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

Displaying One Session

Foyer ABC Thu, 08.12.2022
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Location
Foyer ABC
Poster Display (ID 17) Poster Display

Biomarker development (ID 801)

Lecture Time
12:30 - 12:30
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Poster Display (ID 17) Poster Display

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 643)

Presentation Number
4P
Lecture Time
12:30 - 12:30
Speakers
  • F. Dall'Olio (Bologna, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • F. Dall'Olio (Bologna, Italy)
  • C. Garcia (Villejuif, France)
  • W. Zrafi (Villejuif, France)
  • E. Fortunati (Bologna, Italy)
  • A. Bettayeb (Villejuif Cedex, France)
  • G. Zalcman (Paris, CEDEX 18, France)
  • J. Remon Masip (Barcelona, Spain)
  • L. Hendriks (Maastricht, Netherlands)
  • M. Tagliamento (Genova, Italy)
  • G. Bonardel (Saint-Denis, France)
  • C. Helissey (Saint-Mandé, France)
  • V. Roelants (Woluwe-Saint-Lambert, Belgium)
  • A. Fourquet (Paris, France)
  • F. Aboubakar (Brussels, Belgium)
  • N. Chaput-Gras (Villejuif, Cedex, CEDEX, France)
  • F. Passiglia (Orbassano, Italy)
  • I. Monnet (Creteil, France)
  • D. Planchard (Villejuif, Cedex, France)
  • F. Barlesi (Villejuif, CEDEX 20, France)
  • B. Besse (Villejuif, CEDEX, France)

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

Collapse
Poster Display (ID 17) Poster Display

5P - Tumor microenvironment cellular crosstalk predicts response to adoptive TIL therapy in melanoma patients (ID 497)

Presentation Number
5P
Lecture Time
12:30 - 12:30
Speakers
  • D. Dangaj (Lausanne, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • D. Barras (Lausanne, Switzerland)
  • E. Ghisoni (Lausanne, Switzerland)
  • J. Chiffelle (Lausanne, Switzerland)
  • A. Orcurto (Lausanne, Switzerland)
  • J. Dagher (Lausanne, Switzerland)
  • N. Fahr (Lausanne, Switzerland)
  • U. Dafni (Athens, Attica, Greece)
  • C. Sempoux (Lausanne, Switzerland)
  • O. Michielin (Lausanne, Switzerland)
  • L. Trueb (Lausanne, Switzerland)
  • A. Harari (Lausanne, Switzerland)
  • G. Coukos (Epalinges, Switzerland)

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

Collapse
Poster Display (ID 17) Poster Display

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 372)

Presentation Number
6P
Lecture Time
12:30 - 12:30
Speakers
  • I. Reijers (Amsterdam, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • P. DIMITRIADIS (Amsterdam, Netherlands)
  • J. Traets (Amsterdam, Netherlands)
  • R. Saw (Camperdown, Australia)
  • J. Versluis (Amsterdam, Netherlands)
  • T. Pennington (Wollstonecraft, Australia)
  • E. Kapiteijn (Leiden, Netherlands)
  • A. Van der Veldt (Rotterdam, Netherlands)
  • K. Suijkerbuijk (Utrecht, Netherlands)
  • G. Hospers (Groningen, Netherlands)
  • W. Van Houdt (Amsterdam, Netherlands)
  • A. Broeks (Amsterdam, Netherlands)
  • S. Cornelissen (Amsterdam, Netherlands)
  • A. Spillane (Sydney, Australia)
  • R. Scolyer (Camperdown, Australia)
  • B. Van de Wiel (Amsterdam, Netherlands)
  • A. Menzies (Wollstonecraft, Australia)
  • A. Van Akkooi (Wollstonecraft, Australia)
  • G. Long (Wollstonecraft, NSW, Australia)
  • C. Blank (Amsterdam, Netherlands)

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-y signature and TMB.

Methods

TMB and the IFN-y 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

Table 1 shows the association between IFN-y GES and TMB with pRR, MPR, and EFS. IFN-y GES and logTMB were not correlated (R = 0.065; p = 0.579). Pts with a high IFN-y GES had a higher pRR and MPR rate than pts with a low IFN-y 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-y H/TMB H was 90%, IFN-y H/TMB L 79%, IFN-y L/TMB H 67% and IFN-y L/TMB L 42%. For MPR rates this was 85%, 63%, 67% and 19%, respectively.

Table 1

Univariable analysis

OR / HR

95% CI

p-value

pRR

IFN-y score (continuous)

1.089

(1.027-1.156)

0.004

IFN-y 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-y score (continuous)

1.092

(1.031-1.157)

0.003

IFN-y 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-y score (continuous)

0.965

(0.922-1.011)

0.130

IFN-y 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-y GES and TMB were independent biomarkers for pathologic response and MPR, and the IFN-y 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

Collapse
Poster Display (ID 17) Poster Display

7P - PrimeCUTR: Identifying clinically relevant neoantigens in the untranslated regions of cancer genomes. (ID 683)

Presentation Number
7P
Lecture Time
12:30 - 12:30
Speakers
  • C. Sng (London, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Sng (London, United Kingdom)
  • B. Simpson (London, United Kingdom)
  • J. Van De Haar (Amsterdam, Netherlands)
  • H. Cha (Seoul, Korea, Republic of)
  • A. Coulton (London, United Kingdom)
  • K. Thakkar (London, United Kingdom)
  • K. Litchfield (London, United Kingdom)

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 included 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. Wilcoxon signed-rank test and survival analysis were used to assess associations with CPI treatment response and overall survival (OS).

Results

Within HMF, 75.5% of patients had one or more start-gain neoORFs while 30.5% had at least one stop-loss neoORF. Among those with start-gain neoORFs, 86.9% had neoORFs spanning 20 amino acids or more. These long neoORFs were most frequent in the grouping of lung and head and neck cancer. CPI treatment response was associated with a longer total length of start-gain (but not frameshift or stop-loss) neoORFs (P = 0.0047). Importantly, this association was also demonstrated in the subset of tumours with a low tumour mutational burden (TMB) (P = 0.014). Among GEL patients, survival benefit was specifically seen in patients with melanoma (n = 152), where those whose tumours had longer start-gain neoORFs (>29 amino acids, n = 63) had greater OS (HR 0.59, 95% CI 0.37 - 0.96, 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.

Collapse
Poster Display (ID 17) Poster Display

8P - Phenotypic characterization of infused tumor-infiltrating lymphocytes (TIL) correlates with response to adoptive cellular therapy (ACT) in patients with metastatic melanoma (MM) (ID 387)

Presentation Number
8P
Lecture Time
12:30 - 12:30
Speakers
  • J. Stoltenborg Granhøj (Herlev, Denmark)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Stoltenborg Granhøj (Herlev, Denmark)
  • M. Rohaan (Amsterdam, Netherlands)
  • T. Holz Borch (, Denmark)
  • M. Presti (Herlev, Denmark)
  • C. Nijenhuis (Amsterdam, Netherlands)
  • M. Van Zon (Amsterdam, Netherlands)
  • I. Jedema (Amsterdam, Netherlands)
  • B. Nuijen (Amsterdam, Netherlands)
  • R. Kessels (Amsterdam, Netherlands)
  • A. Torres Acosta (Amsterdam, Netherlands)
  • F. Lalezari (Amsterdam, Netherlands)
  • J. Van den Berg (Amsterdam, Netherlands)
  • M. Donia (Herlev, Denmark)
  • J. Haanen (Amsterdam, Netherlands)
  • I. Svane (Herlev, Denmark)

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 1) 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.

Table 1: 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.

Collapse
Poster Display (ID 17) Poster Display

9P - Early circulating tumor DNA (ctDNA) kinetics and gene expression analysis to predict treatment outcomes with anti-PD-1 therapy (ID 519)

Presentation Number
9P
Lecture Time
12:30 - 12:30
Speakers
  • E. Sanz Garcia (Toronto, Canada)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • E. Sanz Garcia (Toronto, Canada)
  • G. Laliotis (San Carlos, United States of America)
  • L. Avery (Toronto, Canada)
  • A. Spreafico (Toronto, CO, Canada)
  • A. Hansen (Woolloongabba, Ontario, Australia)
  • L. Eng (Toronto, Ontario, Canada)
  • N. Singaravalan (Toronto, Canada)
  • S. Willingham (San Carlos, United States of America)
  • M. Liu (San Carlos, United States of America)
  • S. Soleimani (Toronto, Canada)
  • T. Pugh (Toronto, Canada)
  • S. Bratman (Toronto, Canada)
  • L. Siu (Toronto, Ontario, Canada)

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

Collapse
Poster Display (ID 17) Poster Display

10P - Deep Learning-based prediction of patient’s TLS status from HE images in pan-cancer cohort (ID 563)

Presentation Number
10P
Lecture Time
12:30 - 12:30
Speakers
  • L. Vanhersecke (Bordeaux, France)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Vanhersecke (Bordeaux, France)
  • L. GILLET (Paris, France)
  • J. Le Douget (Paris, France)
  • B. Schmauch (Paris, France)
  • C. Maussion (Paris, France)
  • A. Italiano (Bordeaux, CEDEX, France)
  • I. Soubeyran (Bordeaux, CEDEX, France)
  • F. Le Loarer (Bordeaux, France)

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.

Collapse
Poster Display (ID 17) Poster Display

11P - Association of VISTA expressing CD66b positive neutrophils, with response and survival benefit from pembrolizumab in advanced malignant pleural mesothelioma (ID 685)

Presentation Number
11P
Lecture Time
12:30 - 12:30
Speakers
  • K. Homicsko (Lausanne, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • K. Homicsko (Lausanne, Switzerland)
  • P. Zygoura (Athens, Greece)
  • S. Tissot (Lausanne, Switzerland)
  • M. Norkin (Lausanne, Switzerland)
  • S. Popat (London, United Kingdom)
  • A. Curioni-Fontecedro (Zurich, Switzerland)
  • M. O'Brien (London, United Kingdom)
  • T. Pope (, United Kingdom)
  • R. Shah (Maidstone, Kent, United Kingdom)
  • R. Kammler (Bern, Switzerland)
  • S. Finn (Dublin, Ireland)
  • G. Coukos (Epalinges, Switzerland)
  • U. Dafni (Athens, Attica, Greece)
  • S. Peters (Lausanne, Switzerland)
  • R. Stahel (Bern, Switzerland)

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

ClinicalTrials.gov, number NCT02991482.

ETOP 9-15 PROMISE-meso trial

Collapse
Poster Display (ID 17) Poster Display

12P - Multi-omics correlation with clinical outcome in metastatic melanoma patients treated with ipilimumab plus guadecitabine: the NIBIT-M4 study (ID 715)

Presentation Number
12P
Lecture Time
12:30 - 12:30
Speakers
  • A. Di Giacomo (Siena, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Di Giacomo (Siena, Italy)
  • T. Noviello (Napoli, Italy)
  • F. Caruso (Napoli, Italy)
  • G. Scala (Napoli, Italy)
  • L. Ferraro (Napoli, Italy)
  • A. Covre (Siena, Italy)
  • S. Coral (Siena, Italy)
  • A. Anichini (Milan, Italy)
  • M. Maio (Siena, Italy)
  • M. Ceccarelli (Napoli, Italy)

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

ClinicalTrials EudraCT, number 2015-001329-17,

ClinicalTrials.gov, number NCT02608437.

Collapse
Poster Display (ID 17) Poster Display

13P - Tumor infiltrating CD8/CD103/TIM-3-expressing lymphocytes in Epithelial Ovarian Cancer co-express CXCL13 and associate with improved survival (ID 265)

Presentation Number
13P
Lecture Time
12:30 - 12:30
Speakers
  • V. Bilemjian (Groningen, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • V. Bilemjian (Groningen, Netherlands)

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.

Collapse
Poster Display (ID 17) Poster Display

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 548)

Presentation Number
14P
Lecture Time
12:30 - 12:30
Speakers
  • J. Spronck (Nijmegen, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Spronck (Nijmegen, Netherlands)
  • L. Eekelen (Nijmegen, Netherlands)
  • L. Tessier (Angers, Cedex 02, France)
  • J. Bogaerts (Nijmegen, Netherlands)
  • L. Van der Woude (Nijmegen, Netherlands)
  • M. Van den Heuvel (Nijmegen, Netherlands)
  • W. Theelen (Amsterdam, Netherlands)
  • F. Ciompi (Nijmegen, Netherlands)

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

Collapse
Poster Display (ID 17) Poster Display

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 562)

Presentation Number
15P
Lecture Time
12:30 - 12:30
Speakers
  • G. Yao (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • G. Yao (Shanghai, China)
  • H. Fan (Shanghai, China)
  • R. Wang (Xiamen, China)
  • Y. Zhang (Shanghai, China)
  • C. Du (Xiamen, China)
  • B. Chen (Xiamen, China)
  • Z. Lin (Xiamen, China)
  • T. Zhang (Xiamen, China)
  • Z. Wu (Xiamen, China)

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.

Collapse
Poster Display (ID 17) Poster Display

16P - High fibrinogen levels affect the clinical outcomes of nivolumab in the second-line treatment of metastatic renal cell carcinoma patients: a prospective cohort study (ID 158)

Presentation Number
16P
Lecture Time
12:30 - 12:30
Speakers
  • I. Tsimafeyeu (Moscow, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • I. Tsimafeyeu (Moscow, United States of America)
  • M. Volkova (Moscow, Russian Federation)
  • I. Utyashev (Moscow, Russian Federation)
  • I. Gerk (Saint-Petersburg, Russian Federation)
  • K. Zakurdaeva (Moscow, Russian Federation)

Abstract

Background

A number of coagulation-related studies found that fibrinogen was elevated in renal cell carcinoma (RCC) patients and correlated with more advanced disease. The impact of hyperfibrinogenemia on the clinical outcomes of immunotherapy remains uncertain.

Methods

In this prospective cohort study, fibrinogen levels were measured one week prior to the second-line nivolumab therapy (240 mg or 480 mg every 2 or 4 weeks) and then monthly until disease progression or unacceptable toxicity. High fibrinogen level was defined as = >5 g/L. Key eligibility criteria included metastatic clear cell RCC resistant to the first-line targeted therapy, <= 3 MSKCC risk factors, ECOG PS 0-2, platelet count <400×10(9)/L, no heart diseases, no anticoagulation therapy, and age 40-65 years. The primary endpoint was overall survival (OS) in cohorts with high (H) and normal (N) fibrinogen levels. Secondary endpoints included progression-free survival (PFS), objective response rate (ORR), and fibrinogen levels during therapy.

Results

Between November 2018 and January 2021, 82 patients were enrolled, of whom 41 and 41, respectively, were recruited in H and N cohorts. The groups were well balanced at baseline. The median follow-up time was 32.4 months. Compared to patients in N cohort, patients in H cohort had shorter OS (P=0.001), shorter PFS (P<0.01), and lower ORR (P=0.012). In H cohort, a below-median fibrinogen level was associated with longer observed OS (median, 23.7 vs. 19.4 months; P=0.03). In N cohort, 6 (15%) patients with stable disease had an increase in fibrinogen levels during nivolumab therapy. The table summarizes patients’ characteristics and clinical outcomes.

H cohort,

N=41

N cohort,

N=41

Age (years), mean (range)

59 (40-65)

62 (43-65)

MSKCC risk factors, N (%)

2

3

35 (85)

6 (15)

33 (80)

8 (20)

ECOG PS, N (%)

0-1

2

29 (71)

12 (29)

28 (68)

13 (32)

First-line therapy, N (%)

Sunitinib

Pazopanib

Others

24 (59)

10 (24)

7 (17)

21 (51)

15 (37)

5 (12)

Fibrinogen (baseline), g/L, mean (SD)

6.4 (5.1-8.4)

2.9 (1.8-3.7)

OS, median (95% CI)

20.2 (17.4-25)

31.5 (26.8-NR)

PFS, median (95% CI)

4.0 (2.9-5.1)

9.4 (5.5-14.1)

ORR, N (%)

7 (17)

13 (32)

Conclusions

Detection of high baseline levels of fibrinogen in plasma derived from patients with metastatic RCC may be associated with poor clinical outcomes with nivolumab. These findings require large-scale prospective validation.

Clinical trial identification

KCRB20180412

Collapse
Poster Display (ID 17) Poster Display

17P - Using ex vivo organ culture technology to evaluate a T-Cell Bispecific (TCB) CEACAM5 Therapy (ID 406)

Presentation Number
17P
Lecture Time
12:30 - 12:30
Speakers
  • A. Zundelevich (Rehovot, Israel)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Zundelevich (Rehovot, Israel)
  • B. Ibrahim (Rehovot, Israel)
  • L. Turovsky (Rehovot, Israel)
  • S. Aharon (Rehovot, Israel)
  • V. Bar (Rehovot, Israel)
  • S. Salpeter (Rehovot, Israel)
  • G. Neev (Rehovot, Israel)
  • R. Straussman (Rehovot, Israel)
  • I. Silva (Basel, Switzerland)
  • S. Li (Basel, Switzerland)
  • S. Herter (Basel, Switzerland)
  • M. Bacac (Schlieren, Switzerland)
  • S. Sadok (Basel, Switzerland)
  • N. Gjorevski (Basel, Switzerland)

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.

Collapse
Poster Display (ID 17) Poster Display

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 439)

Presentation Number
18P
Lecture Time
12:30 - 12:30
Speakers
  • A. Tarhini (Tampa, PA, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Tarhini (Tampa, PA, United States of America)
  • S. Coleman (Tampa, United States of America)
  • I. El Naqa (Tampa, United States of America)
  • V. Sukrithan (Columbus, United States of America)
  • R. Aakrosh (Charlottesville, United States of America)
  • M. McCarter (Denver, United States of America)
  • J. Carpten (Los Angeles, United States of America)
  • H. Colman (Salt Lake City, UT, United States of America)
  • A. Ikeguchi (Oklahoma City, United States of America)
  • I. Puzanov (Buffalo, United States of America)
  • S. Arnold (Lexington, United States of America)
  • M. Churchman (Tampa, United States of America)
  • P. Hwu (Tampa, United States of America)
  • J. Conejo-Garcia (Tampa, United States of America)
  • W. Dalton (Tampa, United States of America)
  • G. Weiner (Iowa City, United States of America)
  • A. Tan (Tampa, United States of America)

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 1). 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 1. 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.

Table 1. 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

ClinicalTrials.Gov NCT03977402

Collapse
Poster Display (ID 17) Poster Display

19P - Effects of CTLA-4 Single Nucleotide Polymorphisms (SNPs) on toxicity of ipilimumab-containing regimens in patients with advanced stage melanoma (ID 569)

Presentation Number
19P
Lecture Time
12:30 - 12:30
Speakers
  • K. Joode (Rotterdam, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Rojas Mora (Bern, Switzerland)
  • R. Van Schaik (Rotterdam, Netherlands)
  • A. Zippelius (Basel, Switzerland)
  • A. Van der Veldt (Rotterdam, Netherlands)
  • C. Gerard (Lausanne, Switzerland)
  • H. Läubli (Basel, Switzerland)
  • O. Michielin (Lausanne, Switzerland)
  • R. Von Moos (Chur, Switzerland)
  • M. Joerger (St. Gallen, Switzerland)
  • M. Levesque (Zurich, Switzerland)
  • S. Aeppli (St. Gallen, Switzerland)
  • J. Mangana (Zurich, Switzerland)
  • C. Mangas (Bellinzona, Switzerland)
  • S. Meyer (Schlieren, Switzerland)
  • S. Leoni-Parvex (Locarno, Switzerland)
  • R. Mathijssen (Rotterdam, Netherlands)
  • Y. Metaxas (Muensterlingen, Switzerland)
  • K. Joode (Rotterdam, Netherlands)

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.

Collapse
Poster Display (ID 17) Poster Display

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 570)

Presentation Number
20P
Lecture Time
12:30 - 12:30
Speakers
  • M. Naigeon (Villejuif, Cedex, France)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Naigeon (Villejuif, Cedex, France)
  • M. Roulleaux Dugage (Villejuif Cedex, France)
  • F. Danlos (Villejuif, France)
  • C. De Oliveira (Paris, France)
  • L. Boselli (Villejuif Cedex, France)
  • J. Jouniaux (Villejuif Cedex, France)
  • F. Griscelli (Villejuif Cedex, France)
  • A. Marabelle (Villejuif, France)
  • L. Cassard (Villejuif, Cedex, France)
  • G. Roman (Baltimore, United States of America)
  • T. Hulett (Baltimore, United States of America)
  • B. Besse (Villejuif, CEDEX, France)
  • N. Chaput-Gras (Villejuif, Cedex, CEDEX, France)

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

Collapse
Poster Display (ID 17) Poster Display

21P - Peripheral pre-existing T-cell immunity as predictive biomarker in cancer immunotherapy for NSCLC patients (ID 689)

Presentation Number
21P
Lecture Time
12:30 - 12:30
Speakers
  • A. Xagara (LARISSA, Greece)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Xagara (LARISSA, Greece)
  • S. Fortis (Athens, Greece)
  • M. Goulielmaki (Athens, Greece)
  • F. Koinis (Larissa, Greece)
  • E. Chantzara (Larissa, Greece)
  • I. Samaras (Larissa, Greece)
  • V. Papadopoulos (Larissa, Greece)
  • V. Georgoulias (Heraklion, Greece)
  • C. Baxevanis (Athens, Greece)
  • A. Kotsakis (Heraklion, Greece)

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.

Collapse
Poster Display (ID 17) Poster Display

22P - Mean platelet volume to lymphocyte ratio: a new biomarker predictive of response in patients treated with immunotherapy (ID 264)

Presentation Number
22P
Lecture Time
12:30 - 12:30
Speakers
  • H. Yildirim (Ankara, Turkey)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • D. Guven (Ankara, Turkey)
  • F. Kus (Ankara, Turkey)
  • O. Dizdar (Ankara, Turkey)
  • S. Aksoy (Ankara, Turkey)
  • M. Erman (Ankara, Turkey)
  • Ş. Yalçın (Ankara, Turkey)
  • S. Kilickap (Ankara, Turkey)

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.

Collapse
Poster Display (ID 17) Poster Display

23P - Enriching for response: patient selection criteria for A2AR inhibition by EXS-21546 through ex vivo modelling in primary patient material (ID 363)

Presentation Number
23P
Lecture Time
12:30 - 12:30
Speakers
  • G. Vladimer (Vienna, Austria)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • G. Vladimer (Vienna, Austria)
  • I. Alt (Vienna, Austria)
  • R. Sehlke (Vienna, Austria)
  • A. Lobley (Oxford, United Kingdom)
  • C. Baumgärtler (Vienna, Austria)
  • M. Stulic (Vienna, Austria)
  • K. Hackner (Krems an der Donau, Austria)
  • L. Dzurillova (Bratislava, Slovak Republic)
  • E. Petru (Graz, Austria)
  • L. Hadjari (Linz, Austria)
  • J. Lafleur (Linz, Austria)
  • J. Singer (Krems an der Donau, Austria)
  • N. Krall (Vienna, Austria)
  • J. Šufliarsky (Bratislava, Slovak Republic)
  • L. Hefler (Ried im Innkreis, Austria)
  • T. Fuereder (Vienna, Austria)
  • C. Taubert (Vienna, Austria)
  • C. Boudesco (Vienna, Austria)
  • A. Payne (Oxford, United Kingdom)

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.

Collapse
Poster Display (ID 17) Poster Display

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 590)

Presentation Number
24P
Lecture Time
12:30 - 12:30
Speakers
  • A. Boutros (Genova, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Boutros (Genova, Italy)
  • R. Carosio (Genova, Italy)
  • D. Campanella (Genova, Italy)
  • F. Spagnolo (Genova, Italy)
  • B. Banelli (Genova, Italy)
  • A. Morabito (Genova, Italy)
  • M. Pistillo (Genova, Italy)
  • E. Croce (Genova, Italy)
  • F. Cecchi (Genova, Italy)
  • P. Pronzato (La Spezia, Italy)
  • P. Queirolo (Milan, Italy)
  • V. Fontana (Genova, Italy)
  • E. Tanda (Genova, Italy)

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.

Collapse
Poster Display (ID 17) Poster Display

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 661)

Presentation Number
25P
Lecture Time
12:30 - 12:30
Speakers
  • R. Ferrara (Milan, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • R. Ferrara (Milan, Italy)
  • G. Lo Russo (Milan, Italy)
  • C. Ciniselli (Milan, Italy)
  • B. Bassani (Milan, Italy)
  • G. Calareso (Milano, Italy)
  • V. Duroni (Milan, Italy)
  • S. Di Gregorio (Milan, Italy)
  • C. Proto (Milan, Italy)
  • A. Prelaj (Milan, Italy)
  • A. De Toma (Milan, Italy)
  • M. Occhipinti (Milan, Italy)
  • M. Brambilla (Milan, Italy)
  • S. Manglaviti (Milan, Italy)
  • L. Mazzeo (Milan, Italy)
  • M. Ganzinelli (Milan, Italy)
  • F. De Braud (Milan, Italy)
  • M. Garassino (Chicago, United States of America)
  • M. Colombo (Milan, Italy)
  • P. Verderio (Milan, Italy)
  • S. Sangaletti (Milan, Italy)

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.

Collapse
Poster Display (ID 17) Poster Display

26P - Impact of SF3B1 mutation on response to immunotherapy (ID 268)

Presentation Number
26P
Lecture Time
12:30 - 12:30
Speakers
  • B. Lim (Daejeon, Korea, Republic of)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • B. Lim (Daejeon, Korea, Republic of)
  • K. Cho (Daejeon, Korea, Republic of)

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.

Collapse
Poster Display (ID 17) Poster Display

27P - Gene ontology enrichment analysis of peripheral CD3+CD4+T-cells predicts the immune-mediated behavior of melanoma (ID 484)

Presentation Number
27P
Lecture Time
12:30 - 12:30
Speakers
  • D. Ziogas (Athens, Greece)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • D. Ziogas (Athens, Greece)
  • E. Palli (Athens, Greece)
  • P. Verginis (Heraklion, Greece)
  • M. Lavigne (Heraklion, Greece)
  • T. Alissafi (Athens, Greece)
  • P. Diamantopoulos (Athens, Greece)
  • H. Gogas (Athens, Attiki, Greece)

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.

Collapse
Poster Display (ID 17) Poster Display

28P - Epithelial Mesenchymal transition confers resistance to dual PDL1-VEGF inhibition in relapsed mesothelioma (ID 604)

Presentation Number
28P
Lecture Time
12:30 - 12:30
Speakers
  • E. Baitei (Leicester, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • E. Baitei (Leicester, United Kingdom)
  • M. Zhang (Beijing, China)
  • C. Poile (Leicester, United Kingdom)
  • J. Luo (Leicester, United Kingdom)
  • J. Dzialo (Leicester, United Kingdom)
  • Z. Zhou (Leicester, United Kingdom)
  • J. Harber (Leicester, United Kingdom)
  • C. Pritchard (Leicester, United Kingdom)
  • T. Kamata (Leicester, United Kingdom)
  • A. King (Leicester, United Kingdom)
  • A. Branson (Leicester, United Kingdom)
  • S. Barber (Leicester, United Kingdom)
  • A. Thomas (Leicester, United Kingdom)
  • H. Yang (Leicester, United Kingdom)
  • D. Fennell (Leicester, United Kingdom)

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.

Collapse
Poster Display (ID 17) Poster Display

29P - Circulating proteins associated with immunotherapy efficacy in patients with pancreatic ductal adenocarcinoma. (ID 457)

Presentation Number
29P
Lecture Time
12:30 - 12:30
Speakers
  • T. Christensen (Herlev, Denmark)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • T. Christensen (Herlev, Denmark)
  • E. Maag (Aarhus, Denmark)
  • S. Theile (Herlev, Denmark)
  • K. Madsen (Herlev, Denmark)
  • S. Lindgaard (Herlev, Denmark)
  • D. Nielsen (Herlev, Denmark)
  • I. Chen (Herlev, Denmark)
  • J. Johansen (Herlev, Denmark)

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

Collapse
Poster Display (ID 17) Poster Display

30P - Frequency of peripheral CD8+ T cells expressing chemo-attractant receptors increases in NPC patients with EBV clearance upon radiotherapy (ID 595)

Presentation Number
30P
Lecture Time
12:30 - 12:30
Speakers
  • S. Mahajan (Rotterdam, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Mahajan (Rotterdam, Netherlands)
  • A. Oostvogels (Rotterdam, Netherlands)
  • E. Balcioglu (Rotterdam, Netherlands)
  • K. Lo (Sha Tin, Hong Kong PRC)
  • A. Chan (Hong Kong, Hong Kong PRC)
  • E. Hui (Sha Tin, Hong Kong PRC)
  • A. Tsang (Sha Tin, Hong Kong PRC)
  • B. Ma (Sha Tin, Hong Kong PRC)
  • R. Debets (Rotterdam, Netherlands)

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.

Collapse
Poster Display (ID 17) Poster Display

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 646)

Presentation Number
31P
Lecture Time
12:30 - 12:30
Speakers
  • K. Joode (Rotterdam, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • H. Groen (Groningen, Netherlands)
  • M. Van den Heuvel (Nijmegen, Netherlands)
  • J. Kloover (Tilburg, Netherlands)
  • F. Van der Meer (Utrecht, Netherlands)
  • J. Stigt (Zwolle, Netherlands)
  • C. Van Der Leest (Breda, Netherlands)
  • B. Van Den Borne (Eindhoven, Netherlands)
  • R. De Wijn ('s-Hertogenbosch, Netherlands)
  • D. Hurkmans (Rotterdam, Netherlands)
  • D. Van den Heuvel ('s-Hertogenbosch, Netherlands)
  • T. Van Doorn ('s-Hertogenbosch, Netherlands)
  • B. Pinedo (Willemstad, Curacao)
  • E. Kapiteijn (Leiden, Netherlands)
  • R. Debets (Rotterdam, Netherlands)
  • E. Verdegaal (Leiden, Netherlands)
  • S. Van der Burg (Leiden, Netherlands)
  • J. Groten ('s-Hertogenbosch, Netherlands)
  • J. Aerts (Rotterdam, Noord Brabant, Netherlands)
  • R. Mathijssen (Rotterdam, Netherlands)

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.

