect was studied by qPCR. Cell survival and proliferation were tested in both cell lines with MTT and WST-8 assay respectively.

Results

Confocal microscopy analysis revealed that complexes of siRNA (siVASH2) and tyrosine-modified PEIs efficiently entered the cells. Moreover, nanoparticles escaped from lysosomes, the same preventing siRNA from early release and degradation. Cytotoxicity studies revealed that macrophages viability after 24-hours treatment with nanoparticles was about 80%. Proliferation rate of pancreatic cancer cells was reduced, depending on the type of polymer used in complex from 40 to 75% after 72-hour incubation. qPCR analysis confirmed reduced level of VASH2 in both cell types. In M2-type tumor associated macrophages decreased level of vasohibin 2 correlated with the decrease in CD206 – marker of M2-type macrophages. In pancreatic cancer cells decrease in E-cadherin and vimentin level was observed when VASH2 was targeted.

Conclusions

Our studies revealed that vasohibin 2 plays an important role in pancreatic cancer progression. It also affects tumor associated macrophages. There is no available vasohibin 2 inhibitor, thus, gene therapy seems to be a promising strategy for VASH2 inhibition. We have chosen tyrosine-modified PEIs as a siRNA carriers. Due to their low toxicity, good biocompatibility and high biological activity they should be considered as a gene carriers in tumor immunooncology gene therapies.

Legal entity responsible for the study

The authors.

Funding

National Science Centre, Poland (2020/37/N/NZ5/02359).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

233P - Tumor characteristics of mixed response upon immune checkpoint inhibition (ICI) in advanced melanoma (ID 323)

Presentation Number
233P
Lecture Time
12:30 - 12:30
Speakers
  • Judith M. Versluis (Amsterdam, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

ICI has improved outcomes in advanced melanoma with often durable response. Mixed responders are an interesting subgroup of responding patients (pts) to identify mechansims of resistance to ICI.

Methods

Stage IV melanoma pts treated with ICI achieving mixed response were retrospectively screened for available samples containing sufficient tumor at at least 2 time-points. Included pts were divided among groups based on sample characteristics: regressive and progressive lesion at same site (group A, n=1), regressive and new lesion at other site (group B, n=2), and regressive and progressive lesion at different sites (group C, n=3). Of these pts, matched CD8+ IHC stainings (4 pts) plus RNAseq (5 pts) and DNAseq (3 pts) were performed.

Results

The table gives an overview of patients and their tumor lesions. The progressive vs regressive lesion of pt 1 (group A) had more CD8+ cells/mm2 but a decrease of RNAseq T cell expression and a decreased IFNy score [Table]. Unlike group A, both pts in group B had increased IFNγ scores in their new progressive vs regressive lesion. However, pt 2 had less CD8+ cells/mm2 and stable RNAseq T cell expression, while pt 3 had more CD8+ cells/mm2 and increased RNAseq T cel expression in the progressive lesion. Pts 4 and 5 (group C) had increased IFNγ scores as well, but lower RNAseq T cell expression in the progressive vs regressive lesions. Mutational profiles, based on DNA base changes, of pt 5 and 6 (group C) differed slightly between both lesions (14% and 4%, resp.) but tumor mutional burden (TMB) was much higher in the progressive lesions (540% and 429% increase, resp.). Pt 2 (group B) had similar mutational profiles between the lesions (2% difference) but a 35% decrease in TMB.

Patient group Patient Regressive lesion CD8+ cells/mm2 IFNγ score TMB level Progressive lesion CD8+ cells/mm2 IFNγ score TMB level
A 1 Lymph node 314 0.64 - Lymph node 842 -1.30 -
B 2 Cutis 1742 0.05 742 Adrenal gland 1629 0.53 482
B 3 Liver 225 -2.16 Cutis 521 -0.23
C 4 Cutis - 0.15 717 Cutis 1094 0.88 4588
C 5 Lymph node 172 -1.15 - Gall bladder 250 1.22 -
C 6 Lymph node - - 169 Cutis - -0.39 894

Conclusions

Progressive lesions at a different site have greatly increased TMB, while the newly emerged lesion showed decrease in TMB. In both groups, mutational profile showed only a moderate change between regressive and progressive lesions.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