Collapse
Poster Display (ID 17) Poster Display

32P - Discovering markers that identify pro-metastatic immune cell subsets. (ID 660)

Presentation Number
32P
Lecture Time
12:30 - 12:30
Speakers
  • A. Bokil (Trondheim, Norway)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Bokil (Trondheim, Norway)
  • M. Børkja (Trondheim, Norway)
  • C. Wolowczyk (Trondheim, Norway)
  • A. Bofin (Trondheim, Norway)
  • S. Hak (Trondheim, Norway)
  • M. Giambelluca (Trondheim, Norway)

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.

Collapse
Poster Display (ID 17) Poster Display

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 290)

Presentation Number
33P
Lecture Time
12:30 - 12:30
Speakers
  • Y. Xu (Hangzhou, Zhejiang, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • H. Li (Hangzhou, China)
  • Z. Huang (Hangzhou, China)
  • M. Xie (Hangzhou, China)
  • Z. Zhou (Hangzhou, China)
  • Y. Fan (Hangzhou, China)

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 .

Collapse
Poster Display (ID 17) Poster Display

34TiP - Prospective Analysis of INteStinal Microbiome and Autoimmune PanEls as PrediCtors of Toxicity in ImmunOncology Patients (INSPECT-IO Study) (ID 167)

Presentation Number
34TiP
Lecture Time
12:30 - 12:30
Speakers
  • A. Salawu (Toronto, South Yorkshire, Canada)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Salawu (Toronto, South Yorkshire, Canada)
  • S. Genta (Toronto, Canada)
  • P. Spiliopoulou (Glasgow, United Kingdom)
  • N. Yee (Toronto, Canada)
  • M. Ye (Toronto, Canada)
  • L. Ly (Toronto, Canada)
  • R. Taylor (Toronto, Canada)
  • O. Vornicova (Afula, Israel)
  • A. Abdul Razak (Toronto, Canada)
  • P. Bedard (Toronto, Ontario, Canada)
  • L. Wang (Toronto, Canada)
  • L. Siu (Toronto, Ontario, Canada)
  • B. Coburn (Toronto, Canada)
  • A. Spreafico (Toronto, CO, Canada)

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

Collapse
Poster Display (ID 17) Poster Display

Cell therapy immune engineering (ID 802)

Lecture Time
12:30 - 12:30
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Poster Display (ID 17) Poster Display

37P - Synthetic tumour-infiltrating interleukin(IL)-12 for targeted and tolerable immunotherapy reduces metastasis in pancreatic cancer (ID 691)

Presentation Number
37P
Lecture Time
12:30 - 12:30
Speakers
  • M. Herpels (London, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Herpels (London, United Kingdom)
  • A. Mansurov (Chicago, United States of America)
  • C. Ragulan (Sutton, United Kingdom)
  • A. Hussain (London, United Kingdom)
  • J. Ishihara (London, United Kingdom)
  • A. Sadanandam (London, United Kingdom)

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.

Collapse
Poster Display (ID 17) Poster Display

39P - The expanded CD8+ tumor-infiltrating lymphocyte tumor-specific granzyme response is dominated by granzyme B (ID 540)

Presentation Number
39P
Lecture Time
12:30 - 12:30
Speakers
  • A. Rasmussen (Herlev, Denmark)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Rasmussen (Herlev, Denmark)
  • C. Chamberlain (Herlev, Denmark)
  • K. Harbst (Lund, Sweden)
  • S. Khan (Herlev, Denmark)
  • A. Schina (Herlev, Denmark)
  • A. Draghi (Herlev, Denmark)
  • M. Presti (Herlev, Denmark)
  • A. Jensen (Herlev, Denmark)
  • G. Jönsson (Lund, Sweden)
  • I. Svane (Herlev, Denmark)
  • M. Donia (Herlev, Denmark)

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).

Collapse
Poster Display (ID 17) Poster Display

40P - Can we predict which TIL products will react against NSCLC tumours based on the immune infiltrates? (ID 239)

Presentation Number
40P
Lecture Time
12:30 - 12:30
Speakers
  • S. Castenmiller (Amsterdam, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Castenmiller (Amsterdam, Netherlands)
  • N. Kanagasabesan (Amsterdam, Netherlands)
  • R. De Groot (Amsterdam, Netherlands)
  • A. Guislain (Amsterdam, Netherlands)
  • M. Van Loenen (Amsterdam, Netherlands)
  • K. Monkhorst (Amsterdam, Netherlands)
  • K. Hartemink (Amsterdam, Netherlands)
  • H. Klomp (Amsterdam, Netherlands)
  • E. Smit (Amsterdam, Netherlands)
  • J. Haanen (Amsterdam, Netherlands)
  • M. Wolkers (Amsterdam, Netherlands)

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.

Collapse
Poster Display (ID 17) Poster Display

41P - Single-Cell Signalling Analysis of Engineered γδ T cell Biotherapeutics for the Treatment of Colorectal Cancer (ID 203)

Presentation Number
41P
Lecture Time
12:30 - 12:30
Speakers
  • C. Nattress (London, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • P. Vlckova (London, United Kingdom)
  • D. Fowler (London, United Kingdom)
  • J. Sufi (London, United Kingdom)
  • M. Ramos Zapatero (London, United Kingdom)
  • F. Cardoso Rodriguez (London, United Kingdom)
  • A. Campbell (London, United Kingdom)
  • A. Southern Navarrete (London, United Kingdom)
  • X. Qin (London, United Kingdom)
  • J. Opzoomer (London, United Kingdom)
  • M. Barisa (London, United Kingdom)
  • J. Fisher (London, United Kingdom)
  • C. Tape (London, United Kingdom)

Abstract

Background

Colorectal cancer (CRC) is a devastating disease that kills ~700,000 people worldwide annually, with immunotherapies struggling against the immunosuppressive CRC tumour microenvironment (TME). Tumour infiltrating γδ T cells confer a prognostic benefit to CRC patients and can kill cancer via multiple innate and adaptive mechanisms, including antibody-dependent cellular cytotoxicity (ADCC) against tumour antigens (e.g., B7-H3). We hypothesise that γδ T cells can be exploited as an anti-CRC biotherapeutic but their complex interactions within the CRC TME need to be elucidated.

Methods

We performed 3D TME cultures of CRC patient-derived organoids (PDOs) with CRC cancer-associated fibroblasts (CAFs) and human Vγ9Vδ2 T cells, either unmodified or engineered to secrete a modified IL15 cytokine. The addition of anti-B7-H3 IgG also allows the modelling of anti-PDO ADCC, with 3D CRISPR-Cas9 PDO models generated for demonstration of antigen specificity. Using 126-plex Thiol Organoid Barcoding in situ (TOBis) mass cytometry (MC), we measure over 60 parameters per cell across hundreds of culture conditions (Qin et al., Nature Methods, 2020) (Sufi and Qin et al., Nature Protocols, 2021). Cell-type specific signalling analysis across multiple γδ donors and CRC PDOs including post-translational modifications (PTMs), cell-state and immunological phenotype were analysed computationally.

Results

Engineered γδ T cells exhibit superior proliferation, purity, cytotoxicity, and viability, with γδ donor-specific responses to cytokine engineering (127IdU+ and pSTAT5 [Y694]+) and PDO co-culture (GranzymeB+ and CD69+). Multiple engineered γδ T cells generate a substantial killing phenotype against various CRC PDOs (cCaspase3 [D175]+ and cPARP [D214]+). We also detect the transfer of Granzyme B from γδ T cells to PDOs, executing PDO apoptosis. ADCC varies between γδ donors and is associated with FcγR CD16 expression.

Conclusions

We have identified marker signatures for the selection of candidate γδ biotherapeutics. CAFs can protect PDOs from γδ ADCC and work is continuing to understand the mechanistic basis of biotherapeutic stromal protection.

Collapse
Poster Display (ID 17) Poster Display

42P - Tumor-reactive CD8+ T cells in ovarian and colon cancer in tumors and cell products (ID 670)

Presentation Number
42P
Lecture Time
12:30 - 12:30
Speakers
  • S. Bobisse (Lausanne, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Bobisse (Lausanne, Switzerland)
  • B. Navarro Rodrigo (Lausanne, Switzerland)
  • Q. Ngo (Lausanne, Switzerland)
  • J. Chiffelle (Lausanne, Switzerland)
  • R. Genolet (Lausanne, Switzerland)
  • A. Michel (Lausanne, Switzerland)
  • D. Saugy (Lausanne, Switzerland)
  • C. Sauvage (Lausanne, Switzerland)
  • D. Tarussio (Lausanne, Switzerland)
  • M. Arnaud (Epalinges, Switzerland)
  • P. Guillaume (Lausanne, Switzerland)
  • B. Stevenson (Lausanne, Switzerland)
  • M. Bassani-Sternberg (Lausanne, CH, Switzerland)
  • S. Tissot (Lausanne, Switzerland)
  • S. Rusakiewicz (Lausanne, Switzerland)
  • J. Schmidt (Lausanne, Switzerland)
  • D. Dangaj (Lausanne, Switzerland)
  • L. Kandalaft (Lausanne, Switzerland)
  • G. Coukos (Epalinges, Switzerland)
  • A. Harari (Lausanne, Switzerland)

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.

Collapse
Poster Display (ID 17) Poster Display

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 711)

Presentation Number
43P
Lecture Time
12:30 - 12:30
Speakers
  • L. Tas (Amsterdam, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Tas (Amsterdam, Netherlands)
  • M. Van Zon (Amsterdam, Netherlands)
  • I. Djuretic (South San Francisco, United States of America)
  • K. Moynihan (South San Francisco, United States of America)
  • Y. Yeung (South San Francisco, United States of America)
  • N. Mathewson (South San Francisco, United States of America)
  • J. Haanen (Amsterdam, Netherlands)
  • I. Jedema (Amsterdam, Netherlands)

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.

Collapse
Poster Display (ID 17) Poster Display

44P - In situ CAR Therapy Using oRNA™ Lipid Nanoparticles Regresses Tumors in Mice (ID 174)

Presentation Number
44P
Lecture Time
12:30 - 12:30
Speakers
  • R. Mabry (Cambridge, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Becker (Cambridge, United States of America)
  • A. Wesselhoeft (Cambridge, United States of America)
  • A. Horhata (Cambridge, United States of America)

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 imunotropic 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.
Collapse
Poster Display (ID 17) Poster Display

45P - The RevCAR T cell platform: a switchable and combinatorial therapeutic strategy for glioblastoma (ID 428)

Presentation Number
45P
Lecture Time
12:30 - 12:30
Speakers
  • N. Mitwasi (Dresden, Germany)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • N. Mitwasi (Dresden, Germany)
  • H. Hassan (Dresden, Germany)
  • C. Arndt (Dresden, Germany)
  • L. Loureiro (Dresden, Germany)
  • C. Neuber (Dresden, Germany)
  • A. Kegler (Dresden, Germany)
  • M. Kubeil (Dresden, Germany)
  • M. Toussaint (Dresden, Germany)
  • W. Deuther-Conrad (Dresden, Germany)
  • M. Bachmann (Dresden, Germany)
  • A. Feldmann (Dresden, Germany)

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.

Collapse
Poster Display (ID 17) Poster Display

46P - Development of an allogeneic CAR-T targeting MUC1-C (MUC1, cell surface associated, C-terminal) for epithelial derived tumors (ID 407)

Presentation Number
46P
Lecture Time
12:30 - 12:30
Speakers
  • D. Oh (San Francisco, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • D. Oh (San Francisco, United States of America)
  • J. Henry (Nashville, United States of America)
  • J. Baranda (Kansas City, KS, United States of America)
  • E. Dumbrava (Houston, TX, United States of America)
  • E. Cohen (La Jolla, IL, United States of America)
  • J. Eskew (San Diego, United States of America)
  • R. Belani (San Diego, United States of America)
  • J. McCaigue (San Diego, United States of America)
  • H. Namini (San Diego, United States of America)
  • C. Martin (San Diego, United States of America)
  • A. Murphy (San Diego, United States of America)
  • E. Ostertag (San Diego, United States of America)
  • J. Coronella (San Diego, United States of America)
  • D. Shedlock (San Diego, United States of America)
  • I. Rodriguez Rivera (San Antonio, United States of America)

Abstract

Background

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

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

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

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

Collapse
Poster Display (ID 17) Poster Display

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 705)

Presentation Number
47P
Lecture Time
12:30 - 12:30
Speakers
  • M. Kocoglu (Baltimore, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Kocoglu (Baltimore, United States of America)
  • A. Asch (Oklahoma City, United States of America)
  • A. Ramakrishnan (Austin, United States of America)
  • C. Bachier (San Antonio, United States of America)
  • T. Martin (San Francisco, United States of America)
  • T. Rodriguez (Park Ridge, United States of America)
  • K. McArthur (San Diego, United States of America)
  • C. Martin (San Diego, United States of America)
  • H. Namini (San Diego, United States of America)
  • E. Ostertag (San Diego, United States of America)
  • M. Spear (San Diego, United States of America)
  • E. Christie (San Diego, United States of America)
  • R. Belani (San Diego, United States of America)
  • M. Zhang (San Diego, United States of America)
  • S. Cranert (San Diego, United States of America)
  • J. Coronella (San Diego, United States of America)
  • D. Shedlock (San Diego, United States of America)
  • C. Costello (La Jolla, United States of America)

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

Collapse
Poster Display (ID 17) Poster Display

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 443)

Presentation Number
48P
Lecture Time
12:30 - 12:30
Speakers
  • L. Ridolfi (Meldola, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Ridolfi (Meldola, Italy)
  • L. Gurrieri (Meldola, (MI), Italy)
  • M. Riva (Forlì, Italy)
  • J. Bulgarelli (Meldola, Italy)
  • V. Fausti (Meldola, Forlì-Cesena, Italy)
  • F. De rosa (Meldola, Italy)
  • M. Guidoboni (Meldola, Forlì-Cesena, Italy)
  • F. Foca (Meldola, Italy)
  • M. Tazzari (Meldola, Italy)
  • M. Petrini (Meldola, Italy)
  • A. Granato (Meldola, Italy)
  • E. Pancisi (Meldola, Italy)
  • M. Dall'Agata (Meldola, Italy)
  • E. Amadori (Meldola, Italy)
  • A. Gamboni (Lugo, Italy)
  • G. Pasini (Rimini, Italy)
  • P. Cortesi (Meldola, Italy)
  • L. Mercatali (Meldola, Italy)
  • A. Bongiovanni (Meldola, Forlì-Cesena, Italy)
  • L. Tosatto (Cesena, Italy)

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 number: 2020-003755-15

Collapse
Poster Display (ID 17) Poster Display

49P - Exploiting multi-omic integrated data from DC-vaccinated melanoma patients for the generation of an advanced adoptive T cell therapy (ID 579)

Presentation Number
49P
Lecture Time
12:30 - 12:30
Speakers
  • J. Bulgarelli (Meldola, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Bulgarelli (Meldola, Italy)
  • M. Tazzari (Meldola, Italy)
  • S. Carloni (Meldola, Italy)
  • S. Pignatta (Meldola, Italy)
  • M. Bochicchio (Meldola, Italy)
  • M. Bocchini (Meldola, Italy)
  • D. Angeli (Meldola, Italy)
  • M. Tebaldi (Meldola, Italy)
  • A. Granato (Meldola, Italy)
  • C. Piccinini (Meldola, Italy)
  • E. Pancisi (Meldola, Italy)
  • F. De rosa (Meldola, Italy)
  • V. Ancarani (Meldola, Italy)
  • F. Limarzi (Forlì, Italy)
  • M. Petrini (Meldola, Italy)
  • L. Ridolfi (Meldola, Italy)

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.

Collapse
Poster Display (ID 17) Poster Display

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 455)

Presentation Number
50P
Lecture Time
12:30 - 12:30
Speakers
  • D. Quixabeira (Helsinki, Finland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • D. Quixabeira (Helsinki, Finland)
  • S. Pakola (Helsinki, Finland)
  • E. Jirovec (Helsinki, Finland)
  • S. Basnet (Helsinki, Finland)
  • J. Santos (Helsinki, Finland)
  • T. Kudling (Helsinki, Finland)
  • J. Clubb (Helsinki, Finland)
  • L. Haybout (Helsinki, Finland)
  • V. Arias (Helsinki, Finland)
  • S. Grönberg-Vähä-Koskela (Helsinki, Finland)
  • R. Havunen (Helsinki, Finland)
  • V. Cervera-Carrascon (Helsinki, Finland)
  • A. Pasanen (Helsinki, Finland)
  • J. Tapper (Helsinki, Finland)
  • A. Kanerva (Helsinki, Finland)
  • A. Hemminki (Helsinki, Finland)

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

Collapse
Poster Display (ID 17) Poster Display

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 355)

Presentation Number
51P
Lecture Time
12:30 - 12:30
Speakers
  • S. Pakola (Helsinki, Finland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Pakola (Helsinki, Finland)
  • J. Santos (Helsinki, Finland)
  • V. Cervera-Carrascon (Helsinki, Finland)
  • C. Kistler (Helsinki, Finland)
  • S. Sorsa (Helsinki, Finland)
  • R. Havunen (Helsinki, Finland)
  • A. Hemminki (Helsinki, Finland)

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)

Collapse
Poster Display (ID 17) Poster Display

52P - MUC1 targeted immunotherapy with an oncolytic adenovirus coding for a bispecific T cell engager (ID 466)

Presentation Number
52P
Lecture Time
12:30 - 12:30
Speakers
  • S. Basnet (Helsinki, Finland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Basnet (Helsinki, Finland)
  • J. Santos (Helsinki, Finland)
  • D. Quixabeira (Helsinki, Finland)
  • J. Clubb (Helsinki, Finland)
  • S. Grönberg-Vähä-Koskela (Helsinki, Finland)
  • S. Pakola (Helsinki, Finland)
  • T. Kudling (Helsinki, Finland)
  • C. Heiniö (Helsinki, Finland)
  • R. Havunen (Helsinki, Finland)
  • V. Cervera-Carrascon (Helsinki, Finland)
  • S. Sorsa (Helsinki, Finland)
  • M. Anttila (Helsinki, Finland)
  • A. Kanerva (Helsinki, Finland)
  • A. Hemminki (Helsinki, Finland)

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.

Collapse
Poster Display (ID 17) Poster Display

53P - Combining cancer vaccines based on arenavirus vectors with 4-1BB (CD137) agonists enhances therapeutic efficacy in a non-inflamed tumor model (ID 504)

Presentation Number
53P
Lecture Time
12:30 - 12:30
Speakers
  • J. Strauss (New York, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Strauss (New York, United States of America)
  • D. Reckendorfer (Vienna, Austria)
  • K. Pojar (Vienna, Austria)
  • T. Pölzlbauer (Vienna, Austria)
  • M. Habbeddine (Tuebingen, Germany)
  • M. Scheinost (Vienna, Austria)
  • S. Ahmadi-Erber (Vienna, Austria)
  • S. Schmidt (Vienna, Austria)
  • J. Raguz (Vienna, Austria)
  • J. Lampert (Vienna, Austria)
  • K. Orlinger (Vienna, Austria)
  • H. Lauterbach (Vienna, Austria)

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.

Collapse
Poster Display (ID 17) Poster Display

54P - Oncolytic virus combined PD-1 antibody Toripalimab in advanced lung cancer with liver metastases: An Early Stage, Single Arm, Study (TROJAN 2201) (ID 658)

Presentation Number
54P
Lecture Time
12:30 - 12:30
Speakers
  • J. Yang (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Yang (Shanghai, China)
  • H. Wang (Shanghai, China)
  • Z. Zhou (Shanghai, China)
  • X. Niu (Shanghai, China)
  • C. Qu (Shanghai, China)
  • X. Guo (Shanghai, China)
  • J. Wu (Shanghai, China)
  • S. Lu (Shanghai, China)
  • Q. Xu (Shanghai, China)

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.

Collapse
Poster Display (ID 17) Poster Display

55P - Mutations Localized at the Membrane Predict Immunotherapeutic Efficacy in Cancer Treatment (ID 505)

Presentation Number
55P
Lecture Time
12:30 - 12:30
Speakers
  • Z. Goldberger (Montreal, Canada)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Z. Goldberger (Montreal, Canada)
  • S. Hauert (Chicago, United States of America)
  • K. Chang (Chicago, United States of America)
  • T. Kurtanich (Chicago, United States of America)
  • A. Alpar (Chicago, United States of America)
  • J. Hubbell1 (Chicago, United States of America)
  • P. Briquez (Freiburg im Breisgau, Germany)

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.

Collapse
Poster Display (ID 17) Poster Display

Clinical practice (ID 803)

Lecture Time
12:30 - 12:30
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Poster Display (ID 17) Poster Display

63P - Activity and safety of first-line treatments for advanced melanoma: a network meta-analysis (ID 204)

Presentation Number
63P
Lecture Time
12:30 - 12:30
Speakers
  • A. Boutros (Genova, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Boutros (Genova, Italy)
  • E. Tanda (Genova, Italy)
  • E. Croce (Genova, Italy)
  • F. Catalano (Genova, Italy)
  • M. Ceppi (Genova, Italy)
  • M. Bruzzone (Genova, Italy)
  • F. Cecchi (Genova, Italy)
  • L. Arecco (Genova, Italy)
  • M. Fraguglia (Genova (GE), Italy)
  • P. Pronzato (La Spezia, Italy)
  • L. Del Mastro (Genova, Italy)
  • M. Lambertini (Genova, Italy)
  • F. Spagnolo (Genova, Italy)

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.

Collapse
Poster Display (ID 17) Poster Display

64P - Safety and efficacy of immune checkpoint inhibitors (ICIs) in patients (pts) with solid tumors and cardiac metastases (ID 633)

Presentation Number
64P
Lecture Time
12:30 - 12:30
Speakers
  • A. Nassar (New Haven, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Nassar (New Haven, United States of America)
  • T. Zarif (Boston, United States of America)
  • S. Abou Alaiwi (Boston, United States of America)
  • R. Denu (Houston, United States of America)
  • W. Macaron (Houston, United States of America)
  • E. El-Am (Rochester, United States of America)
  • C. Malvar (La Jolla, United States of America)
  • J. Ocejo Gallegos (Miami, United States of America)
  • A. Cortellini (London, United Kingdom)
  • J. Korolewicz (London, United Kingdom)
  • P. Sackstein (Washington DC, United States of America)
  • F. Aboubakar (Brussels, Belgium)
  • S. Foderaro (Rome, Italy)
  • J. Baena Espinar (Madrid, Spain)
  • R. Woodford (Wollstonecraft, Australia)
  • S. Grynberg (Ramat Gan, Israel)
  • A. Asnani (St. Louis, United States of America)
  • S. Hayek (Ann Arbor, United States of America)
  • T. Choueiri (Boston, United States of America)
  • A. Naqash (Oklahoma City, United States of America)

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 table 1.

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.

Collapse
Poster Display (ID 17) Poster Display

65P - Atezolizumab plus bevacizumab versus Lenvatinib for unresectable hepatocellular carcinoma: a large real life worldwide population (ID 592)

Presentation Number
65P
Lecture Time
12:30 - 12:30
Speakers
  • M. Rimini (Modena, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Casadei Gardini (Milan, Forlì-Cesena, Italy)
  • M. Persano (Cagliari, Italy)
  • G. Suda (Sapporo, Japan)
  • T. Tada (Himeji, Japan)
  • S. Shimose (Kurume, Japan)
  • M. Kudo (Osaka, Japan)
  • J. Cheon (Ulsan, Korea, Republic of)
  • F. Finkelmeier (Frankfurt am Main, Germany)
  • L. Rimassa (Rozzano, (MI), Italy)
  • J. Presa (Vila real, Portugal)
  • G. Masi (Pisa, Italy)
  • C. Yoo (Seoul, Songpa-gu, Korea, Republic of)
  • S. Lonardi (Padova, Italy)
  • F. Tovoli (Bologna, Italy)
  • F. Piscaglia (Bologna, Italy)
  • M. Iavarone (Milan, Italy)
  • M. Scartozzi (Monserrato, Italy)
  • V. Burgio (Catanzaro, Italy)
  • S. Cascinu (Modena, Italy)
  • A. Cucchetti (Bologna, Italy)

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.

Collapse
Poster Display (ID 17) Poster Display

66P - Real-world data of first-line chemo-immunotherapy for patients with extensive stage SCLC: A multicentre experience from Switzerland and the UK (ID 676)

Presentation Number
66P
Lecture Time
12:30 - 12:30
Speakers
  • L. Moliner (Manchester, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Moliner (Manchester, United Kingdom)
  • N. Zellweger (Basel, Switzerland)
  • S. Schmidt (St. Gallen, Switzerland)
  • C. Waibel (Baden, Aargau, Switzerland)
  • P. Froesch (Locarno, Switzerland)
  • P. Häuptle (Liestal, Switzerland)
  • V. Blum (Luzern, Switzerland)
  • L. Holer (Bern, Switzerland)
  • M. Frueh (St. Gallen, Switzerland)
  • S. Bhagani (Leicester, United Kingdom)
  • H. Gray (Glasgow, United Kingdom)
  • S. Cox (Cardiff, United Kingdom)
  • T. Khalid (Sligo, Ireland)
  • D. Scott (Swindon, United Kingdom)
  • S. Robinson (Brighton, East Sussex, United Kingdom)
  • L. Hennah (London, United Kingdom)
  • C. Handforth (Huddersfield, Yorkshire, United Kingdom)
  • L. Mauti (Winterthur, Switzerland)
  • R. Califano (Manchester, United Kingdom)
  • S. Rothschild (Basel, Switzerland)

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.

Collapse
Poster Display (ID 17) Poster Display

67P - Real Word Data for Atezolizumab plus Bevacizumab in unresectable Hepatocellular Carcinoma (ID 645)

Presentation Number
67P
Lecture Time
12:30 - 12:30
Speakers
  • M. Persano (Cagliari, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Persano (Cagliari, Italy)
  • M. Rimini (Modena, Italy)
  • T. Tada (Himeji, Japan)
  • G. Suda (Sapporo, Japan)
  • S. Shimose (Kurume, Japan)
  • M. Kudo (Osaka, Japan)
  • J. Cheon (Ulsan, Korea, Republic of)
  • F. Finkelmeier (Frankfurt am Main, Germany)
  • L. Rimassa (Rozzano, (MI), Italy)
  • J. Presa (Vila real, Portugal)
  • G. Masi (Pisa, Italy)
  • C. Yoo (Seoul, Songpa-gu, Korea, Republic of)
  • S. Lonardi (Padova, Italy)
  • F. Piscaglia (Bologna, Italy)
  • V. Burgio (Catanzaro, Italy)
  • M. Scartozzi (Monserrato, Italy)
  • S. Cascinu (Modena, Italy)
  • A. Casadei Gardini (Milan, Forlì-Cesena, Italy)

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.

Collapse
Poster Display (ID 17) Poster Display

69P - A Real-World Analysis of Treatment-Free survival for Advanced Melanoma Patients treated with First-Line Immune Checkpoint Inhibitors (ID 435)

Presentation Number
69P
Lecture Time
12:30 - 12:30
Speakers
  • M. Gupta (Calgary, Canada)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Gupta (Calgary, Canada)
  • I. Stukalin (Calgary, Canada)
  • S. Goutam (Calgary, Canada)
  • D. Meyers (Calgary, Canada)
  • D. Heng (Calgary, OH, Canada)
  • T. Cheng (Calgary, Alberta, Canada)
  • V. Navani (Calgary, Canada)

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.

Collapse
Poster Display (ID 17) Poster Display

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 258)

Presentation Number
70P
Lecture Time
12:30 - 12:30
Speakers
  • J. Liu (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • X. Zhu (Shanghai, China)
  • Y. Cheng (Shanghai, China)
  • H. Li (Shanghai, China)
  • X. Ma (Shanghai, China)
  • X. Fu (Shanghai, China)

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

Collapse
Poster Display (ID 17) Poster Display

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 277)

Presentation Number
71P
Lecture Time
12:30 - 12:30
Speakers
  • Y. Yu (Harbin, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Y. Yu (Harbin, China)
  • L. Shang (Harbin, China)
  • Q. Xie (Chongqing, China)
  • K. Ying (Hangzhou, China)
  • B. Jin (Shenyang, Liaoning, China)
  • L. Zhang (Shanghai, China)
  • L. Liu (Shanghai, China)
  • J. Li (Shanghai, China)
  • W. Ding (Shanghai, China)

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.

Table: 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.

Collapse
Poster Display (ID 17) Poster Display

72P - Treatments response in non-small cell lung cancer patients according to BRCA status on liquid biopsy: a retrospective analysis (ID 653)

Presentation Number
72P
Lecture Time
12:30 - 12:30
Speakers
  • L. Provenzano (Milan, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Provenzano (Milan, Italy)
  • A. Bottiglieri (Milan, Italy)
  • A. Spagnoletti (Milan, Italy)
  • G. Di Guida (Caserta, Italy)
  • J. Filosa (Napoli, Italy)
  • L. Mazzeo (Milan, Italy)
  • C. Giani (Milan, Italy)
  • V. Miskovic (Milan, Italy)
  • C. Della Corte (Napoli, Italy)
  • G. Viscardi (Napoli, Italy)
  • A. Prelaj (Milan, Italy)

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.

Collapse
Poster Display (ID 17) Poster Display

73P - A randomized phase II study of neoadjuvant immunotherapy or immunochemotherapy in locally advanced oral squamous cell carcinoma (ID 682)

Presentation Number
73P
Lecture Time
12:30 - 12:30
Speakers
  • G. Chen (Wuhan, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • H. Liu (Wuhan, China)
  • X. Xiong (Wuhan, China)
  • Z. Yu (Wuhan, China)
  • Z. Shao (Wuhan, China)
  • Y. Liu (Wuhan, China)
  • X. Wang (Wuhan, China)
  • Q. Fu (Wuhan, China)
  • X. Cheng (Wuhan, China)
  • J. Li (Wuhan, China)
  • J. Jia (Wuhan, China)
  • B. Liu (Wuhan, China)

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

Arm A

Camrelizumab

(n = 30)

Arm B

Camrelizumab + TPF

(n = 30)
Pathological response 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

ClinicalTrials.gov identifier: NCT04649476

Collapse
Poster Display (ID 17) Poster Display

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 333)

Presentation Number
74P
Lecture Time
12:30 - 12:30
Speakers
  • L. Liu (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Fan (Shanghai, China)
  • X. Jin (Shanghai, China)
  • Y. Xu (Shanghai, China)
  • S. Wu (Shanghai, China)
  • Y. Yang (Shanghai, China)
  • L. Chen (Shanghai, China)
  • W. Zhang (Shanghai, China)
  • L. Ma (Shanghai, China)
  • X. Hu (Shanghai, China)
  • Z. Wang (Shanghai, China)
  • Y. Jiang (Shanghai, China)
  • Z. Shao (Shanghai, China)

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

Collapse
Poster Display (ID 17) Poster Display

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 473)

Presentation Number
75P
Lecture Time
12:30 - 12:30
Speakers
  • N. Girard (Paris, France)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Popat (London, United Kingdom)
  • S. Shoshkova (Sofia, Bulgaria)
  • S. Fear (Basel, Switzerland)
  • J. Perez Gracia (Pamplona, Spain)

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

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.

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.

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.

Clinical trial identification

NCT03782207

Editorial acknowledgement

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

Collapse
Poster Display (ID 17) Poster Display

76P - Safety profile of adjuvant pembrolizumab (pembro) in melanoma, non–small cell lung cancer (NSCLC), and renal cell carcinoma (RCC): pooled analysis of phase 3 clinical trials (ID 703)

Presentation Number
76P
Lecture Time
12:30 - 12:30
Speakers
  • J. Luke (Pittsburgh, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • G. Long (Sydney, Australia)
  • C. Robert (Villejuif, Cedex, France)
  • M. Carlino (Westmead, NSW, Australia)
  • T. Choueiri (Boston, United States of America)
  • N. Haas (Philadelphia, United States of America)
  • M. O'Brien (London, United Kingdom)
  • L. Paz-Ares (Madrid, Spain)
  • S. Peters (Lausanne, Switzerland)
  • T. Powles (London, United Kingdom)
  • M. Leiby (Hoddesdon, PA, United Kingdom)
  • J. Lin (Rahway, United States of America)
  • Y. Zhao (Lebanon, United States of America)
  • C. Krepler (North Wales, United States of America)
  • R. Perini (Whitehouse Station, United States of America)
  • M. Pietanza (Whitehouse Station, NJ, United States of America)
  • A. Samkari (North Wales, United States of America)
  • T. Gruber (Lebanon, United States of America)
  • N. Ibrahim (Whitehouse Station, United States of America)
  • A. Eggermont (Utrecht, Netherlands)

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.

Collapse
Poster Display (ID 17) Poster Display

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 118)

Presentation Number
77P
Lecture Time
12:30 - 12:30
Speakers
  • Y. Cheng (Changchun, Jilin, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Y. Cheng (Changchun, Jilin, China)
  • B. Liu (Harbin, China)
  • L. Wu (Changsha, China)
  • T. Zhang (Beijing, China)
  • K. Wang (Chengdu, Sichuan, China)
  • L. Miao (Nanjing, China)
  • J. Wu (Xiamen, China)
  • H. Wang (Zhengzhou, China)
  • J. Fang (Beijing, China)
  • X. Li (Zhengzhou, China)
  • W. Zhang (Nanchang, China)
  • Y. Hu (Wuhan, China)
  • X. Mei (Hefei, China)
  • Y. Song (Nanjing, China)
  • C. Wu (Changchun, China)
  • J. Zhu (Changchun, Jilin, China)
  • Y. Xin (Changchun, China)
  • Y. Chen (Beijing, China)
  • G. Han (Beijing, China)
  • J. Ye (Beijing, China)

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

Collapse
Poster Display (ID 17) Poster Display

78P - Clinical outcomes with atezolizumab plus bevacizumab (AB) or lenvatinib (L) in hepatocellular carcinoma (HCC). (ID 515)

Presentation Number
78P
Lecture Time
12:30 - 12:30
Speakers
  • M. Persano (Cagliari, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Persano (Cagliari, Italy)
  • M. Rimini (Modena, Italy)
  • S. Cascinu (Modena, Italy)
  • M. Scartozzi (Monserrato, Italy)
  • A. Casadei Gardini (Milan, Forlì-Cesena, Italy)

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.

Collapse
Poster Display (ID 17) Poster Display

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 121)

Presentation Number
79P
Lecture Time
12:30 - 12:30
Speakers
  • X. Wu (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • X. Wu (Shanghai, China)
  • L. Xia (Shanghai, China)
  • J. Wang (Changsha, China)
  • C. Wang (Shenyang, China)
  • Q. Zhang (Lanzhou, China)
  • J. Zhu (Hangzhou, China)
  • Q. Rao (Guangzhou, China)
  • H. Cheng (Zhengzhou, China)
  • Z. Liu (Handan, China)
  • Y. Yin (Nanjing, Jiangsu Province, China)
  • X. Ai (Hengyang, China)
  • K. Gulina (Urumqi, Xinjiang, China)
  • H. Zheng (Beijing, China)
  • X. Luo (Luoyang, China)
  • B. Chang (Luoyang, China)
  • L. Li (Nanning, China)
  • H. Liu (Taian, China)
  • Y. Li (Yinchuan, China)
  • J. Zhu (Beijing, China)

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 (ClinicalTrials.gov identifier: 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-lineplatinumcontaining standard chemotherapy.

Methods

In this single-arm, phase II study, eligible patients in 27 Chinese sites were assigned to receive intravenouszimberelimab 240 mg as monotherapy every 2 weeks until confirmed disease progression, death, intolerableadverse 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. Completeand 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 notreached 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

Collapse
Poster Display (ID 17) Poster Display

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 367)

Presentation Number
80P
Lecture Time
12:30 - 12:30
Speakers
  • C. Han (Beijing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Han (Beijing, China)
  • S. Ye (Beijing, China)
  • C. Hu (Changsha, Hunan, China)
  • L. Shen (Changsha, China)
  • Q. Qin (Changsha, China)
  • Y. Bai (Harbin, China)
  • S. Yang (Beijing, China)
  • C. Bai (Beijing, CN, China)
  • A. Zang (Baoding, China)
  • S. Jiao (Beijing, China)
  • L. Bai (Beijing, China)

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

The trial protocol number was NCT04172571 and release date was November 21, 2019.

Collapse
Poster Display (ID 17) Poster Display

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 508)

Presentation Number
81P
Lecture Time
12:30 - 12:30
Speakers
  • Y. Li (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Y. Li (Shanghai, China)
  • H. Jiang (Shanghai, China)
  • F. Qian (Shanghai, China)
  • Y. Cheng (Shanghai, China)
  • Y. Zhang (Shanghai, China)
  • J. Lu (Shanghai, China)
  • Y. Lou (Shanghai, China)
  • B. Han (Shanghai, China)
  • W. Zhang (Shanghai, China)

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.

Collapse
Poster Display (ID 17) Poster Display

82P - Preliminary analysis of tislelizumab (TIS) and chemotherapy as neoadjuvant therapy for potentially resectable stage IIIA/IIIB non-small cell lung cancer (NSCLC) (ID 577)

Presentation Number
82P
Lecture Time
12:30 - 12:30
Speakers
  • T. Wang (Chengdu, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • T. Wang (Chengdu, China)
  • L. Li (Chengdu, China)
  • L. Huang (Chengdu, China)
  • J. Liu (Chengdu, China)
  • D. Zhu (Chengdu, China)
  • C. Qin (Chengdu, China)
  • J. Dong (Chengdu, China)
  • H. Deng (Chengdu, China)
  • X. Zheng (Chengdu, China)
  • L. Tian (Chengdu, China)
  • L. Jiang (Chengdu, China)
  • W. Wang (Chengdu, China)
  • P. Zhou (Chengdu, China)
  • M. Su (Chengdu, China)
  • D. Pu (Chengdu, China)
  • X. Cai (Chengdu, China)
  • M. Feng (Chengdu, China)
  • X. Ou (Chengdu, China)
  • Y. Wang (Chengdu, China)
  • Q. Zhou (Chengdu, Sichuan, China)

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 1). 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%).

Table 1

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

Clinical trial information: NCT04865705

Editorial acknowledgement

Acknowledgments: We would like to thank Chunlu Shu from Medical Affairs, BeiGene, Ltd.

Collapse
Poster Display (ID 17) Poster Display

83P - The Immune-related adverse event (IRAE) Likelihood Score (ILS) identifies “pure” IRAEs strongly associated with outcome in a phase 1-2 trial population (ID 291)

Presentation Number
83P
Lecture Time
12:30 - 12:30
Speakers
  • L. Mazzarella (Milan, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Mazzarella (Milan, Italy)
  • E. Nicolo (Milan, Italy)
  • A. Esposito (Bristol, United Kingdom)
  • E. Crimini (Milan, Italy)
  • G. Tini (Milan, Italy)
  • J. Uliano (Milan, Italy)
  • C. Corti (Milan, Italy)
  • P. Trillo Aliaga (Milan, Italy)
  • C. Valenza (Milan, Italy)
  • M. Repetto (Milan, Italy)
  • G. Antonarelli (Milan, Italy)
  • I. Minchella (Milan, Italy)
  • C. Belli (Milan, Italy)
  • M. Locatelli (Milan, Italy)
  • C. Criscitiello (Milan, Italy)
  • G. Curigliano (Milan, Italy)

Abstract

Background

Immune-related Adverse Events (IRAE) pose a significant diagnostic and therapeutic challenge in patients treated with immune-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 immune-oncology (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 siggnificant 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.

Collapse
Poster Display (ID 17) Poster Display

84P - Ambulatory management of ICI-induced hepatitis: a safe and effective management approach (ID 742)

Presentation Number
84P
Lecture Time
12:30 - 12:30
Speakers
  • A. Olsson-Brown (Liverpool, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Olsson-Brown (Liverpool, United Kingdom)
  • N. Garbutt (Liverpool, United Kingdom)
  • F. Dulson (Liverpool, United Kingdom)
  • T. Guinan (Liverpool, United Kingdom)

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.

Collapse
Poster Display (ID 17) Poster Display

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 394)

Presentation Number
85P
Lecture Time
12:30 - 12:30
Speakers
  • L. Chen (Fuzhou, Fujian, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Chen (Fuzhou, Fujian, China)
  • Z. Ye (Fuzhou, China)
  • G. Liu (Putian, China)
  • Q. Lin (Sanming, China)
  • Y. Chi (Sanming, China)
  • J. Wang (Fuzhou, China)
  • S. Wei (Fuzhou, China)
  • C. Wei (Fuzhou, China)
  • S. Liu (Fuzhou, China)
  • Y. Zeng (Fuzhou, China)
  • S. Chen (Fuzhou, China)
  • Y. Wang (Fuzhou, China)

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

Collapse
Poster Display (ID 17) Poster Display

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 461)

Presentation Number
86P
Lecture Time
12:30 - 12:30
Speakers
  • C. Zhou (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Zhou (Shanghai, China)
  • N. Xu (Hangzhou, Zhejiang, China)
  • A. Xiong (Shanghai, China)
  • W. Li (Shanghai, China)
  • L. Wang (Shanghai, China)
  • F. Wu (Shanghai, China)
  • J. Yu (Shanghai, China)
  • C. Mao (Hangzhou, China)
  • J. Qian (Hangzhou, China)
  • Y. Zheng (Hangzhou, China)
  • H. Jiang (Hangzhou, China)
  • Y. Gao (Hangzhou, China)
  • C. Xiao (Hangzhou, China)
  • W. Wang (Changsha, China)
  • W. Zhuang (Fuzhou, China)
  • J. Yang (Guangzhou, China)
  • J. Sun (Beijing, China)
  • H. Wang (Beijing, China)
  • Y. Chen (Beijing, China)

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

Collapse
Poster Display (ID 17) Poster Display

87P - Infliximab use in patients with checkpoint inhibitor toxicities - a tertiary centre experience (ID 523)

Presentation Number
87P
Lecture Time
12:30 - 12:30
Speakers
  • S. Zhang (London, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Zhang (London, United Kingdom)
  • E. Cartwright (London, United Kingdom)
  • S. Mullings (London, United Kingdom)
  • L. Ferro Lopez (London, United Kingdom)
  • D. Cunningham (Sutton, United Kingdom)
  • I. Chau (Sutton, Surrey, United Kingdom)
  • N. Starling (London, United Kingdom)
  • S. Popat (London, United Kingdom)
  • M. O'Brien (London, United Kingdom)
  • J. Bhosle (Sutton, Surrey, United Kingdom)
  • A. Minchom (London, Surrey, United Kingdom)
  • M. Davidson (London, United Kingdom)
  • N. Tokaca (London, United Kingdom)
  • S. Lalondrelle (London, United Kingdom)
  • L. Pickering (London, United Kingdom)
  • A. Furness (London, London, United Kingdom)
  • S. Turajlic (London, United Kingdom)
  • J. Larkin (London, United Kingdom)
  • R. José (London, United Kingdom)
  • K. Young (London, United Kingdom)

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.

Collapse
Poster Display (ID 17) Poster Display

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 557)

Presentation Number
88P
Lecture Time
12:30 - 12:30
Speakers
  • W. Guo (guangzhou, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • W. Guo (guangzhou, China)
  • S. Chen (guangzhou, China)

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.

Collapse
Poster Display (ID 17) Poster Display

89P - Durvalumab could be effective in combination with anti-HER2 agents in HER2-low breast cancer (ID 625)

Presentation Number
89P
Lecture Time
12:30 - 12:30
Speakers
  • M. Arshad (London, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Arshad (London, United Kingdom)

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.

Collapse
Poster Display (ID 17) Poster Display

90P - Long-term outcomes in melanoma patients achieving a complete response under immune checkpoint inhibition (ID 696)

Presentation Number
90P
Lecture Time
12:30 - 12:30
Speakers
  • E. Chatziioannou (Tuebingen, Germany)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • E. Chatziioannou (Tuebingen, Germany)
  • U. Leiter-Stoppke (Tuebingen, Germany)
  • I. Thomas (Tuebingen, Germany)
  • U. Keim (Tuebingen, Germany)
  • A. Forschner (Tuebingen, Germany)
  • L. Flatz (Tuebingen, Germany)
  • T. Amaral (Tübingen, PT, Germany)

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.

Collapse
Poster Display (ID 17) Poster Display

91P - Association between weight variation and survival in patients treated with immune checkpoint inhibitors (ID 687)

Presentation Number
91P
Lecture Time
12:30 - 12:30
Speakers
  • R. Romao (Porto, Portugal)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • R. Romao (Porto, Portugal)
  • A. Mendes (Porto, Portugal)
  • R. Ranchor (Porto, Portugal)
  • M. Ramos (Porto, Portugal)
  • R. Pichel (Porto, Portugal)
  • J. Coelho (Porto, Portugal)
  • S. Azevedo (Porto, Portugal)
  • P. Fidalgo (Porto, Portugal)
  • A. Ferreira Araujo (Porto, Portugal)

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.

Collapse
Poster Display (ID 17) Poster Display

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 251)

Presentation Number
92P
Lecture Time
12:30 - 12:30
Speakers
  • X. Wang (Beijing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • X. Wang (Beijing, China)
  • X. Chen (Beijing, China)
  • X. Kang (Beijing, China)
  • R. Zhang (Beijing, China)
  • D. Qu (Beijing, China)
  • L. Xue (Beijing, China)
  • G. Cheng (Beijing, China)
  • G. Xi (Beijing, China)
  • T. Zhang (Beijing, China)
  • L. Deng (Beijing, China)
  • W. Liu (Beijing, China)
  • N. Bi (Beijing, China)
  • Y. Li (Beijing, China)

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

Collapse
Poster Display (ID 17) Poster Display

93P - Cetuximab-based chemotherapy after progression on immune-checkpoint inhibitors in recurrent/metastatic head and neck squamous cell cancer (ID 506)

Presentation Number
93P
Lecture Time
12:30 - 12:30
Speakers
  • S. Cabezas-Camarero (Madrid, Spain)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Cabezas-Camarero (Madrid, Spain)
  • M. Sotelo Lezama (Lima, Peru)
  • S. Merino-Menéndez (Madrid, Spain)
  • M. Cabrera-Martín (Madrid, Spain)
  • P. Pérez Segura (Madrid, Spain)

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 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 m (0-71), 18 m (0-71) and 11 m (0-63), respectively. Median overall survival (OS) since first-line was 23 m (95%CI: 18.8-27.2) since the start of ICI was 19 m (95%CI 14-24) and since the start of PC was 12 m (95%CI 8.8-15.1). Median OS with PC in first-line (n=21, OS= 12 m (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 m (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.

Collapse
Poster Display (ID 17) Poster Display

94P - Does hyperprogressive disease predict poor survival? (ID 702)

Presentation Number
94P
Lecture Time
12:30 - 12:30
Speakers
  • G. Alkan (Istanbul, Turkey)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • G. Alkan (Istanbul, Turkey)
  • N. Senturk Oztas (Istanbul, Turkey)
  • E. Degerli (Istanbul, Turkey)
  • Z. Turna (Istanbul, Turkey)
  • M. Ozguroglu (Istanbul, Turkey)

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.

Collapse
Poster Display (ID 17) Poster Display

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 168)

Presentation Number
95P
Lecture Time
12:30 - 12:30
Speakers
  • J. Xie (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Xie (Shanghai, China)
  • L. Yi (Shanghai, China)
  • Z. Meng (Shanghai, China)

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)

Collapse
Poster Display (ID 17) Poster Display

96P - Long-term outcomes after initial disease progression with anti-PD-1 in melanoma (ID 173)

Presentation Number
96P
Lecture Time
12:30 - 12:30
Speakers
  • K. Loo (New York, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • K. Loo (New York, United States of America)
  • B. Park (Nashville, United States of America)
  • H. Kalvin (New York, United States of America)
  • K. Panageas (New York, United States of America)
  • D. Johnson (Nashville, United States of America)
  • A. Betof Warner (New York, United States of America)

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.

Collapse
Poster Display (ID 17) Poster Display

97P - Toxicity of bispecific antibody glofitamab in patients with aggressive B-cell non-Hodgkin lymphoma in real clinical practice (ID 211)

Presentation Number
97P
Lecture Time
12:30 - 12:30
Speakers
  • L. Fedorova (Saint-Petersburg, Russian Federation)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Fedorova (Saint-Petersburg, Russian Federation)
  • O. Smykova (St. Petersburg, Russian Federation)
  • A. Chekalov (Saint-Petersburg, Russian Federation)
  • E. Lepik (Saint-Petersburg, Russian Federation)
  • V. Markelov (St. Petersburg, Russian Federation)
  • M. Popova (Saint-Petersburg, Russian Federation)
  • E. Kondakova (St. Petersburg, Russian Federation)
  • I. Moiseev (St. Petersburg, Russian Federation)
  • K. Lepik (St. Petersburg, Russian Federation)
  • N. Mikhailova (St. Petersburg, Russian Federation)
  • A. Kulagin (St. Petersburg, Russian Federation)

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 1). 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.

Collapse
Poster Display (ID 17) Poster Display

98P - First-line atezolizumab/durvalumab plus platinum–etoposide combined with radiotherapy in extensive-stage small-cell lung cancer (ID 271)

Presentation Number
98P
Lecture Time
12:30 - 12:30
Speakers
  • A. Shi (Beijing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Shi (Beijing, China)
  • L. Li (Beijing, China)
  • D. Yang (Beijing, China)
  • Y. Min (Beijing, China)
  • L. Jiang (Beijing, China)
  • X. Dong (Beijing, China)
  • W. Deng (Beijing, China)
  • H. Yu (Beijing, China)
  • R. Yu (Beijing, China)
  • J. Zhao (Beijing, China)

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.

Collapse
Poster Display (ID 17) Poster Display

99P - Sintilimab plus nab-paclitaxel in platinum-refractory head and neck squamous cell carcinoma: a phase 2 trial (ID 311)

Presentation Number
99P
Lecture Time
12:30 - 12:30
Speakers
  • S. Zhou (Beijing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Zhou (Beijing, China)
  • L. Zuo (Beijing, China)
  • S. Yang (Beijing, China)
  • X. He (Beijing, China)
  • J. Yang (Beijing, China)
  • L. Gui (Beijing, China)
  • R. Li (Beijing, China)
  • Y. Yang (Beijing, China)

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

Collapse
Poster Display (ID 17) Poster Display

100P - Immunotherapy around the clock: impact on stage IV melanoma (ID 354)

Presentation Number
100P
Lecture Time
12:30 - 12:30
Speakers
  • L. Goncalves (Lisbon, Portugal)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Goncalves (Lisbon, Portugal)
  • D. Gonçalves (London, United Kingdom)
  • T. Esteban Casanelles (London, United Kingdom)
  • I. Soares de Pinho (Lisbon, Portugal)
  • T. Barroso (Lisbon, Portugal)
  • V. Patel (Lisbon, Portugal)
  • M. Esperanca-Martins (Lisboa, Portugal)
  • R. Brás (Lisbon, Portugal)
  • S. Lobo-Martins (Lisbon, Portugal)
  • P. Semedo (Lisboa, Portugal)
  • C. Moreira (Lisbon, Portugal)
  • A. Teixeira Sousa (Lisbon, Portugal)
  • A. Mansinho (Lisbon, Portugal)
  • L. Marques Da Costa (Lisbon, Portugal)

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.

Collapse
Poster Display (ID 17) Poster Display

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 366)

Presentation Number
101P
Lecture Time
12:30 - 12:30
Speakers
  • F. Yan (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • F. Yan (Shanghai, China)
  • M. Ying (Shanghai, China)
  • L. Chen (Shanghai, China)
  • Q. Fu (Shanghai, China)

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.

Collapse
Poster Display (ID 17) Poster Display

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 450)

Presentation Number
102P
Lecture Time
12:30 - 12:30
Speakers
  • G. Tan (Dalian, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • G. Tan (Dalian, China)
  • J. Lin (Shenyang, China)
  • F. Wen (Shenyang, China)
  • M. Chi (Dalian, China)
  • G. Yu (Shenyang, China)
  • Z. Lin (Dalian, China)
  • Z. Ning (Dalian, China)

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.

Collapse
Poster Display (ID 17) Poster Display

103P - Phase IIIb study of durvalumab plus platinum–etoposide in first-line treatment of Chinese extensive-stage small-cell lung cancer (ORIENTAL): preliminary safety and efficacy results (ID 464)

Presentation Number
103P
Lecture Time
12:30 - 12:30
Speakers
  • Y. Cheng (Changchun, Jilin, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Y. Cheng (Changchun, Jilin, China)
  • J. Wang (Beijing, China)
  • Y. Yu (Harbin, China)
  • A. Zang (Baoding, China)
  • D. Lv (Taizhou, China)
  • S. Li (Shanghai, China)
  • L. Cao (Hefei, China)
  • Z. Meng (Huai'an, China)
  • W. Mao (Jiangyin, China)
  • J. Zhang (Guangzhou, China)
  • A. Liu (Nanchang, China)
  • Y. Zhang (Nanjing, China)
  • K. Tang (Guangzhou, China)
  • J. Liu (Dalian, China)
  • J. Zheng (Wenzhou, China)
  • Z. Wang (Yangzhou, China)
  • E. Chen (Hangzhou, China)
  • X. Zhang (Qingdao, China)
  • Q. Guo (Jinan, Shandong, China)
  • D. Huang (Tianjin, China)

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

Collapse
Poster Display (ID 17) Poster Display

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 530)

Presentation Number
104P
Lecture Time
12:30 - 12:30
Speakers
  • Y. Zhao (Harbin, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Y. Zhao (Harbin, China)
  • X. Wang (Harbin, China)
  • M. Zhang (Harbin, China)
  • J. Liu (Harbin, China)
  • H. Pu (Harbin, China)
  • X. Li (Heilongjiang, China)
  • H. Zhao (Harbin, China)
  • S. Xu (Harbin, China)
  • M. Yang (Harbin, China)
  • S. Bai (Harbin, China)
  • L. Guo (Harbin, China)
  • L. Zhao (Harbin, China)
  • Y. Li (Harbin, China)
  • Y. Wang (Harbin, Heilongjiang, China)

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

Collapse
Poster Display (ID 17) Poster Display

105P - Neoadjuvant tislelizumab combined with chemoradiotherapy for resectable locally advanced esophageal squamous cell carcinoma (ESCC): single arm phase II study (ID 565)

Presentation Number
105P
Lecture Time
12:30 - 12:30
Speakers
  • P. Jin (Jinan, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • P. Jin (Jinan, China)
  • Y. Gao (Jinan, China)
  • Z. Fu (Jinan, China)
  • W. Yang (Jinan, China)
  • X. Meng (Jinan, China)

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

Collapse
Poster Display (ID 17) Poster Display

106P - Zanubrutinib in combination with tislelizumab in patients with refractory diffuse large B-cell lymphoma (DLBCL): a phase II study (ID 582)

Presentation Number
106P
Lecture Time
12:30 - 12:30
Speakers
  • L. Zuo (Beijing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Zhou (Beijing, China)
  • P. Liu (Beijing, China)
  • S. Yang (Beijing, China)
  • J. Yang (Beijing, China)
  • X. He (Beijing, China)
  • L. Gui (Beijing, China)
  • R. Li (Beijing, China)
  • Y. Yang (Beijing, China)

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.

Collapse
Poster Display (ID 17) Poster Display

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 481)

Presentation Number
107P
Lecture Time
12:30 - 12:30
Speakers
  • S. Peters (Lausanne, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Peters (Lausanne, Switzerland)
  • R. Salomonsen (Phoenix, United States of America)
  • R. Tattersfield (Luton, United Kingdom)
  • A. Wang (Phoenix, United States of America)
  • Y. Xiao (Waltham, United States of America)
  • L. Cai (South San Francisco, United States of America)
  • S. Sadow (Phoenix, United States of America)
  • R. Jassim (Phoenix, United States of America)
  • S. Liu (Washington, United States of America)

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

Dataset
Analytic

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

RCT
400

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

RCT
361

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

RCT
396

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.

Collapse
Poster Display (ID 17) Poster Display

108P - Role of the prognostic nutritional index in predicting survival in advanced hepatocellular carcinoma treated with atezolizumab plus bevacizumab. (ID 513)

Presentation Number
108P
Lecture Time
12:30 - 12:30
Speakers
  • M. Persano (Cagliari, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Persano (Cagliari, Italy)
  • M. Rimini (Modena, Italy)
  • S. Cascinu (Modena, Italy)
  • M. Scartozzi (Monserrato, Italy)
  • A. Casadei Gardini (Milan, Forlì-Cesena, Italy)

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.

Collapse
Poster Display (ID 17) Poster Display

109P - Impact of anti-drug antibody (ADA) on naxitamab efficacy and safety (ID 616)

Presentation Number
109P
Lecture Time
12:30 - 12:30
Speakers
  • B. Kushner (New York, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • B. Kushner (New York, United States of America)
  • G. Chan (Hong Kong, Hong Kong PRC)
  • D. Morgenstern (Toronto, Canada)
  • L. Amoroso (Genoa, Italy)
  • K. Nysom (Copenhagen, Denmark)
  • J. Faber (Mainz, Germany)
  • A. Wingerter (Mainz, Germany)
  • M. Bear (Indianapolis, United States of America)
  • A. Rubio San Simon (Madrid, Spain)
  • K. Tornøe (Hoersholm, Denmark)
  • P. Sørensen (Hoersholm, Denmark)
  • J. Mora (Esplugues de Llobregat, Spain)

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.

Table: 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

Collapse
Poster Display (ID 17) Poster Display

110P - Fecal Microbiota Transplantation in Combination with Fruquintinib and Tislelizumab in Refractory Microsatellite Stable Metastatic Colorectal Cancer: A single center phase II trial (ID 109)

Presentation Number
110P
Lecture Time
12:30 - 12:30
Speakers
  • W. Zhao (Wuhan, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • W. Zhao (Wuhan, China)
  • Y. Chen (Wuhan, China)

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

Collapse
Poster Display (ID 17) Poster Display

111P - Identification of Atezolizumab plus BEvacizumab prognostic index via recursive partitioning analysis in advanced hepatocellular carcinoma: the ABE index. (ID 511)

Presentation Number
111P
Lecture Time
12:30 - 12:30
Speakers
  • M. Persano (Cagliari, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Persano (Cagliari, Italy)
  • M. Rimini (Modena, Italy)
  • S. Cascinu (Modena, Italy)
  • M. Scartozzi (Monserrato, Italy)
  • A. Casadei Gardini (Milan, Forlì-Cesena, Italy)

Abstract

Background

This study aims 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.

Collapse
Poster Display (ID 17) Poster Display

112P - Pseudoprogression predicts better prognosis in advanced non small cell lung cancer treated with immune-check point inhibitors (ID 651)

Presentation Number
112P
Lecture Time
12:30 - 12:30
Speakers
  • N. Senturk Oztas (Istanbul, Turkey)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • N. Senturk Oztas (Istanbul, Turkey)
  • G. Alkan (Istanbul, Turkey)
  • E. Degerli (Istanbul, Turkey)
  • Z. Turna (Istanbul, Turkey)
  • M. Ozguroglu (Istanbul, Turkey)

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.

Table. 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.

Collapse
Poster Display (ID 17) Poster Display

113P - The prognostic and predictive roles of serum C-reactive protein and PD-L1 in non-small cell lung cancer (ID 370)

Presentation Number
113P
Lecture Time
12:30 - 12:30
Speakers
  • S. Kuusisalo (Oulu, Finland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Kuusisalo (Oulu, Finland)
  • A. Tikkanen (Oulu, Finland)
  • S. Iivanainen (Oulu, Finland)
  • J. Koivunen (Oulu, Finland)

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.

Collapse
Poster Display (ID 17) Poster Display

114P - Systemic Inflammatory Index as a prognostic biomarker in metastatic melanoma patients under immune checkpoint inhibitors (ID 162)

Presentation Number
114P
Lecture Time
12:30 - 12:30
Speakers
  • M. Rebordão Pires (Coimbra, Portugal)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Rebordão Pires (Coimbra, Portugal)
  • A. Caetano (Coimbra, Portugal)
  • C. Amorim Costa (Coimbra, Portugal)
  • J. Monteiro (Coimbra, Portugal)
  • R. Santos (Coimbra, Portugal)
  • R. Félix Soares (Coimbra, Portugal)
  • I. Piedade Domingues (Coimbra, Portugal)
  • E. Costa Jesus (Coimbra, Portugal)
  • G. Sousa (Coimbra, Portugal)

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.