C.U. Blank: Financial Interests, Institutional, Advisory Board: BMS, MSD, Roche, Novartis, GSK, AZ, Pfizer, Lilly, GenMab, Pierre Fabre; Financial Interests, Personal, Expert Testimony: Third Rock Ventures; Financial Interests, Personal, Stocks/Shares: Immagene; Financial Interests, Personal, Stocks/Shares, intention to develop IFN signature algorithm: NewCo, no name yet; Financial Interests, Institutional, Invited Speaker: BMS, Novartis, NanoString, 4SC; Other, Personal, Other, pending patent: WO 2021/177822 A1. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

234P - Oncolytic adenovirus-based therapeutics to reprogram the glioblastoma microenvironment for improved CAR T cell therapy (ID 324)

Presentation Number
234P
Lecture Time
12:30 - 12:30
Speakers
  • Jana De Sostoa PomŽs (Geneva, Switzerland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Glioblastoma (GBM) is a highly aggressive and heterogeneous tumor, with a strong immunosuppressive tumor microenvironment (TME), in which tumor-associated macrophages (TAM) play a crucial role. This immunosuppressive phenotype is detrimental to the efficacy of chimeric antigen receptor (CAR) T cell therapy. We believe that oncolytic adenoviruses can overcome some of the mechanisms involved in TME immunosuppression. Here, we investigate the capacity of the oncolytic adenovirus ICO15K to reprogram the GBM TME, with a special focus on promoting the transition from a pro-tumoral M2 to a pro-inflammatory M1 TAM phenotype.

Methods

ICO15K was previously engineered to specifically infect and replicate in cancer cells. We used patient-derived GBM and mouse GBM cell lines and assessed their susceptibility to ICO15K in vitro. To study ICO15K-induced pro-inflammatory TAM phenotype both in human and mouse models in vitro, monocytes-derived macrophages or bone marrow-derived macrophages, respectively, were treated, and their gene profile was characterized by qRT-PCR. In vivo, immunocompetent mice bearing the SB28 mouse GBM cell line or the SB28-hCAR line modified to express the human coxsackie and adenovirus receptor (hCAR) for adenovirus entry were treated with PBS or ICO15K and the immune landscape of tumors was characterized.

Results

We show that ICO15K is able to infect and exert a significant cytotoxicity effect in a panel of patient-derived cell lines, as well as in mouse SB28 and GL261 cell lines. In addition, both human and mouse M2 macrophages showed an ICO15K-induced transition towards an M1-like pro-inflammatory phenotype in vitro. In vivo, ICO15K-treated mice showed significant increased T cell infiltration and increased pro-inflammatory TME phenotype compared to non-treated mice.

Conclusions

Altogether, we present preliminary data of ICO15K-mediated TME modulation that could help to improve CAR T cell therapy. Further preclinical development of this strategy is needed to better understand its therapeutic impact for the improvement of GBM patient outcomes.

Legal entity responsible for the study

The authors.

Funding

ISREC.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

235P - Multiplexed Imaging Reveals the Immunomodulatory cGAS as a Predictor of Response in Patients With Malignant Pleural Mesothelioma (ID 325)

Presentation Number
235P
Lecture Time
12:30 - 12:30
Speakers
  • Alexander Laure (Zurich, Switzerland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The cGAS/STING pathway is an important part of the DNA sensing machinery and plays an important role in fundamental processes of the anti-tumor immune response. Recently it has been hypothesized that cGAS expression might as well have a pro-tumorigenic role through chronic inflammation. Exposure to asbestos fibers can lead to a long-lasting inflammation of the pleura, which is promoting development of malignant pleural mesothelioma (MPM). The aim of this work was to investigate the expression of cGAS and STING as predictive or prognostic factors for patients with MPM.

Methods

We analysed tissue micro arrays (TMAs) with tumor tissue from 190 MPM patients by multiparameter immunofluorescence and multispectral microscopy. We evaluated the expression of cGAS and STING in tumor cells, non-tumor cells and CD8 T cells in chemotherapy naïve and matching treated MPM tissue samples. The different cell phenotypes were assessed for cGAS and STING expression on a single cell level and results were correlated to response to treatment and progression free survival.