Collapse
Poster Display (ID 17) Poster Display

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 416)

Presentation Number
115P
Lecture Time
12:30 - 12:30
Speakers
  • M. Slowinska (Warsaw, Poland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Slowinska (Warsaw, Poland)
  • A. Szumera-Cieckiewicz (Warsaw, PL, Poland)
  • I. Czarnecka (Warsaw, Poland)
  • M. Chelstowska (Warsaw, Poland)
  • A. Dawidowska (Warsaw, Poland)
  • S. Jaczewska (Warsaw, Poland)
  • P. Rutkowski (Warsaw, Poland)
  • I. Lugowska (Warsaw, Poland)

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

Collapse
Poster Display (ID 17) Poster Display

116P - Clinical Significance of Serum-derived Exosomal PD-L1 Expression in Patients with Advanced Pancreatic Cancer (ID 510)

Presentation Number
116P
Lecture Time
12:30 - 12:30
Speakers
  • S. Park (Seoul, Korea, Republic of)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Park (Seoul, Korea, Republic of)
  • I. Kim (Seoul, Korea, Republic of)
  • M. Lee (Seoul, Korea, Republic of)
  • K. Shin (Seoul, Korea, Republic of)
  • T. Hong (Seoul, Korea, Republic of)

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.

Collapse
Poster Display (ID 17) Poster Display

117P - Beta-Blockers in Lung Cancer Patients Receiving Immunotherapy (ID 624)

Presentation Number
117P
Lecture Time
12:30 - 12:30
Speakers
  • A. Duarte Mendes (Amadora, Portugal)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Duarte Mendes (Amadora, Portugal)
  • R. Ferreira (Amadora, Portugal)
  • T. Martins (Amadora, Portugal)
  • S. Prada (Amadora, Portugal)
  • M. Silva Sousa (Amadora, Lisboa, Portugal)

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.

Collapse
Poster Display (ID 17) Poster Display

118P - Immunosenescence and response to immunotherapy in elderly patients: A possible prognostic tool (ID 734)

Presentation Number
118P
Lecture Time
12:30 - 12:30
Speakers
  • A. Pretta (Cagliari, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Pretta (Cagliari, Italy)
  • C. Donisi (Cagliari, Italy)
  • P. Ziranu (Monserrato, Ca, Italy)
  • N. Liscia (Cagliari, Italy)
  • F. Loi (Monserrato, Italy)
  • G. Saba (Cagliari, Italy)
  • G. Pretta (Hove, United Kingdom)
  • D. Spanu (Cagliari, Italy)
  • M. Dubois (Monserrato, Italy)
  • F. Atzori (Monserrato, (CA), Italy)
  • V. Pusceddu (Monserrato, (CA), Italy)
  • M. Puzzoni (Monserrato, (CA), Italy)
  • E. Massa (Monserrato, (CA), Italy)
  • C. Madeddu (Monserrato, (CA), Italy)
  • E. Lai (Cagliari, Italy)
  • G. Astara (Monserrato, (CA), Italy)
  • M. Scartozzi (Monserrato, Italy)

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 an higher T-cells activity and a better response to immunotherapy. Further studies are needed to validate it.

Collapse
Poster Display (ID 17) Poster Display

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 432)

Presentation Number
119TiP
Lecture Time
12:30 - 12:30
Speakers
  • C. Gessner (Leipzig, Germany)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Gessner (Leipzig, Germany)
  • F. Griesinger (Oldenburg, Germany)
  • R. Shah (Maidstone, Kent, United Kingdom)
  • T. Talbot (Truro, Cornwall, United Kingdom)
  • S. Venkateshan (Tarrytown, United States of America)
  • A. Vadanahalli Shankar (Mumbai, India)
  • A. Seluzhytsky (Cambridge, United States of America)
  • D. Marsden (Oxford, United Kingdom)

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.

Collapse
Poster Display (ID 17) Poster Display

120TiP - Conversion of Response to Immune Checkpoint Inhibition by Fecal Microbiota Transplantation in Patients With Metastatic Melanoma: a Randomized Phase Ib/IIa Trial (ID 163)

Presentation Number
120TiP
Lecture Time
12:30 - 12:30
Speakers
  • J. Borgers (Amsterdam, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Borgers (Amsterdam, Netherlands)
  • F. Burgers (Amsterdam, Netherlands)
  • E. Terveer (Leiden, Netherlands)
  • M. Van Leerdam (Leiden, Netherlands)
  • T. Korse (Amsterdam, Netherlands)
  • R. Kessels (Amsterdam, Netherlands)
  • J. Henderickx (Leiden, Netherlands)
  • C. Flohil (Amsterdam, Netherlands)
  • M. Van Dijk (Amsterdam, Netherlands)
  • J. Keller (Leidschendam, Netherlands)
  • H. Verspaget (Leiden, Netherlands)
  • E. Kuijper (Leiden, Netherlands)
  • J. Haanen (Amsterdam, Netherlands)

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

Collapse
Poster Display (ID 17) Poster Display

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 528)

Presentation Number
121TiP
Lecture Time
12:30 - 12:30
Speakers
  • L. Qi (Beijing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Qi (Beijing, China)
  • X. Wang (Beijing, China)
  • Z. Wang (Bejing, China)
  • W. Liu (Shijiazhuang, China)
  • F. Li (Shijiazhuang, China)
  • X. Zhang (Shijiazhuang, China)
  • J. He (Bejing, China)
  • Y. Li (Bejing, China)

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

Collapse
Poster Display (ID 17) Poster Display

Therapeutic development (ID 804)

Lecture Time
12:30 - 12:30
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Poster Display (ID 17) Poster Display

123P - IMpower010: exploratory overall survival (OS) with adjuvant atezolizumab (atezo) vs best supportive care (BSC) in stage II-IIIA NSCLC with high PD-L1 expression (ID 479)

Presentation Number
123P
Lecture Time
12:30 - 12:30
Speakers
  • A. Rittmeyer (Immenhausen, Germany)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Rittmeyer (Immenhausen, Germany)
  • E. Felip (Barcelona, Spain)
  • N. Altorki (New York, United States of America)
  • E. Vallieres (Seattle, WA, United States of America)
  • C. Zhou (Shanghai, China)
  • A. Martinez-Marti (Barcelona, Spain)
  • T. Csoszi (Szolnok, Hungary)
  • M. Reck (Grosshansdorf, Germany)
  • M. Teixeira (Lisbon, Portugal)
  • Y. Deng (South San Francisco, United States of America)
  • M. Huang (South San Francisco, United States of America)
  • V. McNally (Welwyn Garden City, Herts, United Kingdom)
  • E. Bennett (South San Francisco, United States of America)
  • B. Gitlitz (South San Francisco, United States of America)
  • M. Srivastava (South San Francisco, United States of America)
  • H. Wakelee (Stanford, CA, United States of America)

Abstract

Background

IMpower010 (NCT02486718) showed a statistically significant benefit in primary-endpoint disease-free survival (DFS) with adjuvant atezo vs BSC in resected stage II-IIIA PD-L1 tumour cell (TC) ≥1% (SP263) NSCLC after platinum-based chemotherapy. This finding led to the approval of atezo in this setting in the US, China, Japan and other countries and to its approval for stage II-IIIA PD-L1 TC ≥50% NSCLC in the EU and other countries. At the first pre-specified interim analysis (IA) of OS (cutoff 18 Apr 2022), the secondary endpoint of OS in the ITT population was not mature, but OS is of clinical interest in this curative setting; in pts with stage II-IIIA and stage II-IIIA PD-L1 ≥1% NSCLC, the respective OS HRs were 0.95 (95% CI: 0.74, 1.24) and 0.71 (95% CI: 0.49, 1.03) (Wakelee JTO 2022; 17:S2). We present OS data in pts with stage II-IIIA PD-L1 TC ≥50% NSCLC.

Methods

The IMpower010 study design and primary endpoint analysis have been reported (Felip Lancet 2021). We report exploratory OS in pts with stage II-IIIA (UICC/AJCC v7) PD-L1 TC ≥50% (VENTANA SP263 assay) NSCLC and updated safety at this OS IA.

Results

In 229 pts with stage II-IIIA PD-L1 TC ≥50% NSCLC, the OS HR was 0.43 (95% CI: 0.24, 0.78); after excluding 20 pts with known EGFR/ALK+ status, the OS HR was 0.42 (95% CI: 0.23, 0.78). The OS subgroup analysis is presented in the Table. No new safety signals emerged since the previous cutoff.

Conclusions

Pts with stage II-IIIA PD-L1 TC ≥50% NSCLC derived OS benefit with atezo vs BSC. OS benefit was consistent across most pt subgroups, albeit with limited numbers in this exploratory analysis. The atezo safety profile remained unchanged. These data support the previously reported positive benefit-risk profile of adjuvant atezo in stage II-IIIA PD-L1+ resected NSCLC and contribute further evidence to support this new standard of care for patients with early-stage resectable NSCLC.

OS in stage II-IIIA PD-L1 TC ≥50% pt subgroups

Pts, n

Unstratified OS HR
(95% CI)

Atezo

115

BSC

114

Age

<65 y

≥65 y

70

45

68

46

0.44 (0.20, 0.97)

0.42 (0.17, 1.04)

Sex

Male

Female

89

26

78

36

0.39 (0.19, 0.80)

0.58 (0.20, 1.68)

ECOG PS

0

1

71

44

60

53

0.38 (0.16, 0.90)

0.51 (0.22, 1.19)

Histology

Squamous

Nonsquamous

47

68

45

69

0.58 (0.22, 1.51)

0.36 (0.17, 0.79)

Stage

II

IIIA

62

53

57

57

0.63 (0.28, 1.44)

0.30 (0.12, 0.74)

Lymph node status

N0

N1

N2

30

43

42

21

52

41

0.74 (0.21, 2.55)

0.38 (0.14, 1.07)

0.36 (0.14, 0.95)

EGFR/ALK mutation

Yes

No

Not tested

9

52

54

11

54

49

0.56 (0.05, 6.14)

0.37 (0.15, 0.89)

0.51 (0.21, 1.24)

Clinical trial identification

NCT02486718

Editorial acknowledgement

Medical writing support for this abstract was provided by Samantha Santangelo, PhD, of Health Interactions, Inc and funded by F. Hoffmann-La Roche, Ltd.

Collapse
Poster Display (ID 17) Poster Display

124P - Phase II Study of Neoadjuvant Concurrent Chemo-immuno-radiation Therapy Followed by Surgery and Adjuvant Immunotherapy for Resectable Stage IIIA-B N2 Non-Small Cell Lung Cancer: SQUAT trial (WJOG 12119L) (ID 507)

Presentation Number
124P
Lecture Time
12:30 - 12:30
Speakers
  • T. Miyoshi (Kashiwa, Japan)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • T. Miyoshi (Kashiwa, Japan)
  • A. Hamada (Yamagata, Japan)
  • J. Soh (Osaka, Japan)
  • A. Hata (Kobe, Hyogo, Japan)
  • K. Nakamatsu (Osaka, Japan)
  • M. Shimokawa (Ube, Japan)
  • Y. Yatabe (Chuo-ku, Aichi, Japan)
  • J. Suzuki (Yamagata, Japan)
  • M. Tsuboi (Kashiwa, Chiba, Japan)
  • H. Horinouchi (Chuo-ku, Japan)
  • I. Yoshino (Chiba, Japan)
  • T. Masayuki (Hamamatsu, Japan)
  • S. Toyooka (Okayama, Japan)
  • M. Okada (Hiroshima, Japan)
  • T. Go (Miki-cho, Japan)
  • M. Yamashita (Matsuyama, Japan)
  • Y. Nishimura (Osaka, Japan)
  • N. Yamamoto (Wakayama, Japan)
  • K. Nakagawa (Osaka, Japan)
  • T. Mitsudomi (Osaka, Japan)

Abstract

Background

Neoadjuvant chemo-immunotherapy (CheckMate 816 regimen) has become a standard of care in treatment of resectable non-small cell lung cancer (NSCLC) patients in some countries. Addition of radiation therapy (RT) may further improve local control in locally advanced NSCLC patients. Here, we report the primary endpoint, major pathologic response (MPR), of the SQUAT trial (WJOG 12119L, JapicCTI- 195069)-a multicenter, prospective, single-arm, phase II trial of neoadjuvant concurrent chemo-immuno-radiation therapy followed by surgical resection and adjuvant immunotherapy for resectable stage IIIA-B N2 NSCLC.

Methods

Patients with resectable stage IIIA-B N2 NSCLC received concurrent chemoradiotherapy [weekly paclitaxel (40 mg/m2)/carboplatin (AUC 2) for 5 weeks plus involved-field RT 50 Gy] and immunotherapy [durvalumab (1500 mg) Q4W for 2 cycles] followed by surgical resection and adjuvant immunotherapy (durvalumab for up to 1 year). The primary endpoint was MPR rate according to an independent central review (ICR). We assumed the threshold of the MPR rate to be 40% and the expected rate to be 65% with a significance level α=0.05 (one-sided) and power 0.8. The sample size was 31 patients, including the 10% ineligible patients.

Results

Between Jan 2020 and Feb 2022, 31 patients were enrolled from 10 institutions in Japan. Thirty-one patients were evaluated for safety, and 30 patients for efficacy. Of 30 patients (median age, 64 years), 70% and 63% had clinical stage IIIA and non-squamous histology, respectively. The MPR and pathological complete response (pCR) rates by ICR were 63% (90% CI, 47-78, 95% CI, 44-80) and 20% (95% CI, 8-39), respectively. Complete resection was not performed due to adverse events (2 pts) and disease progression (3 pts). Among 25 patients who received complete resection, the MPR and pCR rates were 76% (95% CI, 55-91) and 24% (95% CI, 9-45), respectively. No 30-day postoperative mortality was reported.

Conclusions

The primary endpoint of MPR rate was met in SQUAT trial. This result suggests that this treatment strategy is promising for resectable stage IIIA-B N2 NSCLC.

Clinical trial identification

JapicCTI-195069

Collapse
Poster Display (ID 17) Poster Display

125P - A phase 2a study of the novel immunogenic cell death (ICD) inducer PT-112 plus avelumab (“PAVE”) in advanced non-small cell lung cancer (NSCLC) patients (pts) (ID 209)

Presentation Number
125P
Lecture Time
12:30 - 12:30
Speakers
  • M. Imbimbo (Lausanne, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Imbimbo (Lausanne, Switzerland)
  • E. Ghisoni (Epalinges, (TO), Switzerland)
  • A. Mulvey (Lausanne, WA, Switzerland)
  • H. Bouchaab (Lausanne, Switzerland)
  • N. Mederos Alfonso (Lausanne, Vaud, Switzerland)
  • D. Karp (Houston, United States of America)
  • D. Camidge (Aurora, United States of America)
  • A. Mansfield (Rochester, MN, United States of America)
  • C. Yim (New York, United States of America)
  • T. Ames (New York, United States of America)
  • M. Price (New York, United States of America)
  • J. Baeck (Cambridge, United States of America)
  • J. O'Donnell (New York, United States of America)
  • S. Peters (Lausanne, Switzerland)

Abstract

Background

PT-112 is an immunogenic small molecule with reported clinical activity. In preclinical models, it induces ICD and synergizes with immune checkpoint inhibitors. Monotherapy and avelumab combination dose escalation studies have shown PT-112 to be well-tolerated. The recommended phase 2 dose was established at 360 mg/m2 (days 1, 8 and 15) when combined with 800 mg of avelumab (days 1 and 15) over a 28-day cycle. Here, we present safety and efficacy findings from the Phase 2a PAVE cohort in metastatic NSCLC.

Methods

NSCLC pts were eligible following no more than 4 prior lines of therapy (requiring prior anti-PD-(L)1 and platinum treatment). Correlative sampling included baseline tumor biopsy profiling and blood T cell receptor sequencing.

Results

Eighteen pts were enrolled. Common treatment-related adverse events (TRAEs) were anemia (50%), fatigue (50%), thrombocytopenia (44%), nausea (44%) and anorexia (44%). 72% of pts had ≥1 grade 3-4 TRAEs. Treatment-related peripheral neuropathy was noted in 4 pts (22%). There were 5 cases (28%) of infusion-related reactions, which led to the discontinuation of avelumab in two. Of the 15 pts evaluable for efficacy, 6 pts (40%) had stable disease by iRECIST. Two cases of radiographic and clinical improvement were noted: one pt with primary resistance to prior therapy (nivolumab, ipilimumab, carboplatin and gemcitabine) had a complete response in target and non-target lesions and relief of pulmonary symptoms before experiencing single nodal progression; another pt experienced a multi-site FDG-PET response and remained stable with a PFS of 7.3 months. Both pts had PD-L1 TPS of 5% and 10%, respectively; MSS; and TMB <10. Furthermore, the study population experienced a significant increase in the fraction of T cells within total nucleated blood cells while on treatment.

Conclusions

PT-112 in combination with avelumab was safe and AEs were manageable. Evidence of drug activity was observed, including meaningful clinical benefit in pretreated patients without known predictive markers for immunotherapy response. These results support further study of PT-112 combinations in NSCLC, and assessment of PT-112’s immunological effects in pts.

Clinical trial identification

NCT03409458

Collapse
Poster Display (ID 17) Poster Display

126P - AdvanTIG-105: Phase 1b Dose-Expansion Study of Ociperlimab (OCI) + Tislelizumab (TIS) in Patients (pts) With Checkpoint Inhibitor (CPI)-Experienced Advanced Non-Small Cell Lung cancer (NSCLC) (ID 560)

Presentation Number
126P
Lecture Time
12:30 - 12:30
Speakers
  • S. Frentzas (Bentleigh East, Australia)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Frentzas (Bentleigh East, Australia)
  • T. Meniawy (Subiaco, WA, Australia)
  • S. Kao (Camperdown, NSW, Australia)
  • J. Coward (Brisbane, Australia)
  • T. Clay (Subiaco, Australia)
  • N. Singhal (Adelaide, Australia)
  • A. Black (Hobart, Australia)
  • W. Xu (Woolloongabba, Australia)
  • R. Kumar (Auckland, New Zealand)
  • Y. Lee (Goyang, Korea, Republic of)
  • G. Lee (Jinju, Korea, Republic of)
  • W. Liao (Seoul, Korea, Republic of)
  • D. Zhong (Tianjin, Tianjin City, China)
  • H. Shiah (New Taipei City, Taiwan)
  • Y. Chen (Taipei City, Taiwan)
  • R. Gao (Shanghai, China)
  • R. Wang (Shanghai, Not Applicable, China)
  • H. Zheng (Emeryville, United States of America)
  • W. Tan (Shanghai, Not Applicable, China)
  • E. Cho (Incheon, Korea, Republic of)

Abstract

Background

T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) inhibitor with an anti-programmed cell death protein 1 (PD-1) antibody is a promising combination showing antitumor activity in solid tumors. Phase 1/1b open-label study AdvanTIG-105 assessed safety and preliminary antitumor activity of anti-TIGIT monoclonal antibody (mAb) OCI + anti-PD-1 mAb TIS in pts with advanced solid tumors (NCT04047862). During dose-escalation, OCI + TIS was well tolerated showing antitumor activity, establishing the recommended phase 2 dose (RP2D) of OCI 900mg IV Q3W plus TIS 200mg IV Q3W. We report Cohort 5 dose-expansion results.

Methods

Eligible adults had histologically/cytologically confirmed locally advanced/metastatic CPI-experienced NSCLC for which they received ≤2 prior therapies, including anti-PD-(L)1 in the most recent line, and progressed after complete or partial response (CR or PR) or stable disease. Pts received RP2D OCI + TIS until disease progression, intolerable toxicity or withdrawal of consent. Primary endpoint was investigator-assessed objective response rate (ORR) per RECIST v1.1. Secondary endpoints included disease control rate (DCR), duration of response (DOR) and safety.

Results

As of June 20, 2022, 26 pts were enrolled; 25 were efficacy evaluable. Median study follow-up was 46.1 weeks (range 25.4-59.0). The confirmed ORR was 8.0% (95% confidence interval [CI]: 1.0, 26.0), with two pts experiencing PR, and the confirmed DCR was 56.0% (95% CI: 34.9, 75.6); median DOR was not reached. Overall, 23 pts (88.5%) experienced ≥1 treatment-emergent adverse event (TEAE); 11 pts (42.3%) experienced Grade ≥3 TEAEs and nine pts (34.6%) experienced serious TEAEs. The most common TEAEs were fatigue (30.8%) and cough (26.9%). TEAEs leading to treatment discontinuation occurred in four pts (15.4%), and were related to treatment in two patients, with no TEAEs leading to death.

Conclusions

OCI 900mg + TIS 200mg was generally well tolerated and showed preliminary antitumor activity in pts with locally advanced/metastatic CPI-experienced NSCLC.

Clinical trial identification

NCT04047862

Editorial acknowledgement

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

Collapse
Poster Display (ID 17) Poster Display

127P - Phase 1 study of fianlimab, a human lymphocyte activation gene-3 (LAG-3) monoclonal antibody, in combination with cemiplimab in advanced NSCLC (ID 609)

Presentation Number
127P
Lecture Time
12:30 - 12:30
Speakers
  • B. Cho (Seoul, Korea, Republic of)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • B. Cho (Seoul, Korea, Republic of)
  • G. Dy (Buffalo, United States of America)
  • T. Kim (Seoul, Korea, Republic of)
  • D. Sarker (London, United Kingdom)
  • O. Hamid (Los Angeles, CA, United States of America)
  • S. Williamson (Kansas City, United States of America)
  • K. Sang-We (Seoul, Korea, Republic of)
  • H. Hatim (San Diego, United States of America)
  • S. Chen (Tarrytown, United States of America)
  • J. Mani (Tarrytown, United States of America)
  • V. Jankovic (Tarrytown, United States of America)
  • A. Paccaly (Tarrytown, United States of America)
  • S. Masinde (Sleepy Hollow, United States of America)
  • I. Lowy (Tarrytown, United States of America)
  • L. Brennan (Sleepy Hollow, United States of America)
  • G. Gullo (Tarrytown, United States of America)

Abstract

Background

Concurrent blockade of LAG-3 may enhance efficacy of anti–programmed cell death-1 (anti–PD-1) therapies. We present safety and clinical activity data from a Phase 1 study in patients (pts) with NSCLC treated with anti–LAG-3 (fianlimab) + anti–PD-1 (cemiplimab).

Methods

Pts with unresectable stage IIIB–C or IV NSCLC who were anti–PD-1/PD-L1-naive (expansion cohort [EC] 1) or anti–PD-1/L1-experienced within 3 months (mo) of screening (EC2) were eligible. Pts received fianlimab 1600 mg + cemi 350 mg intravenously every 3 weeks (wks) for 12 mo.

Results

As of the 1 July 2022 data cutoff, 15 EC1 and 15 EC2 pts were treated. For EC1 and EC2 respectively, median age was 70.0 and 67.0 years, 73.3% and 73.3% were male, and 33.3% and 73.3% were White. 40.0% of pts in EC1 were systemic treatment-naive. In EC2, 53.3% of pts had at least two lines of therapy including anti–PD1/L1 therapy.

Median treatment duration was 12.1 wks and median follow-up was 8.5 mo and 5.4 mo for EC1 and EC2 respectively. Grade ≥3 treatment-emergent adverse events (TEAEs) and serious TEAEs occurred in 33.3% & 20.0% and 40.0% & 13.3% of EC1 and EC2 pts, respectively. One pt in EC1 and no pts in EC2 discontinued due to a TEAE. No treatment-related deaths were reported.

RECIST 1.1-based investigator-assessed objective response rate (ORR) was 26.7% (four partial responses [PRs]) in EC1 and 6.7% (one PR) in EC2. In EC1, the ORR for treatment-naive pts (n=6) was 50%; the ORR for pts with PD-L1 expression ≥50% (n=3) was 100%. Kaplan–Meier estimation of median progression-free survival was 2.6 mo (95% confidence interval [CI], 1.2–8.3) in EC1 and 4.1 mo (95% CI, 1.3–6.2) in EC2 pts. Median duration of response was not reached in EC1 and was 4.8 mo (95% CI, not evaluable [NE]–NE) in EC2. Fianlimab concentrations in serum in pts were similar in both EC1 and EC2 cohorts. LAG-3 and PD-L1 correlative biomarker analysis will be included in the presentation.

Conclusions

Fianlimab + cemiplimab demonstrated promising signs of clinical activity with durable responses among pts with anti–PD-1/L1-naive NSCLC and in pts with PD-L1 expression ≥50%, with a similar safety profile and with no new safety signals compared to cemiplimab monotherapy.

Clinical trial identification

NCT03005782

Editorial acknowledgement

Medical writing and editorial support provided by John G Facciponte, PhD, of Prime, Knutsford, UK, funded by Regeneron Pharmaceuticals, Inc., according to Good Publication Practice guidelines.

Collapse
Poster Display (ID 17) Poster Display

128P - Neoadjuvant durvalumab plus chemotherapy in stage III non-small cell lung cancer: A phase II single-center exploratory study (ID 561)

Presentation Number
128P
Lecture Time
12:30 - 12:30
Speakers
  • X. Dong (Wuhan, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • X. Dong (Wuhan, China)
  • F. Tong (Wuhan, China)
  • R. Zhang (Beijing, China)
  • B. Liang (Wuhan, China)
  • W. Zhai (Wuhan, China)
  • S. Wang (Wuhan, China)
  • J. Fan (Wuhan, China)
  • Y. Wang (Wuhan, China)
  • Y. Huang (Wuhan, China)

Abstract

Background

Stage III non-small cell lung cancer (NSCLC) is highly heterogeneous with great variations in clinical practice. Though neoadjuvant immunotherapy plus chemotherapy significantly improved pCR and EFS in resectable NSCLC patients compared with chemotherapy alone in previous study, the evidence in stage III NSCLC are limited. This is the first study to evaluate durvalumab neoadjuvant/adjuvant in stage III NSCLC patients.

Methods

A prospective phase II, single-arm study enrolled the patients (according to the MDT) with histologically confirmed stage IIIa-IIIc NSCLC without known EGFR/ALK mutations. Patients received neoadjuvant durvalumab (1500mg) plus platinum-based chemotherapy q3w for 2-4 cycles followed by surgery, then adjuvant durvalumab mono q4w for 12 cycles. The primary endpoint was MPR (≤10% viable tumor cells). Secondary endpoints included pCR ( 0% viable tumor cells), ORR, DFS, OS, and safety. Predictive biomarkers was exploratory endpoint.

Results

From February 7, 2021 to May 30, 2022, a total of 14 patients were enrolled with median follow-up of 9.5 months. The median age was 64.5 and 71.4% were squamous carcinoma histology. The number of patients with stage IIIa, IIIb, IIIc were 2 (14.3%), 10 (71.4%) and 2 (14.3%), respectively. All patients completed neoadjuvant therapy, 6 patients received 3 cycles and 8 patients received 4 cycles. Currently, the ORR was 64.3% (9/14), 10 patients underwent surgery, 4 patients were ineligible for surgery due to 2 with unresectable stage IIIc disease, 1 with tumor wrapping around the right bronchus and 1 with poor lung function. Among 10 resected patients, 50.0% achieved MPR and 20.0% achieved pCR. The TCR clone counts after 1 cycle neoadjuvant therapy was positively corelated with imaging regressions (p=0.044). The median DFS was not reached. Grade 3 or 4 treatment-related adverse events rate was 14.3%.

Conclusions

The results suggest that stage IIIa-IIIb NSCLC can benefit from neoadjuvant Durvalumab plus chemotherapy. Two stage IIIc patients failed to convert from neoadjuvant therapy. TCR diversity is positively correlated with imaging regression, analysis of its correlation with survival outcomes is ongoing.

Clinical trial identification

NCT04897386

Collapse
Poster Display (ID 17) Poster Display

129P - Tislelizumab Combined with Bevacizumab Plus Nab-paclitaxel Single-Agent Chemotherapy for Advanced NSCLC After Resistance to EGFR TKIs (ID 460)

Presentation Number
129P
Lecture Time
12:30 - 12:30
Speakers
  • C. Zhou (Guangzhou, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Zhou (Guangzhou, China)
  • X. Xie (Guangzhou, China)
  • Y. Yang (Guangzhou, China)
  • X. Lin (Guangzhou, China)
  • Z. XIE (Guangzhou, China)
  • M. Zhou (Guangzhou, China)
  • Y. Qin (Guangzhou, China)
  • J. Zhang (Guangzhou, China)

Abstract

Background

IMpower150 subgroup analysis showed patients with EGFR mutated advanced NSCLC could achieve efficacy from ICI-combination therapy after resistance to EGFR TKIs, but the safety should be improved. A retrospective study from our center showed that ICI plus single-agent chemotherapy (chemo-reform1) might be a better choice. This study was designed to further explore the efficacy and safety of tislelizumab (Tis) plus bevacizumab (Beva) and nab-paclitaxel for advanced NSCLC failed to EGFR TKIs. Herein, the preliminary analysis for early safety and efficacy is reported.

Methods

This is a single-arm, phase II study (NCT04310943). Pts with EGFR mutations who had failed to prior EGFR-TKIs accepted Tis (200mg, d1) plus Beva (15mg/kg, d1) and nab-paclitaxel (100mg/m2, d1,8,15) Q3W for up to 4 cycles, followed by Tis plus Beva (7.5mg/kg) maintenance therapy. Primary endpoints are safety and 1-year PFS rate, and secondary endpoints include ORR and 1-year OS rate. We planned to enroll 24 pts.

Results

As of 26 August 2022, 14 pts were enrolled and treated. The median age was 57 (range: 30-70). Seven pts (50.0%) harbored EGFR exon 19del and 7 pts (50.0%) had exon 21 L858R at the initial diagnosis. Ten pts (71.4%) had progression on both 1st/2nd and 3rd EGFR-TKIs. Two pts (14.3%) had prior chemotherapy. The median follow-up time was 11.2 months. Among 14 pts, the safety was manageable: the incidence of ≥grade 3 TEAE and ≥grade 3 irAE were 35.7% and 14.3% respectively. Among 9 efficacy evaluable pts, the ORR was 66.7% (95%CI: 29.9-92.5), mDOR was 5.3 months (95% CI: 4.9-5.7), and DCR was 100.0%.

AEs, n (%)

N=14

Any TEAEs

14 (100)

TEAEs ≥10%

Alopecia

Decreased WBC

Anemia

Hyperglycemia

Mucositis oral

Pruritus

Hypoalbuminemia

Pneumonitis

Fatigue

Neutrophil count decreased

GGT increased

14(100.0)

6 (42.9)

6 (42.9)

6 (42.9)

3 (21.4)

3 (21.4)

2 (14.3)

2 (14.3)

2 (14.3)

2 (14.3)

2 (14.3)

Any irAEs

8 (57.1)

irAEs ≥10%

Hyperglycemia

Pruritus

Pneumonitis

6 (42.9)

3 (21.4)

2 (14.3)

Conclusions

Tislelizumab combined with bevacizumab plus nab-paclitaxel has shown a promising anti-tumor efficacy with a manageable safety profile for patients who failed to EGFR TKIs. The results will be monitored continuously.

Clinical trial identification

NCT04310943

Editorial acknowledgement

BeiGene

Collapse
Poster Display (ID 17) Poster Display

130P - COM701 ± Nivolumab – preliminary results of antitumor activity from a phase 1 study in patients with metastatic NSCLC who have received prior PD-1/PD-L1 inhibitor. (ID 306)

Presentation Number
130P
Lecture Time
12:30 - 12:30
Speakers
  • R. Sullivan (Boston, MA, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • R. Sullivan (Boston, MA, United States of America)
  • M. Sharma (Grand Rapids, United States of America)
  • D. Vaena (Germantown, United States of America)
  • E. Hamilton (Nashville, United States of America)
  • B. Izar (New York, United States of America)
  • D. Rasco (San Antonio, United States of America)
  • J. Gainor (Boston, United States of America)
  • D. Shepard (Cleveland, United States of America)
  • K. Papadopoulos (San Antonio, TX, United States of America)
  • E. Dumbrava (Houston, TX, United States of America)
  • H. Adewoye (South San Francisco, United States of America)
  • P. Ferre (Boulogne-Billancourt, France)
  • E. Ophir (Lausanne, Switzerland)
  • M. Patel (Fort Myers, United States of America)
  • A. Patnaik (San Antonio, United States of America)
  • B. Chmielowski (Los Angeles, United States of America)

Abstract

Background

Novel agents are urgently needed for the treatment [tx] of patients [pts] with metastatic NSCLC including post immune checkpoint inhibitors (ICI). COM701 [anti-PVRIG] is a novel 1st in class ICI. We hypothesized that COM701 ± nivolumab will demonstrate antitumor activity with a favorable safety and tolerability profile in pts with heavily pretreated NSCLC. Here, we present preliminary results on antitumor activity.

Methods

We enrolled 7 pts with NSCLC: 5 COM701 monotherapy [mtx] [4 - COM701 20 mg/kg IV Q4W mtx expansion, 1 pt COM701 0.01 mg/kg IV Q3W], 2 pts COM701 + nivolumab [COM701/nivolumab 3mg/kg/360 mg both IV Q3W, COM701/nivolumab 10 mg/kg/480 mg both IV Q4W]. Key study objectives were safety/tolerability and preliminary antitumor activity; OS was an exploratory endpoint. Key inclusion criteria: Age ≥ 18 yrs, histologically confirmed metastatic solid malignancy and has exhausted all standard tx. Key exclusion criteria: history of immune toxicities on prior ICI tx leading to discontinuation. Safety per CTCAE v4.03, investigator-assessed response per RECIST v1.1.

Results

Age >65 4/7 [57%], female 6/7 [86%], prior lines median [Min, Max] 4 [3, 6], all pts received prior ICI, 4/7 [57%] received ≥2 prior lines with ICI. Disease control rate [CR+PR+SD) 5/7 [71%], no CR or PR. Median [m] PFS [all pts] 84 days 95% CI [22, 231], mOS in all pts 9.5 months [mos] [95% CI, 2.7-11.6] – [COM701 mtx 9.5 mos [2.7, NE]; combination 10.1 mos [95% CI, 8.6, NE]. The most frequent AE was G1 nausea in 2 pts. Post ICI NSCLC data - 1 prior line of ICI in metastatic setting, mOS 14.5 mos for ramucirumab + pembrolizumab1.

Conclusions

COM701 ± nivolumab has preliminary encouraging signal of antitumor activity in a heavily pretreated popn of pts with NSCLC with prior ICI txp comparable to historical data. A P1 study in post IO NSCLC evaluating COM902+COM701+PD-1 inhibitor, COM902+COM701+chemotherapy is planned. Datacut 07/17/2022.

1. Reckamp KL, et al Phase 2 Randomized Study of Ramucirumab and Pembrolizumab vs SOC in Advanced NSCLC Previously Treated With Immunotherapy-Lung-MAP S1800A. J Clin Oncol. 2022 Jul 20;40(21):2295-2306

Clinical trial identification

NCT03667716.

Collapse
Poster Display (ID 17) Poster Display

131P - First-line HDACi plus Tislelizumab combined with chemotherapy in Advanced NSCLC (ID 403)

Presentation Number
131P
Lecture Time
12:30 - 12:30
Speakers
  • L. Wang (Beijing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Wang (Beijing, China)
  • M. Gao (Beijing, China)
  • F. Jing (Beijing, China)
  • J. Ma (Beijing, China)
  • F. Zhang (Beijing, China)
  • H. Tao (Beijing, China)
  • Y. Hu (Beijing, China)

Abstract

Background

Resistance to ICIs remains a challenge. HDAC inhibitors may synergize with PD-1 antibodies by inducing and activating NK cell and cytotoxic T cell (CTL) -mediated cellular immunity.Tislelizumab is an anti-PD-1 mAb approved for the treatment of NSCLC. Chidamide, a subtype-selective HDACi. This Phase 2 study assessed antitumor activity and tolerability of chidamide and tislelizumab combined with chemotherapy in advanced NSCLC.

Methods

This was an open-label, prospective study. Patients with histologically or cytologically confirmed NSCLC(stage IIIB-IV) without prior systemic treatment was enrolled. EGFR/ALK mutation or fusion and symptomatic brain metastases were ineligible. Patients received chidamide 20 mg twice weekly orally, tislelizumab 200 mg Q3W intravenously, chemotherapy Q3W(≤4~6 cycles) until unacceptable toxicity, withdrawal of consent, or death. The primary endpoint was ORR. Secondary endpoints include DCR、PFS and safety. Tumor response was assessed using RECIST V1.1.

Results

Table 1 Incidence of TRAEs

TRAEs (N=20)

Any Grade, n (%)

Grade ≥3, n(%)

Anemia

15(75.0)

2(10.0)

Alopecia

12(60.0)

0(0.0)

neutropenia

12(60.0)

4(20.0)

leukopenia

11(55.0)

5(25.0)

Nausea

9(45.0)

0(0.0)

Thrombocytopenia

9(45.0)

3(15.0)

Increased ALT

9(45.0)

0(0.0)

Increased AST

7(35.0)

0(0.0)

Decreased appetite

7(35.0)

0(0.0)

Fatigue

6(30.0)

0(0.0)

Increased creatinine

6(30.0)

0(0.0)

Blood lipids increased

5(25.0)

1(5.0)

Constipation

5(25.0)

0(0.0)

Diarrhea

5(25.0)

2(10.0)

pneumonia

4 (20.0)

1(5.0)

hypoesthesia

3(15.0)

0(0.0)

Adrenal insufficiency

3(15.0)

1(5.0)

Hypothyroidism

2(10.0)

0(0.0)

hyperglycemia

2(10.0)

1(5.0)

Dermatitis

1(5.0)

1(5.0)

20 patients were enrolled in the study. At the data cut-off September 15, 2022,the median age was 63.5 years (range: 49-75) and all patients were male 90% of who with smoking history. Most patients were stage IV(95%) .40% of the patients whose PD-L1 expression was positive (PD-L1 TPS≥1%) . The confirmed ORR was 73.7%(95%CI:63.6-83.8) and 1 patient was evaluated as CR. The DCR was 100%.The median follow-up was 10.7 months, median PFS was 13.8 months (95%CI:5.4m-22.2m) and 1-year PFS rate was 76.6% (95%CI:64.3%-88.9%). 11 patients (55.0%) had Grade ≥ 3 TRAEs. The most common Grade≥ 3 TRAEs were leukopenia (25.0%), neutropenia (20.0%) and thrombocytopenia(15%).

Conclusions

Adding HDACi(Chidamide) plus Tislelizumab combined with chemotherapy showed encouraging antitumor activity and a favorable safety profile as first-line therapy for advanced NSCLC. Efficacy and safety of the combination will be continuously monitored.

Clinical trial identification

ChiCTR2000041542

Date of Registration-2020.12.28

Collapse
Poster Display (ID 17) Poster Display

132P - Randomized Phase 3 Study of Tislelizumab Plus Chemotherapy Versus Chemotherapy Alone as First-Line Treatment for Advanced Squamous Non-Small Cell Lung Cancer (sq-NSCLC): RATIONALE-307 Updated Analysis (ID 555)

Presentation Number
132P
Lecture Time
12:30 - 12:30
Speakers
  • J. Wang (Beijing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Wang (Beijing, China)
  • S. Lu (Shanghai, China)
  • X. Yu (Hangzhou, Zhejiang, China)
  • Y. Hu (Wuhan, Hubei, China)
  • J. Zhao (Beijing, China)
  • M. Sun (Jinan, China)
  • Y. Yu (Harbin, China)
  • C. Hu (zhuozhou, China)
  • K. Yang (Wuhan, China)
  • Y. Song (Nanjing, Jiangsu Province, China)
  • X. Lin (Shanghai, China)
  • L. Liang (Beijing, China)
  • S. Leaw (Shanghai, China)
  • W. Zheng (Beijing, China)

Abstract

Background

Interim analysis of the open-label phase 3 RATIONALE-307 study (NCT03594747) demonstrated clinical benefit, including significantly improved progression-free survival (PFS) with a manageable safety profile, of tislelizumab (TIS) plus chemotherapy (chemo) as first-line (1L) therapy in patients (pts) with advanced sq-NSCLC vs chemo alone. Here, we report updated results.

Methods

Adults with previously untreated stage IIIB (not amenable to curative surgery/radiotherapy)/IV sq-NSCLC were randomized (1:1:1) to intravenous TIS (200mg, 21-day cycles) + paclitaxel + carboplatin (Arm A); TIS + nab-paclitaxel + carboplatin (Arm B); or paclitaxel + carboplatin (Arm C). The primary endpoint was PFS in Arms A and B vs Arm C, per independent review committee (IRC). Secondary endpoints included overall survival, objective response rate (ORR), duration of response (DoR), and safety.

Results

As of 30 September 2020 (median follow-up 16.7 months), of 360 randomized pts, 355 received treatment. The updated median (95% confidence interval [CI]) PFS benefit was maintained for Arms A (7.7 [95%CI: 6.7, 10.4] months [mo], stratified hazard ratio [HR] 0.45 [95%CI: 0.33, 0.62]) and B (9.6 mo [95%CI: 7.4, 10.8], HR 0.43 [95%CI: 0.31, 0.60]) vs C (5.5 mo [95%CI: 4.2, 5.6]). Consistent improvements in ORR in Arms A (74.2% [95%CI: 65.4, 81.7]) and B (73.9% [95%CI: 65.1, 81.6]) vs C (47.9% [95%CI: 38.8, 57.2]) were observed. Median DoR in Arms A and B was 8.4 (95%CI: 5.0, 15.8) mo and 8.6 (95%CI: 7.1, 12.5) mo, respectively vs 4.3 (95%CI: 2.9, 5.4) mo in Arm C. The incidences of any grade (Arm A 100%; Arm B 99.2%; Arm C 100%) or ≥grade 3 (Arm A 89.2%; Arm B 87.3%; Arm C 84.6%) treatment-emergent adverse events (TEAE) were similar between arms. The rate of treatment discontinuation due to TEAE was similar between Arms A (17.5%) and C (15.4%), and lower than in Arm B (32.2%). No new safety signal was identified.

Conclusions

In RATIONALE-307, the addition of TIS to chemo continued to demonstrate clinical benefit as 1L treatment of advanced sq-NSCLC vs chemo alone after a longer follow-up, with a manageable safety profile.

Clinical trial identification

NCT03594747

Editorial acknowledgement

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

Collapse
Poster Display (ID 17) Poster Display

133P - Efficacy of Nivolumab Immunotherapy in Non Small-cell Lung Cancer Patients with Bone Metastasis (ID 640)

Presentation Number
133P
Lecture Time
12:30 - 12:30
Speakers
  • A. Mishra (Bhopal, Madhya Pradesh, India)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Mishra (Bhopal, Madhya Pradesh, India)
  • S. Mishra (Gurgaon, Madhya Pradesh, India)
  • P. Sharma (New Delhi, India)

Abstract

Background

Bone metastasis is frequently seen in NSCLC (non small-cell cancer )and have got poor prognosis. Bone is a hematopoietic organ and actively regulates our immune system. Nivolumab is a fully human IgG4 PD-1 ICI (immune checkpoint inhibitor) antibody, which disrupts PD-1 mediated signaling and restores antitumor immunity. We have studied the clinical outcome of Nivolumab monotherapy in NSCLC patients with bone metastasis.

Methods

All patients with NSCLC with bone metastasis treated in our institute between Jan2018 to Aug2021 were included in our study. Patients who have received immunotherapies in past were excluded. Out of total 23 patients (16 men and 7 women), with median age of 58 years, 18 had adenocarcinoma and 5 had squamous cell carcinoma. Median followup was 13months.

All patients were previously treated with chemotherapies (platinum based/epidermal growth factor receptor-tyrosine kinase inhibitor), Bone modifying agents, Denosumab, Zolendronic acid. Now they are given IV infusion of Nivolumab every 2 weeks.

Tumor response were defined as complete/partial response and stable/progressive disease according to RECIST1.1. Responders were classified as either CR or PR and non responders as either PD or SD.

Radiological responses were assessed using the MDA (MD Anderson response classification) criteria and it divided response into 4 categories and can assess both the egression of extaskeletal lesions and osteosclerotic change (OC) on computed tomography due to the repairative process.

Results

A median of 5 doses of Nivolumab was given to patients (2-18 doses) with median treatment duration of 2.1 months (1- 16months). It resulted in overall survival(OS) of 58% at 1 year and overall response rate(ORR) of 21%, and time to response(TTR) was 2.5 months, which is consistent with previous studies. Bone response was achieved in 10 lytic lesions in 8 patients. No change was seen in mixed and blastic lesions.

Univariate analysis showed that the no. of bone metastasis was the only risk factor for non responders.

Conclusions

Bone metastasis impairs immunotherapy efficacy. Nivolumab monotherapy is effective for NSCLC patients with bone metastasis. Early bone response to this immunotherapy can be useful for early prediction and prognosis of such patients.

Clinical trial identification

CMCH/32/2021

Collapse
Poster Display (ID 17) Poster Display

134P - Safety and efficacy of multi-target TKI combined with nivolumab in check-point inhibitor-refractory advanced NSCLC patients: a prospective, single arm, two stage study (ID 257)

Presentation Number
134P
Lecture Time
12:30 - 12:30
Speakers
  • B. Han (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • B. Han (Shanghai, China)
  • B. Zhang (Shanghai, China)
  • H. Zhong (Shanghai, China)
  • C. Shi (Shanghai, China)
  • Z. Gao (Shanghai, China)
  • R. Zhong (Shanghai, China)
  • A. Gu (Shanghai, China)
  • T. Chu (Shanghai, China)
  • H. Wang (Shanghai, China)
  • L. Xiong (Shanghai, China)
  • W. Zhang (Shanghai, China)
  • X. Zhang (Shanghai, China)
  • B. Yan (Shanghai, China)
  • J. Teng (Shanghai, China)
  • W. Wang (Shanghai, China)
  • H. Bai (Shanghai, China)
  • R. Qiao (Shanghai, China)
  • L. Cheng (Shanghai, China)
  • Y. Kuang (Shanghai, China)

Abstract

Background

Vascular endothelial growth factor (VEGF) inhibition may reverse suppressive microenvironment and recover sensitivity to subsequent immune checkpoint inhibitors (ICIs).

Methods

This is a phase Ib/IIa, single-arm study, comprising dose finding (Part A) and expansion cohort (Part B). Immune checkpoint-inhibitor refractory NSCLC were enrolled. The first 21-day treatment cycle was a safety observation period (phase Ib) followed by a phase II expansion cohort (nivolumab 360mg, every 3 weeks, plus anlotinib, RP2D, 14 days on and 7 days off). The primary objective is recommended phase 2 dose (RP2D, part A) safety (part B) and ORR (part B), respectively.

Results

Between November 2020 and March 2022, 35 patients were screened and 21 eligible patients were enrolled (6 patients in Part A). The PR2D is anlotinib 12 mg/day orally (14 days on and 7 days off) and nivolumab (360mg every 3 weeks). Adverse events of any cause and treatment-related were reported in all patients. Two patients (9.5%) experience grade 3 TRAE. No grade 4 or higher adverse events were observed. Serious adverse were reported in 4 patients. There were 6 and 4 patients experience anlotinib and nivolumab disruption due to TRAE. Dese reduction to 10mg at any time was required in 5 patients and no patients decreased to 8mg. ORR and DCR was 19.0% and DCR is 76.2%, respectively. Median PFS and OS were PFS was 7.43 months (95% CI, 4.54-10.21m) and 19.1months (95% CI, 10.37-NE).

Conclusions

Our study suggested that full dose anlotinib combined with nivolumab showed positive safety and efficacy signal. Further study is warranted.

Clinical trial identification

NCT04507906

Collapse
Poster Display (ID 17) Poster Display

135P - Camrelizumab plus chemotherapy as first-line therapy for NSCLC: a pooled analysis of two randomized phase 3 trials with extended follow-up (ID 371)

Presentation Number
135P
Lecture Time
12:30 - 12:30
Speakers
  • C. Zhou (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Zhou (Shanghai, China)
  • G. Chen (Heilongjiang, China)
  • Y. Huang (Kunming, China)
  • J. Chen (Changsha, China)
  • Y. Cheng (Changchun, Jilin, China)
  • Q. Wang (Zhengzhou, China)
  • Y. Pan (Hefei, China)
  • J. Zhou (Hangzhou, China)
  • J. Shi (Linyi, China)
  • X. Xu (Yangzhou, China)
  • L. Lin (Guangzhou, China)
  • W. Zhang (Nanchang, China)
  • Y. Zhang (Hangzhou, Zhejiang, China)
  • Y. Liu (Shenyang, China)
  • Y. Fang (Hangzhou, China)
  • J. Feng (Nanjing, China)
  • Z. Wang (Jinan, China)
  • Y. Tai (Shanghai, China)
  • X. Ma (Shanghai, China)
  • X. Lu (Shanghai, China)

Abstract

Background

The phase 3 CameL and CameL-sq studies have proven the superiority of first-line camrelizumab plus chemotherapy (camre-chemo) vs chemo for progression-free survival (PFS) in patients (pts) with advanced non-squamous and squamous NSCLC, respectively. Here, we report pooled outcomes of pts from the two trials with long-term follow-up.

Methods

Pts with stage IIIB–IV NSCLC, no EGFR/ALK alterations, ECOG PS of 0 or 1, and no prior systemic therapy for metastatic disease were randomized to 4–6 cycles of carboplatin-pemetrexed ± camre followed by maintenance pemetrexed ± camre (non-squamous) or carboplatin-paclitaxel + placebo/camre followed by placebo/camre (squamous). Crossover from chemo to camre was permitted at radiographic progression, and RPSFT model was used to adjust for crossover. Total camre exposure was up to 2 yrs.

Results

801 pts were included (camre-chemo, N=398; chemo, N=403). Baseline characteristics were well balanced between the two groups (male, 82% with camre-chemo vs 82% with chemo; median age, 61 vs 61 yrs; ECOG PS of 1, 78% vs 80%; smoking history of ≥20 packs/yr, 73% vs 71%; PD-L1 TPS of ≥1%, 59% vs 52%). As of Jan 31, 2022, median follow-up was 21.6 mo (range 0.2–51.3) in camre-chemo group and 15.2 mo (range 0.5–48.6) in chemo group. Median PFS (mPFS) per investigator was 11.0 mo (95% CI 9.3–12.6) with camre-chemo vs 5.5 mo (95% CI 5.5–5.6) with chemo (HR 0.44 [95% CI 0.38–0.52]; 1-sided p <0.0001). Median overall survival (mOS) was 26.1 mo (95% CI 22.9–29.2) with camre-chemo vs 16.8 mo (95% CI 14.9–19.9) with chemo (HR 0.66 [95% CI 0.55–0.79]; 1-sided p <0.0001); the OS rate was 52.3% (95% CI 47.1–57.3) vs 37.7% (95% CI 32.8–42.6) at 24 mo, 31.9% (95% CI 25.6–38.4) vs 21.0% (95% CI 16.0–26.4) at 36 mo, and 26.8% (95% CI 19.5–34.7) vs 12.4% (95% CI 7.4–18.7) at 48 mo. Crossover-adjusted mOS was 14.5 mo (95% CI 12.9–16.5) with chemo (HR 0.52 [95% CI 0.43–0.63]; 1-sided p <0.0001). No new safety signals were observed.

Conclusions

With extended follow-up, camre plus chemo continued to provide clinically meaningful survival benefits over chemo with manageable safety in pts with previously untreated, advanced non-squamous and squamous NSCLC.

Clinical trial identification

NCT03134872 (First Posted: May 1, 2017) and NCT03668496 (First Posted: September 12, 2018)

Collapse
Poster Display (ID 17) Poster Display

136P - Tislelizumab (TIS) plus chemotherapy (chemo) for EGFR-mutated non-squamous non-small cell lung cancer (nsq-NSCLC) failed to EGFR tyrosine kinase inhibitors (TKIs) therapies: the primary analysis (ID 419)

Presentation Number
136P
Lecture Time
12:30 - 12:30
Speakers
  • B. Han (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • B. Han (Shanghai, China)
  • H. Zhong (Shanghai, China)
  • P. Tian (Chengdu, China)
  • Y. Zhao (Zhengzhou, China)
  • Q. Guo (Jinan, Shandong, China)
  • X. Yu (Hangzhou, Zhejiang, China)
  • Z. Yu (Qingdao, China)
  • X. Zhang (Shanghai, China)
  • Y. Li (Chengdu, China)
  • L. Chen (Zhengzhou, China)
  • Y. Zhang (Jinan, China)
  • X. Shi (Hangzhou, China)
  • J. Wang (Qingdao, Shandong, China)

Abstract

Background

Treatment option is limited for EGFR-mutated NSCLC after failure to EGFR TKIs. This multicenter, open-label, phase II study aims to evaluate the efficacy and safety of TIS plus chemo (cohort 1) or TIS plus chemo and bevacizumab (cohort 2) in EGFR-mutated nsq-NSCLC pts failed to EGFR TKI therapies. Herein, the primary analysis of cohort 1 is reported.

Methods

In cohort 1, pts with EGFR sensitizing mutations who had failed prior EGFR-TKIs received TIS plus carboplatin and nab-paclitaxel (induction), followed by TIS plus pemetrexed (maintenance). Primary endpoint was 1-year PFS rate; the planned sample size was 66 with a historical control of 7% (chemo), an expected value of 20%, one-sided α of 0.05, and power of 85%.

Results

From Jul 2020 to Dec 2021, 69 pts were enrolled; 39 pts (56.5%) harbored EGFR exon 19del; 28 pts (40.6%) had exon 21 L858R. 34 pts (49.3%) had progression on both 1st /2nd and 3rd EGFR-TKIs. As of 30 Jun 2022 (median follow-up, 8.2 months), 23.2% (n=16) of pts remained on treatment. Among 62 pts in EAS (Table), the confirmed ORR and DCR were 50.0% (95% CI 37.0-63.0%) and 87.1 % (95% CI 76.1-94.3%), respectively. Median PFS was 7.6 (95% CI, 6.4-9.8) months, with a 1-year PFS rate of 23.8% (90% CI, 13.1-36.2%). Pts with L858R mutation or having prior 1st/2nd EGFR-TKIs tended to have a longer PFS compared with pts with EGFR exon 19del mutation or progressed on 1st /2nd and 3rd EGFR-TKIs. Median OS was not reached (95% CI, 14.0-NE), and 1-year OS rate was 74.5% (95% CI, 56.5-86.0%). Safety profile was consistent with previous reports of TIS plus chemo in pts with EGFR-wt NSCLC. Grade 3-4 TEAEs occurred in 40.6% (28/69) of pts. 27.5% (19/69) of pts experienced irAEs; grade 3-4 irAEs occurred in 5 (7.2%) pts.

Table

EAS (Efficacy analysis set*, n=62)
BOR
PR 35 (56.5)
SD 19 (30.6)
PD 7 (11.3)
NA 1 (1.6)
Confirmed ORR, % (95% CI) 50.0 (37.0, 63.0)
DCR, % (95% CI) 87.1 (76.1, 94.3)
Median TTR, months (range) 1.70 (1.2, 7.7)
Median DOR, months (95% CI) 6.1 (4.7, 10.3)
Median PFS, months (95% CI) 7.6 (6.4, 9.8)
1-year PFS rate, % (90% CI) 23.8 (13.1, 36.2)
Median OS, months (95% CI) NR (14.0, NE)
1-year OS rate, % (95% CI) 74.5 (56.5, 86.0)
*Included pts receiving ≥1 dose of TIS or chemo, and having completed ≥1 post-treatment tumor assessment unless treatment was discontinued before the first tumor assessment due to disease progression or death; NA, not accessible; NR, not reached; NE, not estimable.

Conclusions

The study met the primary endpoint for cohort 1. TIS plus chemo is effective with acceptable safety profile for EGFR-mutated non-squamous NSCLC after EGFR TKI failure.

Clinical trial identification

NCT04405674

Collapse
Poster Display (ID 17) Poster Display

137P - Tislelizumab Combined with Chemotherapy as Neoadjuvant Therapy for Stage IIIA-IIIB(N2) Potentially resectable Squamous Non-small-cell Lung Cancer (TACT) (ID 527)

Presentation Number
137P
Lecture Time
12:30 - 12:30
Speakers
  • J. Shan (Hangzhou, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Shan (Hangzhou, China)
  • Z. Liu (Hangzhou, China)
  • C. Du (Hangzhou, China)
  • Y. Hu (Shanghai, China)
  • L. Ruan (Hangzhou, China)
  • X. Teng (Hangzhou, China)
  • L. Wang (Hangzhou, China)
  • M. Du (Hangzhou, China)
  • T. Tian (Hangzhou, China)
  • D. Jiang (Beijing, China)
  • Z. Tu (Hangzhou, China)

Abstract

Background

Recently, multiple clinical trials demonstrated neoadjuvant chemoimmunotherapy is a promising treatment option for resectable non-small-cell lung cancer (NSCLC). However, there is still limited evidence for using chemoimmunotherapy as neoadjuvant treatment in potentially resectable stage III squamous NSCLC. TACT(NCT05024266) is a phase II, open label, single arm trial evaluating the efficacy and safety of tislelizumab(Tis) plus chemotherapy(CT) as neoadjuvant treatment for potentially resectable stage IIIA-IIIB(N2) squamous NSCLC.

Methods

Treatment-naive adults confirmed clinical stage IIIA-IIIB (AJCC 8th), potentially resectable squamous NSCLC and ECOG PS 0-1 were eligible. Patients(pts) intravenously received Tis (200mg d1) + Albumin-bound paclitaxel (260mg/m2 d1) + Carboplatin (AUC 5 d1), Q3W for 2-4 cycles before surgery and 0-2 cycles post surgery (totally 4 cycles). The primary endpoint was major pathological response (MPR) rate and safety. Secondary endpoints included pathologic complete response (pCR) rate, R0 resection rate, overall response rate (ORR), median disease-free survival (DFS) and median overall survival (OS). Exploratory endpoints included biomarkers analysis.

Results

Between September 13, 2021 and May 17, 2022, 35 pts (median age: 65, IQR: 48-78; male: 100%; stage IIIB disease:17%; TPS PD-L1≥1% (22C3): 67.9% ,19/28) were enrolled. After 2-4 cycles of neoadjuvant treatment, 32 pts underwent surgical resection and all of them achieved R0 resection. 23 pts (71.9%) achieved MPR (95% CI, 53.3-86.3) and 11 pts (34.4%) achieved pCR (95% CI, 18.6-53.2). 31 pts(96.9%) had pathological downstaging (95% CI, 83.8-99.9) and no major surgical complications were observed. ORR was 88.6% (95% CI,73.3-96.8). DFS and OS data was immature. One patient (2.9%) experienced grade 3 immune-related hepatitis. No grade 4-5 adverse events were reported.

Conclusions

Neoadjuvant Tis with CT was feasible and safe for pts with potentially resectable stage IIIA-IIIB(N2) squamous NSCLC. Ongoing analysis of predictive biomarker on efficacy and safety will be available in the future meeting.

Clinical trial identification

Clinical trial identification: NCT05024266.

First Posted : August 27, 2021

Collapse
Poster Display (ID 17) Poster Display

138P - Randomized Phase 3 Study of Tislelizumab Plus Chemotherapy Versus Chemotherapy Alone as First-Line Treatment for Advanced Non-Squamous Non-Small Cell Lung Cancer (nsq-NSCLC): RATIONALE-304 Updated Analysis (ID 554)

Presentation Number
138P
Lecture Time
12:30 - 12:30
Speakers
  • S. Lu (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Lu (Shanghai, China)
  • J. Wang (Beijing, China)
  • Y. Yu (Harbin, China)
  • X. Yu (Hangzhou, Zhejiang, China)
  • Y. Hu (Wuhan, Hubei, China)
  • Z. Ma (Zhengzhou, China)
  • X. Li (Zhengzhou, Henan, China)
  • W. He (Beijing, China)
  • Y. Bao (Shanghai, China)
  • M. Wang (Beijing, China)

Abstract

Background

Interim analysis of the open-label phase 3 RATIONALE-304 study (NCT03663205) demonstrated clinical benefit of tislelizumab (TIS) plus chemotherapy (chemo) as first-line (1L) therapy in patients with advanced nsq-NSCLC vs chemo alone, with significantly improved progression-free survival (PFS) and a manageable safety profile. Here, we report the updated results.

Methods

Adults with treatment-naive, stage IIIB (not amenable to curative surgery/radiotherapy)/IV nsq-NSCLC were randomized (2:1) to receive platinum (carboplatin or cisplatin) and pemetrexed (PEM) every 3 weeks either with TIS (Arm A) or without (Arm B), followed by maintenance TIS + PEM (Arm A) or PEM (Arm B). The primary endpoint was PFS in Arm A vs Arm B, per independent review committee (IRC). Secondary endpoints included overall survival, objective response rate (ORR), duration of response (DoR), and safety.