Results

A low count of cGAS+ cells before the start of chemotherapy is a favorable prognostic marker for progression free survival. The majority of cGAS+ cells are non-tumor cells followed by tumor cells. After chemotherapy, all cell subtypes showed a significant increase of cGAS+ (p<0.0001) but not STING. In patients who achieved a partial response after chemotherapy, the increases of cGAS+ cells was significantly lower compared to patients with progression of disease (11.8 fold vs. 25.86 fold increase, p=0.022).

Conclusions

Here we show that low cGAS+ expression at baseline is favorable prognostic marker and that the increase of cGAS expression is a negative predictive marker in MPM patients. The possible pro-tumorigenic role for this disease warrants further investivation.

Legal entity responsible for the study

The authors.

Funding

Stiftung für angewandte Krebsforschung Zürich Krebsliga des Kantons Zürich.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

236P - A novel microfluidic platform for understanding the role of PDAC stroma in immune response (ID 326)

Presentation Number
236P
Lecture Time
12:30 - 12:30
Speakers
  • Marlene Geyer (Oegstgeest, Netherlands)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Pancreatic Ductal Adenocarcinoma (PDAC), the most common pancreatic cancer type is estimated to become the second leading cause of cancer-related deaths by 2030 with mortality rates of up to 93%. PDAC is characterized by high-density stroma and an immunosuppressive tumor microenvironment that lead to a lack of efficacy of immunotherapeutic strategies. Those facts highlight the urgent need for more relevant PDAC in vitro models that support the discovery of novel (immuno-)therapies. Here, we describe the establishment of a multiple cell type culture in a microfluidic platform that will be applied for modeling immune and stroma cell interplay in PDAC.

Methods

Microfluidic platforms such as the OrganoPlate® support 3D growth and inclusion of different cell types cultured under flow conditions. The aim of this project was to determine the involvement of stromal cells such as endothelial cells and pancreatic stellate cells in immune cell recruitment in pancreatic cancer. In order to generate a more relevant culture set up, supporting cells and PDAC organoids were used in the 3-lane OrganoPlate® to study immune cell recruitment. Migration of fluorescently labelled peripheral blood mononuclear cells from vasculature through the stromal compartment to the PDAC organoids was tracked for 72h using confocal microscopy.

Results

Migration of immune cells to the tumor cell compartment was significantly inhibited by the stromal compartment, thus confirming in vivo immune responses, as the stroma seems to form both a physical as well as a chemical barrier for immune cells to reach the active tumor site. Interestingly, PDAC organoid conditioned medium efficiently attracts immune cells into the tumor compartment, indicating that secreted factors play a relevant role in immune cell recruitment. Despite the positive effect on recruitment and immune-tumor cell interaction, a lack of tumor cell killing is observed.

Conclusions

The co-culture of PDAC organoids with stromal and immune cells grown on-a-Chip as described in this study indicates the suitability of microfluidic platforms for generating complex models and its use for dissecting complex cellular interplay and processes involved in the lack of anti-tumor immune responses in PDAC.

Legal entity responsible for the study

Mimetas.

Funding

European Union.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

237P - Immune checkpoint inhibitor therapy and outcomes from SARS-CoV-2 infection in patients with cancer: a joint analysis of OnCovid and ESMO-CoCARE registries (ID 327)

Presentation Number
237P
Lecture Time
12:30 - 12:30
Speakers
  • Alessio Cortellini (London, United Kingdom)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

As management and prevention strategies against Coronavirus Disease-19 (COVID-19) evolve, it is still uncertain whether prior exposure to immune checkpoint inhibitors (ICIs) affects COVID-19 severity in patients (pts) with cancer.

Methods

In a joint analysis of ICI recipients from OnCovid (NCT04393974) and ESMO CoCARE registries, we assessed severity and mortality from SARS-CoV-2 in vaccinated and unvaccinated pts with cancer and explored whether prior immune-related adverse events (irAEs) influenced outcome from COVID-19.