Results

As of 15 July 2022, the median PFS per IRC was 9.8 (95% confidence interval [CI]: 8.9, 11.7) vs 7.6 (95% CI: 5.4, 8.0) months (mo) in Arm A vs Arm B, respectively (stratified hazard ratio 0.61 [95% CI: 0.46, 0.82]). ORR was greater in Arm A (51.6% [95% CI: 44.8, 58.3]) vs Arm B (27.9% [95% CI: 19.8, 37.2]) and median DoR was longer (14.5 [95% CI: 10.1, 24.4] vs 8.4 [95% CI: 6.0, 15.5] mo, respectively). TIS plus chemo was tolerable with no new safety signals identified after longer follow-up. The incidences of ≥grade 3 treatment-emergent adverse events (TEAEs) and of TEAEs leading to death (including disease progression-related AEs) in Arm A and Arm B were 69.4% and 56.4%, and 4.1% and 1.8%, respectively.

Conclusions

In RATIONALE-304, the addition of TIS to platinum plus PEM continued to demonstrate favorable efficacy and was generally well tolerated as 1L treatment of advanced nsq-NSCLC vs chemo alone, with no new safety signals identified.

Clinical trial identification

NCT03663205

Editorial acknowledgement

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

Collapse
Poster Display (ID 17) Poster Display

139P - The timing, dynamics, and co-occurrence of immune-related adverse events with a PD-L1 inhibitor in non-small cell lung cancer (ID 446)

Presentation Number
139P
Lecture Time
12:30 - 12:30
Speakers
  • K. Smith (Rochester, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • K. Smith (Rochester, United States of America)
  • S. Pritzl (Rochester, United States of America)
  • W. Yu (South San Francisco, United States of America)
  • I. Bara (South San Francisco, United States of America)
  • G. Thanarajasingam (Rochester, United States of America)
  • M. Kaul (South San Francisco, United States of America)
  • K. Williams (South San Francisco, United States of America)
  • A. Dueck (Wayne, United States of America)
  • A. Mansfield (Rochester, MN, United States of America)

Abstract

Background

Immune related adverse events (irAEs) due to immune checkpoint inhibitors (ICI) can lead to significant morbidity. Most clinical trials only report irAE frequency, but there is a significant need to better understand complicated irAE clinical courses. We sought to comprehensively evaluate irAE data, including timing, dynamics, and occurrence of multiple events.

Methods

The data from 2457 patients who participated in the Impower 130, 132, and 150 clinical trials investigating the PD-L1 inhibitor atezolizumab for metastatic non-small cell lung cancer were pooled for this analysis. Longitudinal irAE data with landmark analysis, changes in grading severity, and concurrent events were summarized.

Results

1557 patients were treated with atezolizumab (A) and 900 patients were in control (C) groups. Median follow up time was 32.3 months and 23.5 months for the A and C groups, respectively. In the A group, 753 patients (48.4%) experienced an irAE compared to 289 patients (32.1%) in the C group who experienced a non-immune adverse event attributed to irAE. The most common events in the A group were rash (28%), hepatitis (15%), hypothyroidism (12%), and pneumonitis (6%). 13% of these patients experienced two irAEs, most commonly rash and hepatitis (4%) followed by rash and hypothyroidism (3%), and 4% experienced three irAEs. Within five months, the cumulative incidence for all irAEs was 39.2%, but this varied between different irAEs with a lower incidence in the first five months for rash (23%), hepatitis (11%), hypothyroidism (7%), and pneumonitis (4%). IrAE grading increased in severity for some patients with grade 1 events increasing for 39% of patients, grade 2 events increasing for 21%, and grade 3 increasing for 16%. We utilized shift tables and upset plots to visually represent these complex patterns.

Conclusions

We identified differences in irAE patterns, including expected time to onset, frequency at which severity increases, and incidence of developing more than one irAE. These results can improve clinical management, represent a new standard for adverse event reporting, and inform better prospective data collection methodology to enable more comprehensive longitudinal analyses.

Clinical trial identification

NCT02367781, NCT02657434, NCT02366143

Collapse
Poster Display (ID 17) Poster Display

140P - Patient-Reported Outcomes (PRO) Following Sintilimab Plus Platinum and Gemcitabine as First-Line Treatment for Advanced or Metastatic Squamous NSCLC: a Randomized, Double-Blind, Phase 3 Study (Orient-12) (ID 178)

Presentation Number
140P
Lecture Time
12:30 - 12:30
Speakers
  • C. Zhou (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Zhou (Shanghai, China)
  • G. Gao (Shanghai, China)
  • Y. Fan (Hangzhou, China)
  • L. Liu (Nanjing, China)
  • L. Zhang (Beijing, China)
  • S. Cang (Zhengzhou, Henan, China)
  • J. Zhou (Hangzhou, China)
  • B. Li (Beijing, China)
  • Y. Yang (Shanghai, China)
  • J. Li (Shanghai, China)

Abstract

Background

In ORIENT-12 trial, sintilimab plus gemcitabine and platinum provided a significant and clinically meaningful PFS improvement comparing to placebo plus gemcitabine and platinum in Chinese patients with advanced/metastatic squamous NSCLC. This abstract summarizes PRO results.

Methods

357 participants were randomized (179 in sintilimab and 178 in placebo arm). PRO was assessed using LCSS and QLQ-C30 before the first dose, at each imaging evaluation and end-of-treatment visit. Clinically meaningful differences were defined as ≥10 points on a 0-100 scale, except for LCSS three-item global index (3-IGI; ≥30 points). Deterioration was defined as the onset of ≥10-point increase from baseline. P-value <0.05 was considered statistically significant.

Results

PRO compliance rates maintained high until week 30. Baseline scores for each item were similar between arms. During the overall treatment period, QLQ-C30 scores were maintained regardless of the addition of sintilimab [Least square mean changes from baseline (95%CI): sintilimab: -1.04 (-3.42, 1.33); placebo: -0.74 (-3.33, 1.85); Between-group difference (95%CI): -0.30 (-3.81, 3.22)]; LCSS total score, 3-IGI, and average symptom burden index were maintained for both groups. LCSS composite endpoint and respiratory symptoms endpoint showed statistically significant improvement at week 6 and 12 for both arms, but changes did not reach a clinically meaningful difference. Sintilimab arm showed a numerical trend towards delayed deterioration across most LCSS and QLQ-C30 items. Notably, sintilimab arm showed significantly delayed in pain and major symptoms in QLQ-C30, as well as blood in sputum (Median: 56.9 vs 44.9 weeks, HR: 0.56; 95% CI: 0.37, 0.84, p=0.006) and painful sensations (Median: 49.1 vs 44.0 weeks, HR: 0.65; 95% CI: 0.44, 0.95, p=0.027) in LCSS.

Conclusions

The addition of sintilimab to chemotherapy treatment maintained the quality-of-life and delayed the main symptoms deterioration compared to placebo. The data support sintilimab plus platinum and gemcitabine as first-line treatment for advanced/metastatic squamous NSCLC.

Clinical trial identification

ClinicalTrials.gov: NCT03629925

Collapse
Poster Display (ID 17) Poster Display

141P - Efficacy and Safety of Tislelizumab Combined with Targeted Therapy as First-Line Treatment for Unresectable Hepatocellular Carcinoma: A Real-world Study (ID 364)

Presentation Number
141P
Lecture Time
12:30 - 12:30
Speakers
  • L. Wang (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Wang (Shanghai, China)
  • Y. Zhao (Shanghai, China)
  • T. Zhang (Shanghai, China)
  • L. Wang (Shanghai, China)

Abstract

Background

Immunotherapy combined with targeted therapy have become recommended regimens for advanced HCC. Considering that patients are strictly screened in clinical trials, while the efficacy and safety in the real-world are affected by various factors, real-world data of anti-PD-1 antibody combined with targeted therapy is urgently needed.

Methods

In this non-interventional study, patients with histologically or clinically confirmed unresectable HCC with BCLC Stage B/C who were planning to receive or have already received tislelizumab combination therapy were enrolled. Treatment included tislelizumab (200 mg IV Q3W) plus a tyrosine kinase inhibitor (TKI) or VEGFR2 inhibitor selected from lenvatinib (12mg/8mg PO QD), sorafenib (400 mg PO BID) and apatinib (750 mg QD). During the treatment, patients assessed as eligible for resection would undergo surgery. The primary endpoint was ORR assessed by RECIST 1.1. Secondary endpoints included DCR, PFS, OS and safety.

Results

From March 2020 to March 2021, 44 patients were enrolled with a median age of 55 (range: 31-71) years old. Among them, 37 (84.1%) patients were male, 13 (29.5%) patients had extrahepatic metastase. Child-Pugh class A (n=42,95.5%) or B (n=2, 4.5%); ECOG PS 0 (n=24, 54.5%) or 1 (n=20, 45.5%). Patients received tislelizumb plus lenvatinib (n=33; 3 of which also received TACE), or plus sorafenib (n=7), or plus apatinib (n=3). The ORR and DCR were 47.7% (21/44) and 84.1% (37/44), respectively. 15 (34.1%) patients were evaluated as operable after the combination treatment; 3 of them achieved pathological complete response (pCR). No severe postoperative complications were observed. Grade 3 or higher AEs were mainly hypertension (11.4%), rash (9.1%), proteinuria (6.8%), thrombocytopenia (6.8%), hand-foot syndrome (6.8%) and febrile neutropenia (6.8%).

Conclusions

In the real-world setting, tislelizumab plus targeted therapy shows favorable efficacy with reasonable tolerability as 1L treatment options for patients with unresectable HCC. The combination may also provide an opportunity for curative resection.

Clinical trial identification

The clinical trial registration number is NCT04996459.

Enrollment of this study began in March 2020.

Collapse
Poster Display (ID 17) Poster Display

142P - CODAK real-world study: Interim analysis of clinical outcomes in unresectable stage III NSCLC patients treated with durvalumab after chemoradiotherapy (CRT) in the United Kingdom. (ID 187)

Presentation Number
142P
Lecture Time
12:30 - 12:30
Speakers
  • K. Franks (Leeds, Yorkshire, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • K. Franks (Leeds, Yorkshire, United Kingdom)
  • M. Ahmed (London, United Kingdom)
  • D. Smith (London, United Kingdom)
  • P. Shaw (Cardiff, United Kingdom)
  • G. Banna (Portsmouth, United Kingdom)
  • M. Cominos (Maidstone, Kent, United Kingdom)
  • J. Walther (Taunton, Somerset, United Kingdom)
  • T. Talbot (Truro, Cornwall, United Kingdom)
  • P. Taylor (Manchester, United Kingdom)
  • B. Blak (Northolt, United Kingdom)
  • L. Lindqvist (Cambridge, United Kingdom)
  • S. Paul (Cambridge, United Kingdom)
  • D. Vincent (Luton, United Kingdom)

Abstract

Background

There is a lack of real-world data on the effectiveness of durvalumab in the UK clinical practice setting. The ongoing observational CODAK (NCT04667312) study addresses this data gap.

Methods

CODAK is a non-interventional cohort study with retrospective data collection and a prospective observational period of patients (pts) with locally advanced, unresectable Stage III NSCLC initiated on durvalumab following concurrent or sequential CRT in the UK from September 2017 through December 2019. Data on demographic and clinical characteristics, treatment with durvalumab and outcomes were extracted from medical records of eligible pts identified at 10 participating centres by direct care teams (planned enrolment, 120 pts). The primary outcome is real-world overall survival (rwOS) rate at 12- and 24-months post durvalumab initiation. Interim results are reported.

Results

Data cut-off for this analysis was 24 March 2022 (median follow-up from durvalumab initiation: 16.5 months). In 47 pts for whom outcomes data on durvalumab were available, 36 (77%) were aged ≥60 years and 30 (64%) were male. The 12- and 24-month rwOS rate was 75.2% (95% CI: 63.5–89.1) and 50.9% (95% CI: 37.3–69.4), and the 12- and 24-month real-world progression-free survival (rwPFS) rate was 53.6% (95% CI: 40.9–70.4) and 43.3% (95% CI: 30.7–61.1), respectively. Median rwOS and rwPFS was not reached and 15.2 months (95% CI: 7.1–not reached), respectively. The median time to subsequent therapy was 16.0 months (95% CI: 11.0–not reached). Mean duration of durvalumab treatment for the 47 pts was 195 days (range: 8–456). Mean radiotherapy dose and number of fractions per pt was 60.8 Gy and 28.4, respectively. Durvalumab treatment was discontinued due to toxicity/adverse event (AE) in 11 (23%) pts. Pneumonitis/interstitial lung disease was the most common AE leading to discontinuation (reported in 8 [17%] pts).

Conclusions

Interim results from CODAK, based on both limited sample size and follow-up, demonstrate the effectiveness of consolidation durvalumab after CRT in a real-world cohort of UK pts with unresectable Stage III NSCLC. Safety data were comparable to results from PACIFIC (NCT02125461).

Clinical trial identification

NCT04667312

Editorial acknowledgement

Medical writing and editorial assistance, which was in accordance with Good Publications Practice(GPP3) guidelines, was provided by Patrick Foley, PhD, of NexGen Healthcare (London, UK) andfunded by AstraZeneca UK.

Collapse
Poster Display (ID 17) Poster Display

143P - Efficacy and safety of bronchial arterial chemoembolization (BACE) in combination with tislelizumab for non-small cell lung cancer (NSCLC): A single-arm phase II trial (ID 509)

Presentation Number
143P
Lecture Time
12:30 - 12:30
Speakers
  • X. Duan (Zhengzhou, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • X. Duan (Zhengzhou, China)
  • H. Li (Zhengzhou, China)
  • D. Kuang (Zhengzhou, China)
  • M. Zhang (Zhengzhou, China)
  • W. Xu (Zhengzhou, China)
  • C. Liang (Zhengzhou, China)
  • J. Wang (Zhengzhou, China)
  • J. Ren (Zhengzhou, China)

Abstract

Background

Immunotherapy and BACE are important therapy for NSCLC. This trial is designed to determine the efficacy and safety of immunotherapy with tislelizumab in addition to BACE in stage III-IV NSCLC patients (pts) who failed, refused or ineligible to receive standard treatments.

Methods

This trial enrolled stage III-IV pts without EGFR, ALK or ROS1 aberrations who have failed, refused, or assessed ineligible to receive conventional treatments (surgery, chemoradiotherapy, chemotherapy). Pts were treated with BACE and tislelizumab as induction therapy during which BACE was performed on the first day and tislelizumab was given 3-5 days later, then tislelizumab was administered at 200mg Q3W as maintenance therapy. Primary endpoint was progression-free survival (PFS), and secondary endpoints included objective response rate (ORR), disease control rate (DCR), overall survival (OS) and safety, etc. Herein, the treatment response at 6 weeks is reported.

Results

30 pts were enrolled with 24 (80%) males pts and a median age of 65.1y; 21 (70%) squamous, 9 (30%) adenocarcinomas; 4 (13%) stage IIIA, 11 (37%) stage IIIB, 15 (50%) stage IV; 8 (27%) with ECOG score 0, 17 (57%) with score 1, and 5(17%) with score 2. Among the 30 pts, 5 (17%) pts had bronchial stenosis, 2 (7%) had superior vena cava syndrome, 8 (27%) had pneumonia, 4 (13%) had pulmonary heart disease, and 4 (13%) had hemoptysis. The ORR and DCR were 66.7% and 83.3% respectively per RECIST1.1, 2 pts (6.7%) achieved complete response (CR), 18 pts (60.0%) and 5 pts (16.7%) got partial response (PR) and stable disease (SD). The main TEAEs related to BACE mainly include nausea (20.0%, 6/30), fever (16.7%, 5/30), hemoglobin decrease (6.7%, 2/30), leukopenia (10.0%, 3/30) and thrombocytopenia (6.7%, 2/30). TEAEs related to tislelizumab mainly included fatigue (6.7%, 2/30),rash (10.0%, 3/30) and cough (10.0%, 3/30). Grade≥3 AEs were not observed.

Conclusions

Tislelizumab combined with BACE for advanced NSCLC pts appears to be safe and feasible, and the comorbidities of patients can be alleviated after treatment. Compared with previous studies on BACE, the addition of immunotherapy may improve the ORR and DCR.

Clinical trial identification

Clinical trial information: NCT05286957.

Collapse
Poster Display (ID 17) Poster Display

144P - Toripalimab plus chemotherapy as neoadjuvant treatment for resectable stage IIB-IIIB NSCLC (RENAISSANCE study): a single-arm, phase 2 trial (ID 533)

Presentation Number
144P
Lecture Time
12:30 - 12:30
Speakers
  • S. Yan (Beijing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Yan (Beijing, China)
  • J. Wang (Beijing, China)
  • C. Lyu (Beijing, China)
  • J. Bi (Beijing, China)
  • Y. Xin (Beijing, China)
  • B. Liu (Beijing, China)
  • S. Li (Beijing, China)
  • Y. Wang (Beijing, China)
  • J. Chen (Beijing, China)
  • X. Li (Beijing, China)
  • Y. Yang (Beijing, China)
  • N. Wu (Beijing, China)

Abstract

Background

Neoadjuvant chemoimmunotherapy have a promising efficacy in resectable non-small cell lung cancer (NSCLC). We aimed to investigate the efficacy and safety of neoadjuvant immunotherapy plus chemotherapy for stage IIB-IIIB NSCLC.

Methods

Pts with stage IIB-IIIB, wildtype EGFR/ALK NSCLC, ECOG PS 0-1 were eligible. All pts received 2-4 cycles of toripalimab (240mg, q3w) plus double platinum-based chemotherapy. All pts were assessed by imaging/surgical indication after 2 cycles of treatment. Pts who cannot undergo surgery will be reassessed after another 1-2 cycles of neoadjuvant therapy. Primary endpoints were major pathological response (MPR), complete pathological response (pCR). Secondary endpoints were objective response rate (ORR), R0 resection rate and safety.

Results

A total of 81 pts (median age: 62, IQR: 45-76; female: 7, 8.6%, squamous cell carcinoma: 63, 77.8%) were enrolled since Dec 2020. Disease distribution in stage IIB, IIIA and IIIB consisted of 27, 41 and 13 pts, respectively. 16 pts were in the preoperative stage or unsuitable for surgery. 65 pts underwent R0 resection. 42 pts (42/65, 64.6%) achieved MPR, including 31 pts (31/65, 47.7%) with pCR. 62 pts received 2 or 3 cycles of treatment, and no significant difference in MPR or pCR rate was observed between 2 and 3 cycles of treatment (32/48, 66.7% vs 8/14, 57.1%, p=0.512; 25/48, 52.1% vs 5/14, 35.7%, p=0.281). After 2 cycles of treatment, the primary tumor shranked significantly with a -40% (IQR: -100%, +25%) median regression rate, and tended to flatten with a -4.63% (IQR: -21.54%, +21.15%) median regression rate after another 1-2 cycles. Among 31 pts who underwent imaging evaluation again after 2-cycle treatment assessment, 19 pts (19/31, 61.3%) still had tumor regression. 10 pts of these (10/19, 52.6%) received 3-4 cycles of treatment, and 9 pts (9/19, 47.4%) did not receive further treatment and continued regression was also observed. 69 pts (69/81, 85.2%) experienced TRAEs.

Conclusions

Neoadjuvant toripalimab and chemotherapy is a promising treatment for pts with stage IIB-IIIB NSCLC. Continued tumor regression after chemoimmunotherapy is not uncommon, and it is not entirely dependent on further cycles of neoadjuvant therapy.

Clinical trial identification

NCT04606303

Editorial acknowledgement

None.

Collapse
Poster Display (ID 17) Poster Display

145P - Preliminary results of a phase Ⅱ study of fruquintinib combined with sintilimab and chemotherapy as the first-line treatment in advanced naive EGFR- and ALK-negative non-squamous non-small cell lung cancer(nsq-NSCLC) (ID 341)

Presentation Number
145P
Lecture Time
12:30 - 12:30
Speakers
  • Y. Shu (Nanjing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Y. Shu (Nanjing, China)
  • P. Ma (Nanjing, China)
  • H. Shen (Nanjing, China)
  • W. Gao (Nanjing, China)
  • X. Chen (Nanjing, Jiangsu Province, China)
  • J. Sun (Nanjing, China)
  • L. Xu (Nanjing, China)

Abstract

Background

Fruquintinib(F) is an orally available VEGFR inhibitor that is highly selective for VEGFR-1,2 and 3. In the phase Ⅱ and Ⅲ trials, F has been shown an improved progression-free survival(PFS) compared to placebo and a manageable safety profile with advanced NSCLC. This study aimed to investigate the efficacy and safety of F plus PD-1 inhibitor sintilimab(S) and platinum-based chemotherapy(chemo) as the first-line treatment for advanced naive EGFR- and ALK-negative nsq-NSCLC.

Methods

This single-arm, open-label, phase Ⅱ study (NCT04956146) consists of a safety lead-in phase(Part 1) and dose expansion phase(Part 2). In Part 1 the F taken orally at 5 mg (2 weeks on/ 1 weeks off, Q3W) plus S (200mg, iv, d1,Q3W) and chemo(Q3W). After 4~6 cycles followed by maintenance therapy with F(RP2D) plus S and pemetrexed or not. DLT was observed for 1 cycle. The primary objective of Part 1 is to assess the safety and confirm the RP2D of F. The primary objective of Part 2 is to estimate the PFS and the secondary endpoints including ORR, DCR, OS, and safety.

Results

From February 2022 to September 2022, 15 pts were enrolled . Median age was 66, male 93.3%, ECOG PS 0 66.7%, adenocarcinoma 80%. No DLTs were reported in Part 1. The dose of F (5mg, 2 weeks on/ 1 weeks off, Q3W) was established as the RP2D. Of 9 evaluable pts, ORR and DCR were 66.7% and 100%(6 partial response and 3 stable disease). Median PFS was not reached yet. The most common treatment-emergent adverse events (TEAEs) (Total; Grade ≥3) were Alanine aminotransferase increased (100%; 7.7%), hypoalbuminaemia (100%; 0), Aspartate aminotransferase increased(92.3%; 0), White blood cell count decreased (84.6%; 38.5%) and Gamma-glutamyltransferase increased(84.6%, 23.1%).

Conclusions

This finding showed promising efficacy for fruquintinib combined with sintilimab and chemo in pts with advanced NSCLC as first-line treatment with a manageable safety profile. These findings support fruquintinib plus sintilimab and chemo as a potential new first-line treatment option for unresectable or metastatic advanced naive EGFR- and ALK-negative nsq-NSCLC.

Clinical trial identification

Trial protocol number: NCT04956146

Release date: February 2, 2022

Collapse
Poster Display (ID 17) Poster Display

146P - Stereotactic body radiotherapy plus anlotinib ± toripalimab in untreated oligometastatic brain metastases NSCLC patients (ID 293)

Presentation Number
146P
Lecture Time
12:30 - 12:30
Speakers
  • G. Han (Wuhan, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • G. Han (Wuhan, China)
  • J. Bi (Wuhan, China)
  • J. Ma (Wuhan, China)
  • M. Yuan (Wuhan, China)
  • Y. Li (Wuhan, China)
  • G. Pi (Wuhan, China)
  • Y. Li (Wuhan, China)
  • D. Hu (Wuhan, China)

Abstract

Background

This is a prospective, open-label, phase 1b study to evaluate the safety and feasibility of stereotactic body radiotherapy (SBRT) and anlotinib ± toripalimab in untreated oligometastatic brain metastases (BMs) NSCLC patients (pts).

Methods

Ten pts were randomized into 2 groups. Induction therapy: SBRT (35 Gy/5 F) plus anlotinib (12 mg, d1~14, q3w) with (group A) or without (group B) toripalimab (240 mg, d1, q3w). Then will continue toripalimab and anlotinib on d22 for 1 year until disease progression or intolerable toxicity. Key inclusion criteria: ≥ 18 years old, ECOG ≤ 1, driver mutation-negative, untreated NSCLC BMs (1~5 lesions). Primary endpoints: intracranial response rate (iORR) and treatment-related adverse events (TRAEs). Secondary endpoints: intracranial progression-free survival, intracranial disease control rate (iDCR), and overall survival.

Results

As of Sep 15, 2022, 8 males (group A: 5, group B: 3) were included. The median age of A was 67 (range 59-71) years. The median age of B was 63 (range 61-68) years, with 1 squamous cell carcinoma in each group. The median follow-up time was 147 days. Intracranial efficacy: after the induction therapy cycle, all pts in group A had SD, 2 in group B had SD and 1 had PR. During 3~4 months of treatment, the iORR and iDCR were 60% and 80% in group A (3 PR, 1 SD, and 1 PD: investigator error, missing brain radiotherapy, later stage salvage SBRT). Group B had 1 PR and 2 SD. In terms of systemic efficacy, group A: ORR was 40% and DCR was 80%; group B: ORR was 33.3% and DCR was 100%. 80% of group A pts developed any grade (G) TRAEs, mainly hypertension (3/5), including 1 G 3 hypertension; 1 concurrent had hypothyroidism, hypertension, elevated myocardial enzymes, and hand-foot syndrome, causing suspension of one cycle of toripalimab therapy, anlotinib was reduced to 8 mg. 66.7% (2/3) in group B experienced any grade TRAEs, 1 had G 3 hand-foot syndrome led to anlotinib dose reduction.

Conclusions

SBRT plus anlotinib ± toripalimab showed good short-term efficacy in untreated BMs NSCLC. The induction part combined immunotherapy increases manageable TRAEs incidence. Further efficacy and safety data require follow-up confirmation after enrollment.

Clinical trial identification

NCT05021328

Collapse
Poster Display (ID 17) Poster Display

147P - Characterizing CRS in phase 1 study of DLL3-targeted T-cell engager tarlatamab in small cell lung cancer (ID 308)

Presentation Number
147P
Lecture Time
12:30 - 12:30
Speakers
  • S. Champiat (Villejuif, Cedex, CEDEX, France)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Champiat (Villejuif, Cedex, CEDEX, France)
  • M. Boyer (Camperdown, NSW, Australia)
  • L. Paz-Ares (Madrid, Spain)
  • A. Schoenfeld (New York, United States of America)
  • H. Izumi (Kashiwa, Japan)
  • R. Govindan (St. Louis, MO, United States of America)
  • J. Carlisle (Atlanta, United States of America)
  • H. Borghaei (Philadelphia, United States of America)
  • M. Johnson (Nashville, TN, United States of America)
  • N. Steeghs (Amsterdam, Netherlands)
  • E. Vokes (Chicago, IL, United States of America)
  • A. Dowlati (Cleveland, United States of America)
  • Y. Zhang (Thousand Oaks, United States of America)
  • A. Pati (Thousand Oaks, United States of America)
  • C. Ju (Thousand Oaks, United States of America)
  • S. Mukherjee (Thousand Oaks, United States of America)
  • X. Chen (Thousand Oaks, United States of America)
  • N. Hashemi Sadraei (Thousand Oaks, CA, United States of America)
  • H. Hummel (Wuerzburg, Germany)

Abstract

Background

Tarlatamab (AMG 757) binds DLL3 on SCLC cells and CD3 on T cells leading to T cell-mediated tumor lysis. Updated phase 1 tarlatamab data in relapsed/refractory SCLC showed an ORR of 23%, median DOR of 13 months, and an acceptable safety profile with cytokine release syndrome (CRS) as the most common treatment-related (TRAE).1 Here, we describe clinical CRS seen with tarlatamab and explore associations between cytokine levels and CRS in cycle 1 (C1)-where CRS mostly occurs.

Methods

Tarlatamab (0.003–100.0 mg) was given intravenously every 2 weeks ± step dosing in patients (pts) with SCLC progressing after ≥1 platinum-based regimen. Safety/efficacy were evaluated as previously described.1 Serum was drawn at time points up to 24 hours (hrs) after first dose from pts receiving ≥1 mg initial dose and up to 100 mg in subsequent doses in C1. Peak level and elevation speed were evaluated for a panel of soluble factors in pts with CRS vs no CRS in C1.

Results

By 15 June 2022, 106 pts had received ≥1 dose tarlatamab with median follow-up time of 8.5 months (range, 0.2–30.7). Median treatment duration was 11.6 weeks (range, 0.1–80.0). TRAEs occurred in 97 pts (92%) and grade (gr) ≥ 3 in 33 pts (31%). CRS occurred in 56 pts (53%) with maximum gr1 in 41 pts and gr3 in 1 (1.0%), and 8 pts received tocilizumab for CRS. Most CRS events were in C1 (55 pts in C1, 5 pts in C2+). CRS had median onset of 17.5 hrs (in pts with both date and hr of onset data), was transient (median duration, 3 days), and resolved in all cases. While initial tarlatamab infusion is characterized by immune activation and inflammatory cytokine induction, among biomarker-evaluable pts (N=86), those with any grade CRS in C1 (n=45 or 46) showed trends in increased peak levels of IL-6, IL-8, IL-10, and TNF-α by 24 hrs after first dose vs those without (n=40).

Conclusions

Prior results from this study show manageable safety and promising efficacy of tarlatamab in pts with heavily pretreated SCLC. CRS is not unexpected based on tarlatamab’s mechanism of action, is associated with rises in some cytokines yet remains generally low-grade, reversible, and confined to C1. Further characterization of tarlatamab’s safety is ongoing.

References

1. Borghaei H, et al. OA12.05. WCLC 2022. Aug 8, 2022.

Clinical trial identification

NCT03319940

Editorial acknowledgement

Medical writing support was provided by Eugene Gillespie, PhD of Amgen Inc.

Collapse
Poster Display (ID 17) Poster Display

148P - AdvanTIG-105: Phase 1b Dose-Expansion Study of Ociperlimab (OCI) + Tislelizumab (TIS) With Chemotherapy in Patients (pts) With Extensive-Stage Small Cell Lung Cancer (ES-SCLC) (ID 654)

Presentation Number
148P
Lecture Time
12:30 - 12:30
Speakers
  • J. Zhang (Kansas City, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Zhang (Kansas City, United States of America)
  • M. Hussein (Fort Myers, United States of America)
  • S. Kao (Camperdown, NSW, Australia)
  • T. Clay (Subiaco, Australia)
  • N. Singhal (Adelaide, Australia)
  • H. Kim (Seoul, Korea, Republic of)
  • E. Cho (Incheon, Korea, Republic of)
  • B. Shim (Suwon, Korea, Republic of)
  • Y. Lee (Goyang, Korea, Republic of)
  • G. Lee (Jinju, Korea, Republic of)
  • J. Zhao (Beijing, China)
  • Y. Yu (Harbin, China)
  • M. Sun (Jinan, China)
  • C. Lin (Hualien City, Taiwan)
  • T. Yang (Taichung City, Taiwan)
  • G. Chang (Taichung City, Taiwan)
  • H. Zheng (Emeryville, United States of America)
  • W. Tan (Shanghai, Not Applicable, China)
  • D. Spigel (Nashville, TN, United States of America)

Abstract

Background

T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) inhibitor with an anti-programmed cell death protein 1 (PD-1) antibody is a promising combination showing antitumor activity in solid tumors. Phase 1/1b open-label study AdvanTIG-105 assessed safety and preliminary antitumor activity of anti-TIGIT monoclonal antibody (mAb) OCI + anti-PD-1 mAb TIS in pts with advanced unresectable solid tumors (NCT04047862). In dose-escalation, OCI + TIS was well tolerated showing preliminary antitumor activity, establishing the recommended phase 2 dose (RP2D) of OCI 900mg IV every 3 weeks (Q3W) plus TIS 200mg IV Q3W. We report dose-expansion results in pts with ES-SCLC.

Methods

Eligible adults had histologically/cytologically confirmed ES-SCLC and had received no prior systemic therapies. Pts received RP2D of OCI + TIS with cisplatin or carboplatin + etoposide Q3W for 4 cycles, followed by RP2D OCI + TIS Q3W until disease progression, intolerable toxicity, or withdrawal of consent. Primary endpoint was investigator-assessed objective response rate (ORR) per RECIST v1.1. Secondary endpoints included progression-free survival (PFS), duration of response (DoR), and safety.

Results

As of June 20, 2022, 42 pts were enrolled, of which 40 were efficacy evaluable; median study follow-up time was 24.9 weeks (range 3.0-67.9). Confirmed ORR was 65.0% (95% confidence interval [CI]: 48.3, 79.4) and median DoR was 4.3 months (95% CI: 3.2, 5.6). Median PFS was 4.9 months (95% CI: 4.2, 5.7) with a 6-month PFS rate of 27.3%. All 42 pts experienced at least 1 treatment-emergent adverse event (TEAE); 25 (59.5%) had Grade ≥ 3 TEAEs and 17 (40.5%) had serious TEAEs. Most common TEAEs were neutrophil count decreased and anemia (54.8% each). Immune-mediated TEAEs were reported in 12 pts (28.6%) and TEAEs led to treatment discontinuation in two pts. Pneumonia (unrelated to treatment) and disease progression led to death in two pts.

Conclusions

OCI 900mg + TIS 200mg with cisplatin/carboplatin plus etoposide was generally well tolerated and showed antitumor activity in pts with ES-SCLC.

Clinical trial identification

NCT04047862

Editorial acknowledgement

This study was sponsored by BeiGene, Ltd. Medical writing support, under the direction of the authors, was provided by Sophie Cook, PhD, of Ashfield MedComms, an Inizio company, and was funded by BeiGene, Ltd. The authors would like to thank Rang Gao and Ruihua Wang for their contributions.

Collapse
Poster Display (ID 17) Poster Display

149P - A phase Ⅰb/Ⅱ trial of surufatinib plus toripalimab and etoposide(E) combined with cisplatin (P) in patients(pts) with untreated advanced small cell lung cancer (SCLC) (ID 532)

Presentation Number
149P
Lecture Time
12:30 - 12:30
Speakers
  • W. Fang (Guangzhou, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • W. Fang (Guangzhou, China)
  • Y. Huang (Guangzhou, Guangdong, China)
  • Y. Yang (Guangzhou, China)
  • Y. Zhao (Qingdao, Shandong, China)
  • Y. Zhang (Guangzhou, China)
  • T. Zhou (Guangzhou, Guangdong, China)
  • S. Zhao (Guangzhou, China)
  • M. Yuxiang (Guangzhou, China)
  • S. Hong (Guangzhou, China)
  • H. Zhao (Guangzhou, China)
  • L. Zhang (Guangzhou, Guangdong, China)

Abstract

Background

Surufatinib (S, a small-molecule inhibitor of VEGFR1-3, FGFR1 and CSF-1R) plus toripalimab (T, an anti-PD-1 antibody) has exhibited encouraging efficacy in an SCLC cohort in a trial evaluating S + T in pts with selected solid tumors. This study is assessing the safety, tolerability, and preliminary efficacy of S+ T+ chemotherapy(EP) as first-line regimen for SCLC pts. Here, we report the preliminary efficacy results.

Methods

This study is a phase Ⅰb/Ⅱ, single-arm study(NCT04996771). Eligible pts were≥18 years old with histologically confirmed SCLC, ECOG PS 0-1, with at least one measurable lesion. Pts with treated, stable, and asymptomatic brain metastases(BMs) are allowed. A 3+3 dose-escalation was done to determine the recommended RP2D of S+T+EP. S was dosed at 150mg, 200mg, and 250mg qd, po, Q3W in the dose level(DL)1, 2, and 3, respectively, in combination with a fixed dose of T(200mg, iv, d1, Q3W) and EP(Q3W). After 4 cycles followed by maintenance therapy with S plus T every 3 weeks. This study started escalation in the DL2. DLT was observed for 1 cycle. The primary objective of phase Ⅰb is to assess the safety and confirm the RP2D of S. The primary objective of phase Ⅱ is to estimate the PFS and the secondary endpoints including ORR, DCR, OS, and safety.

Results

At cutoff date (Sep 20, 2022), 24 pts (phase Ⅰb DL2, n=6; phase Ⅱ, n=18) were enrolled and received treatment(median age 59 years, male 70.8% , ECOG PS 1 62.5%, BMs 8.3% ). Two DLTs occurred in the DL2, The RP2D was identified as S(200mg, po, qd, Q3W). Among pts with at least one post-baseline tumor assessment (evaluable pts, n=17), the confirmed ORR was 88.2%, and DCR(15 PRs, 2 SDs) was 100%. Median PFS was not reached yet. The most common treatment-emergent adverse events (TEAEs) (Total; Grade ≥3) were alopecia(66.7%; 0), white blood cell count decreased (57.1%; 28.6%), anemia(57.1%; 0), decreased appetite(52.4%; 0) and constipation(47.6%; 0).

Conclusions

Surufatinib plus toripalimab and etoposide combined with cisplatin showed promising anti-tumor activity and acceptable toxicity for the 1L treatment of SCLC. The combination of the 4 agents might be a novel 1L therapeutic option for SCLC.

Clinical trial identification

Trial protocol number: NCT04996771

Release date: November 9, 2021

Collapse
Poster Display (ID 17) Poster Display

150P - Phase 1 study of fianlimab, a human lymphocyte activation gene-3 (LAG-3) monoclonal antibody, in combination with cemiplimab in advanced melanoma (mel): subgroup analysis (ID 613)

Presentation Number
150P
Lecture Time
12:30 - 12:30
Speakers
  • O. Hamid (Los Angeles, CA, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • O. Hamid (Los Angeles, CA, United States of America)
  • K. Lewis (Aurora, CO, United States of America)
  • A. Weise (Detroit, United States of America)
  • M. McKean (Nashville, United States of America)
  • K. Papadopoulos (San Antonio, TX, United States of America)
  • J. Crown (Dublin, Ireland)
  • S. Thomas (Orlando, FL, United States of America)
  • J. Kaczmar (Charleston, United States of America)
  • N. Lakhani (Grand Rapids, United States of America)
  • T. Kim (Seoul, Korea, Republic of)
  • K. Kim (San Francisco, United States of America)
  • G. Rabinowits (Miami, United States of America)
  • A. Spira (Fairfax, United States of America)
  • J. Mani (Tarrytown, United States of America)
  • S. Chen (Tarrytown, United States of America)
  • G. Gullo (Tarrytown, United States of America)

Abstract

Background

Concurrent blockade of LAG-3 may enhance efficacy of anti-programmed cell death-1 (PD-1) therapies. We previously presented safety and efficacy data from the phase 1 study in patients (pts) with advanced mel treated with anti-LAG-3 (fianlimab) in combination with anti-PD-1 (cemiplimab). We demonstrated high clinical activity among pts with anti–PD-1/PD-ligand (L)1-naive (expansion cohort [EC] 6 and 15) advanced mel. Among pts in EC6+EC15 (N=80), the objective response rate (ORR) was 63.8%, the disease control rate (DCR) was 80.0%, and the median duration of response (mDOR) was not reached (NR).

Factors associated with a poorer prognosis and reduced response to immunotherapy in pts with advanced mel include advanced stage of disease, elevated lactate dehydrogenase (LDH) levels, and metastasis sites, including liver or other visceral organs (M1c).

Methods

Pts with advanced mel were treated with fianlimab 1600 mg and cemiplimab 350 mg intravenously every 3 weeks for 12 months. In this subgroup analysis we evaluated fianlimab and cemiplimab in pts with advanced mel with poor prognostic features. ORR, DCR and mDOR are reported here for pts with liver mets at baseline (BL), LDH>upper limit of normal (ULN) at BL, and LDH>ULN at BL and M1c stage in pts in EC6+EC15.

Results

As of 1 July 2022, data cutoff date, 80 pts in EC6+EC15 (40 pts each) were treated with fianlimab and cemiplimab. For EC6+EC15 combined, at BL,19 pts (23.8%) had liver mets, 28 pts (35.0%) had LDH>ULN; and 13 pts (16.3%) had LDH>ULN at BL and any M1c.

In pts with liver mets at BL, the ORR for EC6+15 combined was 47.4%, the DCR was 63.2%, and the mDOR was 9.0 months (95% confidence interval [CI], 2.8–not evaluable [NE]). In pts with LDH>ULN at BL, the ORR for EC6+15 was 57.1%, the DCR was 71.4, and the mDOR was NR (95% CI, 7.3–NE). In pts with LDH>ULN at BL and any M1c, the ORR for EC6+15 was 53.8%, the DCR was 69.2%, and the mDOR was NR (95% CI, 5.7–NE).

Conclusions

Despite small numbers in subgroups, the efficacy analysis from EC6 and EC15 combined demonstrates high activity of fianlimab in combination with cemiplimab in pts with advanced mel and poor prognosis features at BL.

Clinical trial identification

NCT03005782

Editorial acknowledgement

Medical writing and editorial support provided by John G Facciponte, PhD, of Prime, Knutsford, UK, funded by Regeneron Pharmaceuticals, Inc., according to Good Publication Practice guidelines.

Collapse
Poster Display (ID 17) Poster Display

151P - Clinical outcome and preliminary immune analysis of Phase II clinical trial of combination of Tocilizumab with Ipilimumab and Nivolumab for patients with treatment naïve metastatic melanoma (ID 737)

Presentation Number
151P
Lecture Time
12:30 - 12:30
Speakers
  • E. Montazari (Houston, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • E. Montazari (Houston, United States of America)
  • N. Abdel-Wahab (Houston, United States of America)
  • D. Johnson (New Orleans, United States of America)
  • C. Spillson (Houston, United States of America)
  • K. Elsayes (Houston, United States of America)
  • F. Duan (Houston, United States of America)
  • S. Yadav (Houston, United States of America)
  • J. Allison (Houston, United States of America)
  • P. Sharma (Houston, United States of America)
  • A. Diab (Houston, United States of America)

Abstract

Background

Background: Immune related adverse events (irAEs) can possess a major challenge in patient symptom burden and risk of fatalities. In addition, it is a barrier for the development of multi-agent immunotherapy regimen. Management of these irAEs using corticosteroids as first-line therapy may have detrimental impact on cancer outcomes and worsen survival. We have previously demonstrated that interleukin-6 (IL-6) plays a role in the pathogenesis of irAEs in tumor resistance. Based on this data, we conducted this trial to assess the safety and efficacy of IL-6 blockade, using Tocilizumab, in combination with ipilimumab and nivolumab in melanoma patients.

Methods

Methods: Phase II clinical study to evaluate the safety and efficacy of the triplet therapy in melanoma (n=35). Pts received subcutaneous tocilizumab 162 mg bi-monthly for up to 12 weeks plus ipi 3mg/kg + nivo 1mg/kg every 3 weeks. The primary endpoints: 1) assess the frequency ofgrade 3/4 irAEs; 2) objective response rate (ORR); and 3) assess biomarker analysis from tumor, blood and inflamed tissue.

Results

Results: A total of 25 melanoma patients were enrolled; duration of treatment ranged from 6 to 41 weeks. Eleven patients (44%) had grade 3/4 irAEs; median time to onset was 8.3 weeks (2.7-17.3). This included colitis (20%), hepatitis (16%), pancreatitis (8%), and fibromyalgia rheumatica-like and type I diabetes (4% each) and led to study disconsolation in 4% but no treatment-related deaths. The ORR was 60% (44% in patients with elevated LDH). Our longitudinal immune analysis from tumor tissue demonstrates increase infiltration of immune cells Th1, Th17, and CD8 cells. Furthermore, gene expression data exhibits enrichment of IL-1, IL-6, and IL-17 pathways in patients with high grade toxicity post treatment.

Conclusions

Conclusions: Our preliminary data showed a trend to mitigate irAEs with the triplet therapy with no negative impact on tumor immunity. Furthermore, exploratory biomarker analysis suggests that we are not sufficiently blocking the IL-6/Th17 pathway with the current dosing of Tocilizumab. The protocol will enroll a new cohort of patients with increased Tocilizumab administration.

Collapse
Poster Display (ID 17) Poster Display

152P - A Phase I Study of HBM4003, an anti-CTLA-4 Heavy Chain Only Monoclonal Antibody, in Combination with Toripalimab in Advanced Melanoma (ID 423)

Presentation Number
152P
Lecture Time
12:30 - 12:30
Speakers
  • T. Bixia (Beijing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • T. Bixia (Beijing, China)
  • Z. Chi (Beijing, China)
  • Y. Chen (Fuzhou, China)
  • Y. Jiang (Chengdu, Sichuan, China)
  • M. Fang (Hangzhou, China)
  • Q. Gao (Zhengzhou, China)
  • G. Huang (Changsha, China)
  • X. Ren (Tianjin, China)
  • Y. Yao (Xi'an, China)
  • J. Chen (Wuhan, China)
  • X. Zhang (Guangzhou, China)
  • R. Li (Kunming, China)
  • G. Humphrey (Shanghai, China)
  • L. Ding (Shanghai, China)
  • Y. Geng (Shanghai, China)
  • S. Zhao (Shanghai, China)
  • Y. Yang (Shanghai, China)
  • Z. Lu (Shanghai, China)
  • D. Ye (Shanghai, China)
  • J. Guo (Beijing, China)

Abstract

Background

HBM4003 is a fully human heavy chain only monoclonal antibody targeting CTLA-4. In addition to blocking the CTLA-4 pathway, HBM4003 is also engineered to deplete Treg cells by enhanced antibody-dependent cellular cytotoxicity (ADCC) activity that was clinically validated. Here we reported updated results of a phase I study that evaluated HBM4003 plus toripalimab (anti-PD-1 antibody) in advanced melanoma.

Methods

This study includes two parts. In the dose-escalation part, patients with solid tumors received HBM4003 at 3 dose levels (0.03 mg/kg [n=1], 0.1 mg/kg [n=3], and 0.3 mg/kg [n=10]) plus toripalimab 240 mg every three weeks (Q3W). In the dose-expansion part, patients with advanced melanoma (n=26) received the recommended phase 2 dose (RP2D) of HBM4003 0.3 mg/kg plus toripalimab 240 mg Q3W. ClinicalTrials.gov number: NCT04727164.

Results

As of 27 Jun 2022, a total of 40 patients had been dosed. Median follow-up time was 106.5 days. Treatment-related adverse events (TRAEs) were reported in 85.0% (34/40) patients, and ≥Grade 3 TRAEs were reported in 20.0% (8/40) patients. The most commonly reported TRAE was rash (30.0%).

Patients with advanced melanoma treated with RP2D (including 8 patients in Part 1 and 26 patients in Part 2) were categorized as anti-PD-(L)1 naïve group (Cohort A, 17 patients) and anti-PD-(L)1 pretreated group (Cohort B, 17 patients). For cohort A, the ORR and DCR were 53.3% (95%CI: 26.6-78.7) and 73.3% (95%CI: 44.9-92.2) respectively in the 15 patients with post-treatment tumor assessment. The ORR of cutaneous, acral, mucosal and unknown subtype were 66.7% (2/3), 50% (2/4), 60.0% (3/5) and 33.3% (1/3), respectively. For Cohort B, the ORR and DCR were 11.8% (95%CI: 1.5-36.4) and 35.3% (95%CI: 14.2-61.7) respectively, including one patient achieving PR after pseudo-progression. Both of the PR cases were mucosal subtype.

Conclusions

HBM4003 0.3 mg/kg plus toripalimab 240mg Q3W showed promising anti-tumor activity in anti-PD-(L)1 naive patients with advanced melanoma including acral and mucosal subtypes, as well as an acceptable safety profile.

Collapse
Poster Display (ID 17) Poster Display

153P - Neoadjuvant therapy with tislelizumab plus chemotherapy followed by concurrent chemoradiotherapy in patients with stage Ⅳa nasopharyngeal carcinoma: A single-arm, phase II trial (ID 373)

Presentation Number
153P
Lecture Time
12:30 - 12:30
Speakers
  • Q. Zhang (Jiangmen, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Q. Zhang (Jiangmen, China)
  • M. Lai (Jiangmen, China)
  • F. Li (Jiangmen, China)
  • J. Chen (Jiangmen, China)
  • G. Chen (Jiangmen, China)

Abstract

Background

The effect of comprehensive treatment for locally advanced nasopharyngeal carcinoma (LANPC) is still unsatisfied, especially for stage Ⅳa NPC. 2 or 3 cycles of neoadjuvant therapy may not be enough. Immunotherapy has been reported to benefit patients with LANPC. Therefore, this study investigated the efficacy and safety of four cycles tislelizumab combined with neoadjuvant chemotherapy in treating stage Ⅳa NPC.

Methods

Patients with stage IVa NPC were enrolled and treated with neoadjuvant chemotherapy (gemcitabine ,1000 mg/m2, days 1 and 8, cisplatin ,80 mg/m2, day 1) and tislelizumab(200mg, day 1) every 3 weeks for 4 cycles followed by standard concurrent chemoradiotherapy. The primary endpoint was complete response(CR) rate after neoadjuvant treatment. Secondary endpoints included overall response rate(ORR) after neoadjuvant treatment, disease-free survival, safety and so on.

Results

From February 2022 to June 2022, 25 patients were enrolled at Jiangmen Central Hospital. As of Sep 15th 2022, the median follow-up time was 124.5 days. Overall, 24 patients were assessed by investigator according to RECIST v1.1 and 1 patient was withdrawn from the trial due to treatment of acute viral parotitis. All the 24 evaluable patients completed protocol-specified 4 cycles of therapy without delaying radiotherapy. 12 patients (50%) achieved CR after neoadjuvant therapy. The ORR of all evaluable patients was 91.7%. 13 (52%) of 25 patients had grade 3-4 treatment-related adverse events. Grade 1-2 immune-related adverse events(irAE) was recorded in 18 patients (72%). There were no grade 3-4 irAEs. Any grade of irAEs included hyperthyroidism, hypothyroidism, rash, and pruritus. A numerically higher CR rate of PD-L1 positive(TC≥1%)patients than PD-L1 negative(TC=0) patients (61% vs 0).

Conclusions

Four cycles tislelizumab in combination with neoadjuvant chemotherapy demonstrated a manageable safety profile and improved clinical response in high-risk LANPC patients. Patients with PD-L1 positive may be associated with favorable response. Long-term survival benefit will be followed continuously in this ongoing trial.

Clinical trial identification

ChiCTR2200056941

Collapse
Poster Display (ID 17) Poster Display

154P - Neoadjuvant chemotherapy plus tislelizumab followed by adjuvant tislelizumab for locoregionally advanced nasopharyngeal carcinoma (NPC): A single-arm, phase II trial (ID 377)

Presentation Number
154P
Lecture Time
12:30 - 12:30
Speakers
  • S. Sun (Bejing, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Sun (Bejing, China)
  • X. Huang (Beijing, China)
  • K. Wang (Beijing, China)
  • R. Wu (Bejing, China)
  • J. Wang (Bejing, China)
  • Y. Zhang (Bejing, China)
  • J. Zhang (Bejing, China)
  • X. Chen (Bejing, China)
  • Y. Qu (Bejing, China)
  • J. Luo (Bejing, China)
  • J. Yi (Beijing, China)
  • S. Zhou (Beijing, China)

Abstract

Background

Prognosis of NPC with T4 or N3 remains unsatisfactory due to high-risk of distant metastasis. More effective treatment strategies are needed for these patients. GP plus immune checkpoint blockade regimen has been shown to improve the survival in recurrent or metastatic NPC We investigated the efficacy and safety of neoadjuvant with GP plus tislelizumab followed by concurrent chemoradiotherapy, and adjuvant treatment with tislelizumab, an anti-PD-1 monoclonal antibody, in previously untreated T4 or N3 NPC.

Methods

In this phase II, single-arm study, eligible patients aged 18-70 yrs who were diagnosed with stage IVa (AJCC 8th) non-keratinizing nasopharyngeal received neoadjuvant therapy with gemcitabine (1000mg/m2 on day 1,8), cisplatin (25 mg/m2 on day 1-3) and tislelizumab (200mg) Q3W for 2 cycles followed by concurrent IMRT and cisplatin (100 mg/m2) Q3W during radiotherapy, then followed by adjuvant therapy with tislelizumab (200 mg) Q3W for 13 cycles. The primary endpoint was 2-years progression-free survival (PFS). The secondary endpoint included objective response rate (ORR), overall survival (OS), progression-free survival (PFS) and safety. This study planned to enroll 50 patients in 2 years.

Results

A total of 27 patients were enrolled from Sep. 2021 to Sep. 2022. 23 patients were evaluable for response assessment to neoadjuvant therapy with a median age of 49 (24-66) yrs and 82.6% (n=19) of male. According to RECIST1.1, 20 (87.0%) patients had objective response to neoadjuvant treatment, including 2 (8.7%) patient with CR, 18 (78.3%) patients with PR. The other 3 patients had SDa with a definition of tumor shrinkage occured and below a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. The incidence of adverse events of grade 3 or 4 was in 52.2% (12/23) patients, including nausea (7 [30.4%]) leukopenia (3 [13.0%]) neutropenia (2 [8.7%]) and hepatotoxicity (2 [8.7%]). Long-term efficacy is awaited.

Conclusions

Neoadjuvant with tislelizumab plus GP and adjuvant tislelizumab treatment achieved an impressive ORR with manageable toxicities. Further follow-up is needed to confirm the long-term efficacy.

Clinical trial identification

NCT05448885

Collapse
Poster Display (ID 17) Poster Display

155P - BNT113 + pembrolizumab as first-line treatment in patients with unresectable recurrent/metastatic HNSCC: Preliminary safety data from AHEAD-MERIT (ID 397)

Presentation Number
155P
Lecture Time
12:30 - 12:30
Speakers
  • K. Klinghammer (Berlin, Germany)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • K. Klinghammer (Berlin, Germany)
  • N. Saba (Atlanta, GA, United States of America)
  • E. Castelluci (Bronx, United States of America)
  • A. Colevas (Stanford, United States of America)
  • T. Rutkowski (Warsaw, Poland)
  • R. Greil (Salzburg, Austria)
  • D. Thurner (Graz, Austria)
  • U. Müller-Richter (Wuerzburg, Germany)
  • A. Di Giacomo (Siena, Italy)
  • J. Grewal (Louisville, United States of America)
  • C. Ottensmeier (Liverpool, United Kingdom)
  • A. Atasoy (Cambridge, United States of America)
  • S. Shpyro (Mainz, Germany)
  • P. Brück (Mainz, Germany)
  • J. Dias (Amadora, Portugal)
  • C. Ganser (Mainz, Germany)
  • Ö. Türeci (Mainz, Germany)
  • U. Sahin (Mainz, Germany)

Abstract

Background

BNT113 is an IV administered liposomal ribonucleic acid (RNA-LPX) cancer vaccine encoding the human papillomavirus (HPV)16 oncoproteins E6 and E7. Preclinically, PD‑L1 inhibition augments the anti‑tumor effect of RNA-LPX-based vaccines, supporting combining BNT113 + pembrolizumab.

Methods

AHEAD-MERIT is an open-label, randomized, Phase II trial of pembrolizumab (q3w) vs pembrolizumab + BNT113 (8xq1w, then q3w) as first-line treatment in patients (pts) with unresectable recurrent/metastatic HPV16 and PD-L1 positive HNSCC. Following a safety run-in of BNT113 + pembrolizumab, pts are randomized 1:1 to BNT113 + pembrolizumab or pembrolizumab. Primary endpoints: safety, OS, and ORR.

Results

As of 05 July 2022, of 15 treated pts, 12 had completed the safety run-in (pembrolizumab + 4 BNT113 doses). In the treated pts, the median age was 66 yrs (range: 41–74) and all were male. All pts in the safety run-in had ≥1 AE, the most frequent were pyrexia (8 pts) and chills (6 pts). There were 3 pts (25%) that had Grade ≥3 AEs with all classed as SAEs (pyrexia, hypercalcemia, pleural effusion, shaking/rigors). In 2/3 pts, the AEs (pyrexia, shaking/rigors) were considered related. No deaths were reported.

Conclusions

Safety was acceptable and in line with the safety profile of BNT113 and pembrolizumab as single agents; no new safety signals were observed for the combination. The randomized part of the trial is ongoing.

Clinical trial identification

BNT113-01: NCT04534205; EudraCT: 2020-001400-41.

Editorial acknowledgement

The authors would like to acknowledge Frank Smeets for clinical trial support and Camilla West (BioNTech SE) for medical writing support.

Collapse
Poster Display (ID 17) Poster Display

156P - A Phase 1b/2 Study of Nanatinostat (Nstat) Plus Valganciclovir (VGCV) in Advanced Epstein-Barr Virus Positive (EBV+) Solid Tumors and with Pembrolizumab (PEM) in Recurrent/Metastatic Nasopharyngeal Carcinoma (RM-NPC) (ID 448)

Presentation Number
156P
Lecture Time
12:30 - 12:30
Speakers
  • A. Colevas (Stanford, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Colevas (Stanford, United States of America)
  • L. Siu (Toronto, Ontario, Canada)
  • D. Lim (Singapore, Singapore)
  • B. Gao (Blacktown, NSW, Australia)
  • S. Khan (Stanford, United States of America)
  • L. Eng (Toronto, Ontario, Canada)
  • P. Voon (Kuching, Malaysia)
  • M. Ahn (Seoul, Korea, Republic of)
  • A. Elguindy (Cardiff-by-the-Sea, United States of America)
  • A. Katkov (Cardiff-by-the-Sea, United States of America)
  • L. Rojkjaer (Cardiff-by-the-Sea, United States of America)
  • Y. Katz (Cardiff-by-the-Sea, United States of America)
  • B. Ma (Sha Tin, Hong Kong PRC)

Abstract

Background

EBV is associated with NPC, with the virus in a latent state. Nstat is a Class-I selective oral HDAC inhibitor that induces expression of the lytic BGLF4 EBV protein kinase in EBV+ tumor cells, activating ganciclovir (GCV) via phosphorylation. This results in GCV-mediated inhibition of viral and cellular DNA synthesis and apoptosis. Nstat plus VGCV (oral prodrug of GCV) showed a favorable safety profile with anti-tumor activity in a phase 1b/2 study in R/R EBV+ lymphoma (recommended phase 2 dose [RP2D]: Nstat 20 mg daily [QD], 4 d/wk + VGCV 900 mg QD). Here, phase 1b uses a 3+3 dose escalation for Nstat + VGCV RP2D selection in RM-NPC, followed by an expansion at the RP2D in other EBV+ solid tumors. In phase 2, up to 60 RM-NPC pts will be randomized 1:1 to receive Nstat + VGCV at the RP2D +/- PEM. Herein we report preliminary safety results from phase 1b in RM-NPC (NCT05166577).

Methods

Pts aged ≥18 with EBV+ RM-NPC (1-3 prior therapies) with measurable disease (RECIST v1.1) and no curative options receive daily Nstat 20-40 mg 4d per wk with VGCV 900-1800 mg daily in phase 1b. Primary endpoints are incidence of dose limiting toxicities (DLTs) (phase 1b) and overall response rate (phase 2); secondary endpoints include duration of response, disease control rate, progression free survival and overall survival. Responses are assessed per RECIST v1.1 from week 8.

Results

As of 15-Sept-22, 7 male pts (median age 51y [19-61y]) were enrolled; 3 in dose level (DL) 1 and 4 in DL2 received Nstat 20 and 30 mg QD, respectively, 4 d/wk, plus VGCV 900 mg QD. Median no. prior systemic therapies was 2; all pts were refractory to last therapy with bone (6/7), liver (5/7), and lung (3/7) metastases. Related AEs were all G1-2, most commonly fatigue, nausea, and increased creatinine (n=3 each); no G3+ AEs/DLTs and 1 SAE (cancer pain) were reported. Of 6 pts evaluable for response, 2 had SD (with decreasing/stable plasma EBV DNA, pEBVd); 4 had PD (with rising pEBVd).

Conclusions

The combination of Nstat and VGCV represents a novel approach for the treatment of EBV+ NPC and is tolerated at doses exceeding the RP2D for lymphoma. Dose escalation continues.