Results

The study population consisted of 240 pts diagnosed with COVID-19 between Jan 2020 and Feb 2022 exposed to ICI within 3 months prior to COVID-19 diagnosis, with a 30-day case fatality rate (CFR30) of 23.6% (95%CI: 17.8-30.7%). 42 (17.5%) were fully vaccinated prior to COVID-19 and experienced decreased CFR30 (4.8% vs 28.1%, p=0.001), hospitalization rate (27.5% vs 63.2%, p<0.001), requirement of oxygen therapy (15.8% vs 41.5%, p=0.003), COVID-19 complication rate (11.9% vs 34.6%, p=0.004), and COVID-19-specific therapy (26.3% vs 57.9%, p=0.001) compared with unvaccinated pts. IPTW-fitted multivariable analysis, following a clustered-robust correction for the data source (OnCovid vs ESMO CoCARE), confirmed that vaccinated pts experienced a decreased risk of death at 30 days (aOR 0.08, 95%CI: 0.01-0.69). 38 pts (15.8%) experienced at least 1 irAE of any grade at any time prior to COVID-19, at a median time of 3.2 months (0.13-48.7) from COVID-19 diagnosis. IrAEs occurred independently of baseline characteristics except for primary tumour (p=0.037) and were associated with a significantly decreased CFR30 (10.8% vs 26.0%, p=0.0462) additionally confirmed by the IPTW-fitted multivariable analysis (aOR: 0.47, 95%CI: 0.33-0.67). Pts who experienced irAEs also presented a higher median absolute lymphocyte count at COVID-19 (1.4 vs 0.8 109 cells/L, p=0.009).

Conclusions

Anti-SARS-CoV-2 vaccination reduces morbidity and mortality from COVID-19 in ICI recipients. History of irAEs might identify pts with pre-existing protection from COVID-19, warranting further investigation of adaptive immune determinants of protection from SARS-CoV-2.

Clinical trial identification

NCT04393974 OnCovid.

Legal entity responsible for the study

Imperial College London & ESMO.

Funding

Imperial Biomedical Research Centre ESMO.

Disclosure

A. Cortellini: Financial Interests, Personal, Advisory Board: MSD, OncoC4; Financial Interests, Personal, Invited Speaker: Eisai, AstraZeneca; Financial Interests, Personal, Expert Testimony: Iqvia. D.J. Pinato: Financial Interests, Personal, Invited Speaker: ViiV Healthcare, Bayer, BMS, Roche, Eisai, Falk Foundation; Financial Interests, Personal, Advisory Board: Mina Therapuetics, Eisai, Roche, DaVolterra, AstraZeneca. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

238P - Exploring blood immune cell dynamics to unravel the immunomodulatory effect of radiotherapy in NSCLC patients undergoing immune checkpoint inhibitors (ID 328)

Presentation Number
238P
Lecture Time
12:30 - 12:30
Speakers
  • Giulia Mazzaschi (Parma, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

The role of radiotherapy (RT) in immunotherapy-based (IO) combinatory approaches to advanced NSCLC is still uncertain due to its dual immune -suppressive and -stimulatory effect. We performed a longitudinal peripheral blood (PB) analysis to determine whether RT, by affecting immune cell phenotypes and dynamics, impacts on clinical outcome of IO-treated NSCLC patients.

Methods

PB samples were prospectively collected at baseline (T0) and first disease assessment (T1) on stage IV NSCLC undergoing 1st line IO-based regimens alone (RTnull) or combined with RT (RTpre, within 4 weeks before IO; RTpost, during IO). Flow cytometric analysis included CD3, CD8, CD4, NK, NKT, CD19, CD14 and Treg cells, expression of functional molecules (PD1, Granzyme B [GZB], Perforin [Perf]) and proliferative index (Ki67). PB parameters and their delta variation ([T1-T0/T0] * 100) were correlated with RT administration, Objective Response Rate (ORR) and Progression-free survival (PFS).

Results

Among 57 patients, 22 underwent RT either before (RTpre, 32%) or during (RTpost, 68%) IO. RT doses ranged from 8 to 54 Gy according to sites of involvement. No significant differences in IO response and survival emerged between RT and RTnull cases. Compared to RTnull, baseline RTpre immune profiles exhibited increased % of CD8, CD19, CD14 and NK cells expressing PD1, reduced CD4+GZB/Perf+ and Tregs. Delta variation revealed that RTpre attenuated the downregulation of PD1 in CD8, CD4 and CD19 cells following IO, and favored the circulating release of GZB/Perf+ CD8 and CD4. RTpost reduced CD8 number, proliferation and PD1 expression, while increasing NKT. At variance from RTpre, RTpost did not affect PD1+ T cell kinetic, although decreased total and PD1+ NKs. We observed a clear trend towards prolonged PFS in RTpre group (median PFS: RTpre= not reached, RTpost= 7.1 mos, RTnull= 5.9 mos) associated with slightly increased ORR, hinting that the positive cytotoxic (CD8+GZB/Perf+, NK) to suppressive (Tregs) balance triggered by RTpre may result in greater benefit from IO.

Conclusions

RT timing may differentially impact on clinical outcome of IO-treated NSCLC patients by shaping immune cells phenotypes and dynamics.

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 Poster Display session

239P - Survival outcomes predicted by irAEs on 18F-FDG-PET in response to PD-1 antibody therapy in metastatic melanoma (ID 329)

Presentation Number
239P
Lecture Time
12:30 - 12:30
Speakers
  • Sarah Lewis (Melbourne, Australia)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

PD-1 antibody therapy has revolutionised the landscape of metastatic cutaneous melanoma treatments and outcomes. The aim of our study is to investigate whether immune related adverse events (irAEs) or granulomatous/reactive nodal changes visible on 18F-FDG-PET during treatment with PD-1 antibody therapy for metastatic cutaneous melanoma predicts improved overall survival.

Methods

Patient demographics, treatment regimes, toxicity profiles and 18F-FDG-PET scans were collected for patients who underwent treatment at Alfred Health in Melbourne, Victoria between 2015 and 2019 for advanced melanoma. Data were extracted from each patient’s electronic medical record. Patients were included if they were treated with 1st line PD-1 antibody +/- CTLA-4 antibody therapy for unresectable stage III or stage IV metastatic cutaneous melanoma. Patients were excluded if an 18F-FDG- PET was not performed both during and prior to commencement of immunotherapy. Two blinded-nuclear medicine physicians reviewed 18F-FDG-PET imaging at baseline and following immunotherapy. The review criteria included granulomatous/reactive changes and PET detected irAEs. Clinically reported irAE were also collected. Statistical analysis was performed using IBM SPSS software.

Results

A total of 103 patients (68% male) met the inclusion criteria for the study. The largest proportion of individuals had M1c disease (26.2%) followed by unresectable stage III (25.2%), M1b (21.4%), M1a (16.5%) and M1d (10.7%) respectively. Most individuals received single agent anti-PD-1 (71.6%) whereas a smaller proportion received combination therapy with CTLA4 antibody therapy (28.4%). Patients with irAEs visible on 18F-FDG-PET during treatment had improved survival (Figure 1) (p=0.014). Granulomatous or reactive nodal changes visible on 18F-FDG-PET did not have any significant impact on survival outcomes (p=0.80) nor did clinically reported irAEs (p=0.60).

Conclusions

This study demonstrates that irAEs visible on 18F-FDG-PET during PD-1 antibody therapy for metastatic melanoma predicts improved overall survival.

Legal entity responsible for the study

Sarah Lewis.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

240P - AMU: Using mRNA Embedding in Transformer Network to Predict Melanoma Immune Checkpoint Inhibitor Response (ID 330)

Presentation Number
240P
Lecture Time
12:30 - 12:30
Speakers
  • Yi Yin (Fuzhou, China)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Melanoma immune checkpoint inhibitor (ICI) response is related to complex biological pathways. A deep learning (DL) model has powerful fitting ability and transformer structure has interactive information extraction function. We designed a transformer neural network fed with multi-gene mRNA expression quantities to predict melanoma ICI response and explored gene interactions from the embedding layer.

Methods

We built our training/validation/testing datasets from four open databases, 208 pre-ICI and 58 post-ICI samples were included in training/validation dataset and testing dataset, respectively. Every sample has 160-dimension mRNA expression quantities sequenced from tumor tissues. The 160 genes were previously described as potentially associated with melanoma, inflammation, immunity, and the PD-L1/CTLA4 pathways. We designed a DL model named AMU with the transformer encoder followed by a convolutional neural network (CNN). We applied the method of upsampling for data enhancement and set the 20-dimension gene embeddings. SVM, Random Forest, AdaBoost, XGBoost and a simple CNN we designed were token as competing models. In model interpretation work, gene feature vectors were extracted from the embedding layer and gene interactions were calculated by t-distributed Stochastic Neighbor Embedding (t-SNE) algorithm, then the gene interactions were compared with that inquired in the Functional Protein Association Network (STRING, https://cn.string-db.org/).

Results

AMU showed the preferred performance with the area under the curve (AUC) of 0.941 and the mean average precision (mAP) of 0.960 in validation dataset and AUC of 0.672, mAP of 0.800 in testing dataset, respectively. In model interpretation, the TNF-TNFRSF1A pathway was indicated as a key pathway to influence melanoma ICI response. Further, gene features learned from AMU showed a local similarity with STRING.

Conclusions

The DL model built with transformer encoder structure has strong power to process mRNA expression data and precisely predicted the melanoma ICI response. Meanwhile, gene vector representation learned from trainable embedding layer is promising for DL application in the biomedical field.

Legal entity responsible for the study

Y. Yin.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

241P - Treatment and outcomes of patients with gastrointestinal toxicity following immunotherapy- a large multi-center retrospective study in the United Kingdom by the National Oncology Trainees Collaborative for Healthcare Research (NOTCH) (ID 331)

Presentation Number
241P
Lecture Time
12:30 - 12:30
Speakers
  • Mirashini Swaminathan (Liverpool, United Kingdom)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Immune checkpoint inhibitors (ICIs) have revolutionised the treatment of many cancers, but their use has been associated with the development of gastrointestinal (GI) toxicities such as colitis and hepatitis.

Methods

A multi-center retrospective study across 12 National Health Service centers across the United Kingdom (UK) was conducted by the UK National Oncology Trainees Collaborative for Healthcare Research (NOTCH) over a 2-year period. The study included patients receiving ICIs for malignant melanoma, non-small lung cancer and renal cell cancer as standard of care. Occurrence of clinically significant (≥grade 2) GI toxicity was assessed and correlated with subsequent treatment and outcomes. Multiple logistic regression was used to assess correlation. For overall survival (OS), Kaplan-Meier and log-rank tests were utilised.

Results

The cohort included 2049 patients. 1230 (60%) were male with a median age of 66. Colitis occurred in 182 (8.9%) patients and hepatitis in 129 (6.3%). Of the patients where treatment was recorded, 129 (70.9%) received treatment with systemic steroids alone and 37 (20.3%) required second-line immunosuppressants (IS) in the colitis group. In the hepatitis group, 101 (78.3%) had steroids alone with 19 (14.7%) having IS. Improved OS was found in patients who experienced colitis (HR 2.59 95%CI: 2.15 to 3.11, p<0.0001) and hepatitis (HR 2.26, 95%CI: 1.84 to 2.79, p=<0.0001) compared to those with no adverse events. Pre-existing autoimmune disease (p=0.02) and combination ICIs (p=0.006) were predictors of colitis that required IS whilst grade 2 and 3 hepatitis (p<0.001) were predictors of hepatitis needing IS. The use of IS did not affect OS significantly in the colitis group (p=0.372) but did correlate with survival in the hepatitis group (p=0.037). Patients that were able to continue treatment with ICIs after toxicity had an increased OS in both groups (p<0.001).

Conclusions

Patients with GI toxicity following treatment with ICIs have improved OS. The use of IS did not significantly affect OS which suggests they should continue to be utilised in the treatment of GI toxicity.

Legal entity responsible for the study

United Kingdom National Oncology Trainees Collaborative for Healthcare Research (NOTCH).

Funding

Has not received any funding.

Disclosure

M. Swaminathan: Financial Interests, Institutional, Funding: Eli Lilly, Roche, UCB, Novartis, Medical Research Council. M. Baxter: Financial Interests, Personal, Other, Honoraria, travel, accommodation and expenses: Ipsen. D.J. Hughes: Financial Interests, Personal, Other, Honoraria: Novartis, Pfizer; Financial Interests, Institutional, Funding: NanoMab. R. Lee: Financial Interests, Personal, Invited Speaker: AstraZeneca; Financial Interests, Personal, Advisory Board: Pierre Fabre; Financial Interests, Institutional, Research Grant: Bristol Myers Squibb, AstraZeneca; Financial Interests, Institutional, Research Grant, Funding for translational research: Pierre Fabre; Financial Interests, Institutional, Other, Advisory role - payment going to research account in melanoma team: Lumisphere Technology. M. Pritchard: Financial Interests, Personal, Other, Consultancy funding: Ipsen, Mayoly Spindler Laboratories; Financial Interests, Personal, Other, Consultancy Funding: Advance Accelerator Application; Financial Interests, Institutional, Funding: Trio Medicines, Ltd. M.P. Rowe: Financial Interests, Personal, Other, Honoraria: MSD; Financial Interests, Personal, Speaker’s Bureau: Servier; Financial Interests, Personal, Other, Travel, accommodation, expenses: Astellas Pharma. A.C. Olsson-Brown: Financial Interests, Personal, Other, Honoraria: Bristol Myers Squibb; Financial Interests, Personal, Speaker’s Bureau: Bristol Myers Squibb; Financial Interests, Personal, Advisory Role: Bristol Myers Squibb, MSC Oncology; Financial Interests, Personal, Other, Honoraria, travel, accommodation and expenses: Roche; Financial Interests, Institutional, Funding: Roche, Lilly, UCB; Financial Interests, Personal, Funding: Novartis; Financial Interests, Personal, Other, Travel, accommodation and expenses: Servier. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

242P - Educational needs of healthcare professionals using immune checkpoint inhibitors across multiple tumour types: A cross-speciality survey (ID 332)

Presentation Number
242P
Lecture Time
12:30 - 12:30
Speakers
  • Judy Brownsword (Stockport, United Kingdom)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Several immune checkpoint inhibitors (ICIs) are available or in development for the treatment of many different cancers. The complexity of the treatment landscape suggests that healthcare professionals (HCPs) will need education to help them optimise the use of these agents in clinical practice. Our aim was to understand from HCPs which educational topics they believe are most important in this area.

Methods

We used a two-part modified Delphi process, in which HCPs from 10 different specialities in France, Germany, Italy, Spain, UK and USA were invited from a pre-existing database to complete a web-based questionnaire. In part 1, HCPs were asked to state in their own words what they believed to be the most important educational topics of benefit to them or their colleagues to improve the safe and effective use of ICIs in patients with cancer. In part 2, a second group of HCPs was asked to assess the importance of each of the topics from a list based on the responses in part 1.

Results

In part 1, 201 HCPs responded to the survey. The mean number of tumour types managed with ICIs by each participant was 4.2, with 33% managing at least 6 different tumour types. For oncologists only, the mean number of tumour types was 4.8, with 42% managing at least 6 different tumour types. On average, 45% of HCPs said that they would like to know more about the use of immune checkpoint inhibitors. This was highest for paediatric cancers (64%) and lowest for urological cancers (39%). The demographics of the 200 HCPs participating in part 2 were similar to part 1. The most frequently cited topic for education in part 1 was adverse event monitoring and management, with 87% of respondents stating this as an educational need. In part 2, 98% of respondents rated education on adverse event monitoring and management as moderately, very or extremely important. Other frequently suggested topics were indications/comparison of different ICIs (43%), the use of combination therapy (27%), and biomarkers (24%).

Conclusions

Our results give a clear indication of which topics are of most importance to HCPs for further education on the use of ICIs in cancer treatment. To our knowledge, this is the only survey to investigate educational needs in this area.

Legal entity responsible for the study

The authors.

Funding

Touch Independent Medical Education Ltd.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

243P - Skin biopsies for pharmacodynamic studies of tebentafusp, a novel bispecific molecule against the melanocytic antigen gp100. (ID 333)

Presentation Number
243P
Lecture Time
12:30 - 12:30
Speakers
  • Ramon Stäger (Zurich, Switzerland)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Tebentafusp (tebe) is a novel bispecific fusion protein redirecting T cells against gp100 (a melanocytic antigen) expressing cells, approved for metastatic uveal melanoma. Skin adverse events (incl. maculopapular rash, depigmentation) are frequent and were shown to be an off-tumor/on-target effect against gp100+ epidermal melanocytes. Thus, analysis of tebe-induced cellular and molecular processes in patient skin samples may improve mechanistic understanding.

Methods

Skin biopsies from patients treated with tebe were collected at baseline and development of skin rash (n=11), as well as from depigmented areas (n=5). Spatial analysis was performed by histology and multiplex immunohistochemistry (7-plex, Akoya Opal). On paired samples (n=6, baseline and rash) from 3 patients, single cell RNA sequencing was conducted. Furthermore, proliferation of T cell subsets (naïve and memory of each CD4+ and CD8+) treated with tebe or anti-CD3 antibody was assessed in vitro (3 healthy donors).

Results

Rash showed increased numbers of CD4+ and CD8+ T cells and markers of activation/proliferation (IFNG, GZMB, IL2RA, MKI67) were upregulated in T cells. In vitro, tebe’s capacity to induce T cell proliferation was validated, predominantly in the CD8+ memory subset, in contrast to the unselective effect observed by high affinity anti-CD3. Recruitment of monocyte-derived macrophages and plasmacytoid dendritic cells (DC) was found. Interestingly, increased activity in regulatory T cells and immunoregulatory DC suggested early homeostatic regulation. Melanocyte numbers remained unchanged in rash but were reduced in 4/5 depigmented lesions. Upregulation of interferon type I/II responses and downregulation of pigment synthesis was seen, resulting in an increased fraction of gp100-negative melanocytes.

Conclusions

Longitudinal skin biopsies are a promising and minimally-invasive tool to study the pharmacodynamics of anti-melanocytic bispecifics. The skin microenvironment on tebe treatment is characterized by T cell proliferation and cytotoxic activity, recruitment of myeloid cells as well as downregulation of the target gp100 in melanocytes and their partial loss in depigmented skin areas.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

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, touchIME, T3 Pharma, Pfizer. All other authors have declared no conflicts of interest.

Collapse
Poster Display Poster Display session

244P - DNA-PK inhibition sustains immune activation in SCLC (ID 334)

Presentation Number
244P
Lecture Time
12:30 - 12:30
Speakers
  • Carminia Maria Della Corte (Napoli, Italy)
Session Name
Poster Display
Room
Foyer mezzanine
Date
Thu, Dec 8, 2022
Time
12:30 - 13:15

Abstract

Background

Recently, 4 biological subtypes have been defined with SCLC-Inflamed (I) experiencing benefit from immunotherapy. Previously, we showed that chemotherapy and PARP/CHK1-i synergize with immunotherapy and activate STimulator of INterferon Genes (STING) pathway. We hypothesize that DDRi may sustain intratumoral shifts toward SCLC-I.

Methods

We tested anti-proliferative effect and changes in immune related protein (WB, ELISA) production after treatment of selected SCLC cell lines (n=10) with various DDRi (ATMi, ATRi, DNA-PKi) +/- radiotherapy (RT) +/- ENPP1-inhibitor (ENPP1i, that inhibits the phosphodiesterase that degrades the STING ligand, 2’3’-cGAMP). We also tested the drug effects on PBMCs. Finally, we investigated expression of innate immune pathways in SCLC cohort.

Results

From George et al. cohort, STING gene levels were correlated with higher expression of other innate immune pathway, MAVS (like IFITH1 and DDX60) and IFI16 (p<0.001). Thus, we studied changes on innate immune pathways activation (STING, MAVS, IFI16), inflammatory cytokine production and anti-proliferative effects induced by DDRi +/- RT +/- ENPP1i. Among all drug tested, we found that DNA-PKi sustain STING, MAVS and IFI16 protein activation in SCLC cells. We also detected STING activation by ELISA for 2’3’-cGAMP and by ENPP1-i. The addition of ENPP1i and RT in vitro is able to further enhance all innate immune pathways activation and is accompanied by significant increase in pro-inflammatory cytokine productions by both cancer and PBMCs cells (e.g. INFbeta, IL-6, IL-1β, TNF-alfa, CCL2) and STING-related chemokines (CCL5 and CXCL10) along with PD-L1. Also, STING activation was confirmed by increased levels of extracellular and intracellular 2’3’-cGAMP following DNA-PKi +/- RT +/- ENPP1i (at various time points up to 72 hours). Finally, we found that TP53 is activated at early time-points of treatment in response to DNA-PKi and is then downregulated by combination with ENPP1i in SCLC cell lines, thus suggesting a potential novel mechanism of interaction between these stress-related pathways.

Conclusions

Our findings highlight for the first time a positive pro-immune effect of DNA-PKi in SCLC models and on immune cells, suggesting various innate immune pathways as mediator of immune responsive.

Legal entity responsible for the study

The authors.

Funding

AIRC.

Disclosure

All authors have declared no conflicts of interest.

Collapse