Clinical trial identification

NCT05166577

Collapse
Poster Display (ID 17) Poster Display

157P - Phase 1 expansion of IMC-C103C, a MAGE-A4×CD3 ImmTAC bispecific protein, in ovarian carcinoma (ID 288)

Presentation Number
157P
Lecture Time
12:30 - 12:30
Speakers
  • R. Sweis (Chicago, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • R. Sweis (Chicago, United States of America)
  • E. Garralda (Barcelona, Spain)
  • O. Saavedra Santa Gadea (Barcelona, Spain)
  • K. Moore (Oklahoma City, Oklahoma, United States of America)
  • D. Davar (Pittsburgh, United States of America)
  • O. Hamid (Los Angeles, CA, United States of America)
  • N. Segal (New York, NY, United States of America)
  • T. Evans (Glasgow, United Kingdom)
  • M. Dar (Conshohocken, United States of America)
  • Y. Yuan (Conshohocken, United States of America)
  • L. Collins (Abingdon-on-Thames, United Kingdom)
  • P. Kirk (Abingdon-on-Thames, United Kingdom)
  • O. Karakuzu (Conshohocken, United States of America)
  • J. Lopez (Sutton, United Kingdom)
  • I. Melero (Pamplona, Spain)

Abstract

Background

ImmTAC® bispecific proteins redirect polyclonal T cells to target intra/extracellular proteins using a T cell receptor as targeting domain. IMC‑C103C (MAGE‑A4 × CD3) is an ImmTAC against MAGE‑A4, which is expressed in several tumors including ovarian carcinoma (OC). ≥ 90 µg IMC‑C103C was demonstrated as the clinically active range (Davar 2021). Pre-selection for MAGE‑A4 expression was not required as a majority of OC express MAGE‑A4 (H score ≥ 1) and tebentafusp demonstrated OS benefit and ctDNA reductions regardless of H score, although RECIST responses were enriched at higher H scores (Leach 2021).

Methods

HLA‑A*02:01+ OC pts were enrolled as all-comers in escalation and expansion cohorts with MAGE‑A4 expression tested by IHC retrospectively. Ph1 primary objective is to identify expansion dose. Other objectives: adverse events (AE), ORR (RECIST v1.1), OS and biomarkers including ctDNA (Guardant 360).

Results

As of 24Jun2022, 31 heavily pretreated OC pts (87% platinum resistant) received 90 - 240 µg IMC‑C103C. Of 28 OC pts with evaluable MAGE‑A4 IHC expression, 16 (57%) were positive (pos) with median H score 28 (maximum 300). 12/28 (43%) pts had H score = 0 (neg). Only 1 pt had H score > 130.

The AE profile is consistent with previously reported (Davar 2021). Most common related AEs were pyrexia (68%), chills (52%), and CRS (45%), the vast majority were Grade 1/2. No related AEs resulted in treatment discontinuation or death.

27 of 31 were response evaluable. 15 pts were MAGE‑A4 pos; 1 had confirmed PR (duration 12+ months) and 5 had SD. 6/15 pts had any tumor shrinkage. Of 12 MAGE‑A4 neg/unknown (unk) pts, 1 had SD and 1 had any tumor shrinkage.

Of 21 ctDNA evaluable pts, 4/11 MAGE‑A4 pos and 1/10 MAGE‑A4 neg/unk had ≥ 50% ctDNA reduction.

After first dose, serum IFNγ and TNFα were not induced in MAGE‑A4 neg pts or minimally induced in MAGE‑A4 pos pts, consistent with mostly low H scores.

Conclusions

When enrolled as all-comers, the vast majority of heavily pre-treated OC pts had either zero or very low MAGE‑A4 expression. IMC-C103C is clinically active but the low MAGE expression may have resulted in few RECIST responses. Dose optimization and signal detection continue in MAGE‑A4 pos pts with additional tumor types.

Clinical trial identification

NCT03973333

Collapse
Poster Display (ID 17) Poster Display

158P - Triple blockade of the DNAM-axis with COM701 + BMS-986207 + nivolumab demonstrates preliminary antitumor activity in patients with platinum resistant OVCA. (ID 420)

Presentation Number
158P
Lecture Time
12:30 - 12:30
Speakers
  • J. Moroney (Chicago, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Moroney (Chicago, United States of America)
  • O. Yeku (Boston, United States of America)
  • G. Fleming (Chicago, IL, United States of America)
  • L. Emens (Pittsburgh, United States of America)
  • D. Vaena (Germantown, United States of America)
  • E. Dumbrava (Houston, TX, United States of America)
  • D. Rasco (San Antonio, United States of America)
  • M. Sharma (Grand Rapids, United States of America)
  • K. Papadopoulos (San Antonio, TX, United States of America)
  • A. Patnaik (San Antonio, United States of America)
  • R. Sullivan (Boston, MA, United States of America)
  • H. Adewoye (South San Francisco, United States of America)
  • E. Ophir (Lausanne, Switzerland)
  • G. Cojocaru (Holon, Israel)
  • P. Ferre (Boulogne-Billancourt, France)
  • B. Izar (New York, United States of America)
  • S. Gaillard (Baltimore, United States of America)

Abstract

Background

Treatment [tx] options for platinum resistant ovarian cancer [PROC] are limited [ltd]. Immune checkpoint inhibitors (ICI) have ltd activity in PROC. Clinical studies evaluating novel therapies are urgently needed. COM701, a novel, 1st in-class ICI binds to PVRIG, leading to activation of T-cells. BMS-986207 is an ICI blocker of TIGIT. We reported a partial response [PR] with COM701 monotherapy in a pt with primary peritoneal CA1. We hypothesized that in pts with PROC, blocking the DNAM axis with the triplet: COM701 + BMS-986207 + nivolumab, would demonstrate antitumor activity with a favorable safety and tolerability profile. We present preliminary results.

Methods

All 20 pts enrolled received COM701 20 mg/kg + BMS-986207 480 mg + nivolumab 480 mg IV Q4W. Primary objectives [obj] were safety/tolerability; secondary obj of antitumor activity. Key inclusion criteria: Age ≥ 18 yrs, histologically confirmed advanced malignancies and exhausted all available standard tx. Key exclusion criteria: prior receipt of any inhibitor of PVRIG, TIGIT, or PD-1/PD-L1. Investigator assessed responses per RECIST v1.1, safety per CTCAE v5.0.

Results

Median [med] age 61yr, med follow-up 51 days [range 1-202], med number of prior lines of therapy - 4 [range 1-10]. Objective response rate 4/20 [20%] pts [all ongoing study tx, 3 confirmed PR], 3 PRs - serous adenoCA, 1 PR - clear cell histology. No complete responses (CR); 4pts with stable disease (SD), disease control rate [CR+PR+SD] 8/20 [40%]. Most frequent [freq] histology - serous adenoCA 10/20 [50%], clear cell 3/20 [15%]. Most freq AE G1/2 fatigue in 11 pts. A sustained immune activation induced by the tx, with a maximum 7.6-fold average increase of peripheral IFNɣ in 16pts evaluated [p<0.005].

Conclusions

The combination of COM701 + BMS-986207 + nivolumab has encouraging signal of antitumor activity with immune activation in pts with heavily pre-treated PROC and is well tolerated. Additional data will be presented at the conference. Data extract 08/29/2022.

1. Vaena D et al, COM701±nivolumab: Results of an ongoing P1 study of safety, tolerability & preliminary antitumor activity in pts with advanced solid malig. J Clin Onco 39, 2021 (suppl 15; abst 2504).

Clinical trial identification

NCT04570839.

Collapse
Poster Display (ID 17) Poster Display

159P - COM701 in combination with nivolumab demonstrates preliminary antitumor activity in patients with platinum resistant epithelial ovarian cancer. (ID 338)

Presentation Number
159P
Lecture Time
12:30 - 12:30
Speakers
  • O. Yeku (Boston, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • O. Yeku (Boston, United States of America)
  • D. Shepard (Cleveland, United States of America)
  • M. Patel (Fort Myers, United States of America)
  • G. Fleming (Chicago, IL, United States of America)
  • D. Vaena (Germantown, United States of America)
  • D. Rasco (San Antonio, United States of America)
  • B. Chmielowski (Los Angeles, United States of America)
  • M. Sharma (Grand Rapids, United States of America)
  • E. Hamilton (Nashville, United States of America)
  • R. Sullivan (Boston, MA, United States of America)
  • K. Papadopoulos (San Antonio, TX, United States of America)
  • B. Izar (New York, United States of America)
  • G. Cojocaru (Holon, Israel)
  • E. Ophir (Lausanne, Switzerland)
  • P. Ferre (Boulogne-Billancourt, France)
  • E. Dumbrava (Houston, TX, United States of America)

Abstract

Background

There is a high unmet medical need for the treatment of patients [pts] with platinum resistant epithelial ovarian cancer [PROC]. Immune checkpoint inhibitors (ICI) have limited activity in this pt population. COM701 is a novel, 1st-in-class ICI that binds to PVRIG, a DNAM-1 axis member, leading to activation of T-and NK-cells. We hypothesized that in pts with PROC, dual blockade of PVRIG and PD1 would demonstrate antitumor activity with a favorable safety and tolerability profile. We present encouraging preliminary results.

Methods

We enrolled 20 patients [pts] with PROC. All pts received COM701 20 mg/kg + nivolumab 480 mg both IV Q4W. Primary objectives were safety/tolerability, with secondary objective of preliminary antitumor activity. Key inclusion criteria: Age ≥ 18 yrs, histologically confirmed locally advanced or metastatic solid malignancy and has exhausted all available standard therapy. Key exclusion criteria: prior receipt of anti-PVRIG, anti-TIGIT, no limitation on the number of prior lines of therapy or prior PD-1/PD-L1 inhibitor. Investigator assessed responses were per RECIST v1.1, safety per CTCAE v4.03.

Results

Median age 61.5yrs, median of 6 prior lines of therapy [Min, Max: 2,9]. Objective response rate (ORR) of 2/20 [10%] pts: 1 pt with fallopian tube CA, 6 prior lines of therapy, clear cell histology; 1 pt with OVCA, serous adenoCA, 7 prior lines of therapy [including prior nivolumab with best response of progressive disease]; both subjects with PR have an ongoing response to therapy, no complete responses (CR); 5 pts with stable disease (SD). Disease control rate [CR + PR + SD] 7/20 [35%]. Most frequent AEs were G1/2 nausea 11 pts, fatigue 11 pts [all G1/2]. Increase of serum IFNg was observed, confirming the expected immune activation induced by COM701 given in combination with nivolumab.

Conclusions

COM701 + nivolumab demonstrates encouraging preliminary signal of antitumor activity and immune activation in pts with heavily pre-treated PROC with a favorable safety/tolerability profile. Additional data analyses and pt followup are ongoing and will be presented at the conference. Data extract 09/18/2022.

Clinical trial identification

NCT03667716.

Collapse
Poster Display (ID 17) Poster Display

160P - COLUMBIA-1: A Phase Ib/II, Open-Label, Randomized, Multicenter Study of Durvalumab plus Oleclumab in Combination with Chemotherapy and Bevacizumab as First-Line (1L) Therapy in Metastatic Microsatellite-Stable Colorectal Cancer (MSS-mCRC) (ID 474)

Presentation Number
160P
Lecture Time
12:30 - 12:30
Speakers
  • N. Segal (New York, NY, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • N. Segal (New York, NY, United States of America)
  • J. Tie (Melbourne, VIC, Australia)
  • S. Kopetz (Houston, United States of America)
  • M. Ducreux (Villejuif, CEDEX, France)
  • E. Chen (Toronto, Canada)
  • R. Dienstmann (Sao Paulo, Brazil)
  • A. Hollebecque (Villejuif, Cedex, France)
  • M. Reilley (Charlottesville, United States of America)
  • M. Elez Fernandez (Barcelona, Spain)
  • J. Cosaert (Zoetermeer, NJ, Netherlands)
  • J. Cain (Phoenix, United States of America)
  • M. Hernandez (Gaithersburg, United States of America)
  • N. Hewson (Northolt, United Kingdom)
  • Z. Cooper (Phoenix, United States of America)
  • M. Dressman (Northolt, United Kingdom)
  • J. Tabernero (Barcelona, Spain)

Abstract

Background

Despite advances in the treatment of mCRC combining chemotherapy regimens with biologics, most patients (pts) still progress within 11 months of receiving 1L chemotherapy. Addition of novel therapies to the standard of care (SoC) to improve antitumor activity is urgently needed. The randomized part 2 of COLUMBIA-1 (NCT04068610) evaluated the safety and efficacy of combining SoC (bevacizumab [BEV] + FOLFOX) with the PD-L1 inhibitor durvalumab (D) and the anti-CD73 monoclonal antibody oleclumab (O).

Methods

Pts with previously untreated, MSS-mCRC and ECOG PS ≤1 received either SoC alone or SoC + D (1500 mg, Q4W) + O (3000 mg Q2W x4 then Q4W) in the experimental arm (EXP). The primary endpoint was objective response rate (ORR) per investigator assessed RECIST v1.1.

Results

As of 10 Dec 2021, 52 pts were enrolled, of whom 51 were response evaluable. The confirmed ORR with SoC was 44.0% (95% confidence interval [CI], 24.4–65.1%) compared to 61.5% (95% CI, 40.6–79.8%) in the EXP arm. Median OS was not reached (SoC) vs 19.1 mos (EXP); median PFS was 11.1 mos (SoC) vs 10.9 mos (EXP; Table). Grade ≥3 treatment emergent adverse events (TEAEs) occurred in 76.9% of pts in SoC and 65.4% EXP. Fatal TEAEs (all unrelated) were observed in 3 pts in the EXP arm: 1 with sepsis and 2 with intestinal perforation. One pt with intestinal perforation deemed related to BEV experienced fatal peritonitis. In the SoC arm, there was a single fatal COVID-19 TEAE. The most frequent treatment-related AEs in the EXP arm were diarrhea (38.5%), peripheral sensory neuropathy (38.5%) and fatigue (26.9%). There was no identified association between CD73 expression and clinical benefit.

Conclusions

Addition of D + O to FOLFOX + BEV SoC showed a moderate response increase without PFS benefit vs SoC alone. Safety was consistent with known safety profiles.

RECIST v1.1

Response Evaluable Population

SoC (n=25)

EXP (n=26)

Best Overall Response, n (%)

Confirmed CR

0

1 (3.8)

Confirmed PR

11 (44.0)

15 (57.7)

SD

12 (48.0)

7 (26.9)

Unconfirmed PR

3 (12.0)

1 (3.8)

PD

1 (4.0)

2 (7.7)

Unknown/not evaluable

1 (4.0)

1 (3.8)

ORR, n (%) [95% CI %]

11 (44) [24.4, 65.1]

16 (61.5) [40.6, 79.8]

Disease Control, n (%) [95% CI %]

23 (92.0) [74.0, 99.0]

23 (88.5) [69.8, 97.6]

Median DoR, mos (95% CI %)

7.6 (3.7, NE)

10.3 (5.8, 14.9)

Median PFS, mos (95% CI %)**

11.1 (7.3, 19.3)

10.9 (6.9, 15.1)

PFS rate at 12 mos, % (95% CI %)**

35.9 (16.2, 56.2)

36.1 (17.1, 55.5)

Median OS, mos (95% CI %)**

NR (18.2, NE)

19.1 (10.6, NE)

*NE=Not Estimable; **ITT Population: SoC (n=26), EXP (n=26)

Clinical trial identification

NCT04068610

Editorial acknowledgement

Editing support for this abstract, under the direction of the authors, was provided by Catherine Crookes of Ashfield MedComms (Macclesfield, UK), an Inizio company, and was funded by AstraZeneca

Collapse
Poster Display (ID 17) Poster Display

161P - A Phase 1b Study to Evaluate IK-007 in Combination with Pembrolizumab (pembro) in Patients with Advanced Microsatellite Stable (MSS) Colorectal Cancer (CRC) (ID 634)

Presentation Number
161P
Lecture Time
12:30 - 12:30
Speakers
  • A. Tolcher (San Antonio, TX, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Tolcher (San Antonio, TX, United States of America)
  • D. Spigel (Nashville, TN, United States of America)
  • A. Leal (Aurora, United States of America)
  • T. Bekaii-Saab (Phoenix, United States of America)
  • T. Lingaraj (Boston, United States of America)
  • E. Talcove-Berko (Boston, United States of America)
  • N. Ortiz-Otero (Boston, United States of America)
  • W. Bartolini (Boston, United States of America)
  • K. Kacena (Boston, United States of America)
  • M. Chisamore (Whitehouse Station, United States of America)
  • S. Santillana (Boston, MA, United States of America)

Abstract

Background

IK-007 is a selective oral inhibitor of prostaglandin (PG) E2 receptor 4 (EP4). EP4 plays a key role in mediating PGE2-dependent immunosuppression and IK-007 may overcome mechanisms driving CPI resistance. MSS CRC is primary refractory to checkpoint inhibitors and high levels of PGE2 metabolite (PGEM) are associated with poor prognosis of CRC and may be predictive of response.

Methods

This is a Phase 1b study of IK-007 in combination with pembro in patients with advanced or metastatic MSS CRC who have received at least 2 lines of prior therapy. Cohort 1 had a 7-day run-in with single agent IK-007 followed by IK-007 in combination with pembro (200 mg IV every 3 weeks), Cohort 2 received combination only. Endpoints included safety (mTPI design), antitumor activity (RECIST 1.1), pharmacokinetics, pharmacodynamics, and RP2D.

Results

Study enrolled 54 patients: 26 in Cohort 1 and 28 in Cohort 2 at 4 dose levels (300, 450, 600, and 900mg BID PO) with 12 patients prospectively enrolled with high urinary PGEM (enrichment). Median age was 70 years (28-82). Events of clinical interest included decreased renal function, liver toxicity, and upper GI ulcers. Treatment-related adverse events ≥ Grade 2 were reported in 26 patients (48%). Of these, increased AST was seen in 5 patients (9.3%). Serious adverse events (SAEs) were reported in 23 patients (43%) including intestinal obstruction, pneumothorax, and acute kidney injury. Two unrelated Grade 5 SAEs (cardiac arrest and aspiration pneumonia) were reported. A 5.3% ORR (2/39) was observed in the response population: 1 cCR (300mg BID) with DoR of 18+ months and 1 cPR (900mg Q12h) with DoR 4+ months (ongoing). Disease control rate was 28% (11/39) with the median duration of response of 8.3 months in the 5 patients with prolonged clinical benefit.

Conclusions

IK-007 in combination with pembro had an acceptable safety profile and showed limited antitumor activity in heavily pretreated MSS CRC patients. Correlation of response with PGEM status was inconclusive. Of note, 28% achieved disease control, many showing prolonged clinical benefit. Investigation in additional indication is warranted, including the ongoing IIT in inflammatory breast cancer.

Clinical trial identification

NCT03658772

Collapse
Poster Display (ID 17) Poster Display

162P - Efficacy and safety of GEMOX (Gemcitabine plus Oxaliplatin) plus Sintilimab and Bevacizumab as a conversion therapy in patients with initially unresectable biliary tract cancers (BTC): A single-arm, phase II study (ID 454)

Presentation Number
162P
Lecture Time
12:30 - 12:30
Speakers
  • N. Zhang (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • N. Zhang (Shanghai, China)
  • J. Zhou (Shanghai, China)
  • L. Wang (Shanghai, China)
  • T. Zhang (Shanghai, China)
  • W. Zhu (Shanghai, China)
  • A. Mao (Shanghai, China)
  • Q. Pan (Shanghai, China)
  • Z. Lin (Shanghai, China)
  • M. Wang (Shanghai, China)
  • Y. Zhang (Shanghai, China)
  • Y. Feng (Shanghai, China)
  • W. Xu (Shanghai, China)
  • Y. Zhao (Shanghai, China)
  • L. Wang (Shanghai, China)

Abstract

Background

Gemcitabine-based chemotherapy and durvalumab plus GC are currently the standard regimens for advanced biliary tract cancers (BTC). Further, chemotherapy combined with PD-1 inhibitor and Bevacizumab improved the survival benefits in lung cancer. The effectiveness and safety of GEMOX plus Sintilimab and Bevacizumab in treating initially unresectable BTC are being evaluated in this study.

Methods

Treatment-naïve patients (pts) with histologically or cytologically confirmed unresectable BTC (TanyN1M0) were enrolled. GEMOX (Gemcitabine 1000mg/m2, iv, d1,8; Oxaliplatin 100mg/m2, iv, d1) plus Sintilimab (200mg, iv, d1) and Bevacizumab (7.5mg/kg, iv, d1) were given every 3 weeks for 5 cycles. pts with partial response (PR) or complete response (CR) and eligible for R0 resection were referred for surgery. After treatment for 5 cycles, subsequent therapy was recommended by the investigator. Tumor response and resectability were assessed every 3 weeks according to RECIST v1.1. The primary endpoint was objective response rate (ORR), and secondary endpoints included surgical conversion, disease control rate (DCR), progression-free survival (PFS), overall survival (OS) and safety.

Results

Between 08/2020 and 6/2022, 37 pts were enrolled with a median age of 63 years (range 36-78), 49% were male, and 76% had an ECOG PS of 1. Of them, 23 pts (62%) were ICC, 4 pts (11%) were ECC and 10 pts (27%) were GBC. All of the pts were diagnosed with stage III. At a median follow-up duration of 12.7 months (range 3.1-22.0), ORR was 49% in 37 evaluable patients and DCR was 86%. 1 pts achieved CR, and 17 pts achieved PR. 9 pts met the pre-designated criteria for surgery and 3 pts received surgery. Median PFS was 9.82 months (95% CI 5.19-NA), and median OS was 12.6 months (95% CI 9.5-13.9). Most treatment-related AEs (TRAEs) were Grade 1-2, and the incidence of grade 3-4 TRAEs was 35%. The most common TRAEs were asthenia (75%), peripheral neuropathy (46%), and nausea (35%).

Conclusions

Given the encouraging ORR and tolerable safety, the combination of GEMOX plus Sintilimab and Bevacizumab as a conversion therapy may be a new option for initially unresectable BTC.

Collapse
Poster Display (ID 17) Poster Display

163P - Neoadjuvant Chemoradiotherapy plus Tislelizumab Followed by Surgery for Esophageal Carcinoma: An Interim Analysis of the Prospective, Single-arm, Phase II Trial (ID 663)

Presentation Number
163P
Lecture Time
12:30 - 12:30
Speakers
  • J. Yang (Wuhan, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Yang (Wuhan, China)
  • Z. Zhang (Wuhan, China)
  • B. Wu (Wuhan, China)
  • Y. Qin (Wuhan, China)
  • J. Wei (Wuhan, China)
  • Y. Qu (Wuhan, China)
  • Q. Sun (Wuhan, China)
  • K. Jiang (Wuhan, China)
  • K. Yang (Wuhan, China)

Abstract

Background

This study aimed to evaluate the safety and efficacy of neoadjuvant chemoradiotherapy (nCRT) combined with sequential tislelizumab followed by surgery for resectable thoracic esophageal squamous cell carcinoma (TESCC).

Methods

Patients with pathologically confirmed TESCC and clinical T1-2N1-3M0 or T3-4aN0-3M0 were allocated to receive neoadjuvant radiotherapy (41.4 Gy in 23 fractions) with concurrent chemotherapy (albumin-bound paclitaxel, 50-100 mg/m2, once weekly for five weeks; carboplatin, area under the curve of 2 mg/mL/min, once weekly for five weeks) plus sequential tislelizumab (200 mg every three weeks for three cycles, beginning within the first to 14th day after completion of radiotherapy) followed by subtotal esophagectomy with two-field lymphadenectomy. The primary endpoints were safety and pCR rate after surgery. The second endpoints included radical (R0) resection and major pathological response (MPR) rate. The trial was registered with ClinicalTrials.gov (NCT04776590).

Results

From January 2021 to June 2022, 26 eligible patients were enrolled. Eighteen patients completed neoadjuvant Tislelizumab and 15 underwent planed surgery. R0 resection rate was 100%. pCR rate for primary tumor and resected lymph nodes was 46.7% (7/15). MPR rate for primary tumor was 86.7% (13/15). During neoadjuvant Tislelizumab, no ≥ grade 3 adverse events (AEs) occurred and grade 1–2 AEs developed in 88.9% (16/18) of the patients, including weakness (66.7%, 12/18), chest pain (61.1%, 11/18), pulmonary infection (PI) (33.3%, 6/18) and radiation-induced lung injury (33.3%, 6/18). Grade 3 postoperative complications occurred in 20.0% (3/15) of the patients, including anastomotic fistula (20.0%, 3/15), injury of recurrent laryngeal nerve (6.7%, 1/15) and pleural effusion (PE) (6.7%, 1/15). And grade 1-2 postoperative complications occurred in 80.0% (12/15) of the patients, including anemia (46.7%, 7/15), PI (26.7%, 4/15), PE (26.7%, 4/15) and liver malfunction (13.3%, 2/15).

Conclusions

nCRT combined with sequential tislelizumab followed by surgery may be safe and effective for resectable TESCC and be worthy of further study.

Clinical trial identification

NCT04776590

Collapse
Poster Display (ID 17) Poster Display

164P - TQB2450 plus anlotinib combined with paclitaxel and cisplatin as first-line treatment of advanced esophageal squamous cell carcinoma (ESCC): a single-arm, multicenter phase Ⅱ trial (ID 453)

Presentation Number
164P
Lecture Time
12:30 - 12:30
Speakers
  • J. Wang (Anyang, Henan, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Wang (Anyang, Henan, China)
  • N. Li (Zhengzhou, China)
  • Y. Guo (Luoyang, China)
  • Y. Cheng (Jinan, China)
  • B. Li (Jinan,China, China)
  • S. Luo (Zhengzhou, China)

Abstract

Background

Immunotherapy combined with chemotherapy as first-line treatment in advanced ESCC has been the standard of care. Previously results demonstrated anlotinib, whether combined with chemotherapy as first-line regimen or monotherapy as second-line treatment, had encouraging efficacy and manageable toxicity in advanced ESCC. TQB2450 is a humanized anti-PD-L1 monoclonal antibody, which could recovery T-cells activity and enhance immune responses by preventing the binding of PD-L1 to PD-1 and B7.1 receptors on the surface of T cells. Here, we conducted a phase II trial to evaluate the efficacy and safety of anlotinib combined with TQB2450, cisplatin and paclitaxel as first-line therapy in advanced ESCC.

Methods

Eligible patients (pts) with advanced ESCC who had not previous systemic therapy received TQB2450 (1200mg, iv, d1, q3w) plus anlotinib (10mg, po, d1~14, q3w) combined with paclitaxel (135mg/m2, iv, d1, q3w) and cisplatin (60~75mg/m2, iv, d1~3, q3w) for 4 to 6 cycles as initial therapy. Patients, who did not have progressive disease (PD), continued received anlotinib (10mg, po, d1~14, q3w) and TQB2450 (1200mg, iv, d1, q3w) as maintenance treatment until PD or unacceptable toxicity. The primary endpoint was PFS. Secondary endpoints were iPFS (iRECIST), ORR (RECIST 1.1), DCR, DOR and safety.

Results

As of 31 May, 2022, 38 pts were enrolled with a median age of 64 years (range 41-74), male (78.9%) and ECOG PS 1 (78.9%). Of 25 evaluable pts, 1 pts had complete response, 20 pts reached partial response and 4 pts had stable disease. The ORR was 84.0% (95%Cl 63.9%-95.5%) and the DCR was 100.0% (95%Cl: 86.3%-100.0%). Median PFS was not reached. ≥ Grade 3 treatment-related adverse events (TRAEs) was 57.9% (22/38), which mainly included neutropenia (42.1%), leukopenia (18.4%) and hypertension (18.4%). 11 pts (28.9%) suffered from serious AEs. AEs led to discontinuation of TQB2450 in 7.9% and anlotinib in 7.9% of enrolled patients.

Conclusions

TQB2450 plus anlotinib combined with paclitaxel and cisplatin showed significant efficacy and manageable toxicities as first-line treatment in advanced ESCC, which might provide a new treatment strategy for those.

Clinical trial identification

NCT05013697

Collapse
Poster Display (ID 17) Poster Display

165P - Efficacy and safety of tislelizumab (TIS) plus lenvatinib (LEN) as first-line treatment in patients (pts) with unresectable hepatocellular carcinoma (uHCC): a single-arm, multicenter, phase II trial (ID 393)

Presentation Number
165P
Lecture Time
12:30 - 12:30
Speakers
  • L. Xu (Guangzhou, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Xu (Guangzhou, China)
  • J. Chen (Guangzhou, China)
  • J. Yang (Chengdu, China)
  • W. Gong (Shanghai, China)
  • Y. Zhang (Harbin, China)
  • H. Zhao (Beijing, China)
  • S. Yan (Hangzhou, China)
  • W. Jia (Hefei, China)
  • Z. Wu (Xi'an, China)
  • C. Liu (Chengdu, China)
  • X. Song (Shanghai, China)
  • Y. Ma (Harbin, China)
  • X. Yang (Beijing, China)
  • Z. Gao (Hangzhou, China)
  • N. Zhang (Hefei, China)
  • X. Zheng (Xi' An, China)
  • M. Li (Beijing, China)
  • X. Zhang (Shanghai, China)
  • M. Chen (Guangzhou, China)

Abstract

Background

The potential advantage of combining anti-PD-1 antibodies with tyrosine kinase inhibitors has been revealed by several trials in advanced HCC. TIS, an anti-PD-1 monoclonal antibody, has shown anti-tumor activity in HCC. LEN is a multikinase inhibitor approved for the first-line treatment of uHCC. This study aims to evaluate the efficacy and safety of TIS plus LEN in pts with uHCC.

Methods

Systemic treatment-naïve pts with uHCC received TIS (200 mg, IV, Q3W) and LEN (body weight ≥60 kg: 12 mg; <60 kg: 8 mg, PO, QD). Tolerability evaluated by assessing dose-limiting toxicities (DLTs) in the first 6 pts (Part 1) was the premise of the remaining enrollment (Part 2). Primary endpoint was objective response rate (ORR) assessed by Independent Review Committee (IRC) per RECIST v1.1. Based on the Simon’s two-stage design, >6 responders were needed in stage 1 (n=30) to continue, and ≥18 responders were needed by the end of stage 2 (n=60) to claim statistical superiority to a historical control of 18.8% (from LEN arm of phase III REFLECT study) (α and β errors of 0.05).

Results

A total of 64 pts were enrolled and received TIS plus LEN (Part 1, n=6; Part 2, n=58) with 73.4% in BCLC stage C. As of 7 July 2022 (median follow-up, 12.5 months), 21.9% pts were on treatment. No DLTs were observed in the first 6 pts. The study met the statistical superiority criteria. There were 24 responders assessed by IRC per RECIST v1.1 in the efficacy evaluable analysis set (n=62). Confirmed ORR and DCR were 38.7% (95% CI, 26.6-51.9) and 90.3% (95% CI, 80.1-96.4), respectively (Table). Median PFS was 9.6 months (95% CI, 6.8-NE), and 12-month PFS rate was 42.0% (95% CI, 25.7-57.4). Any grade of TRAEs occurred in 61 (95.3%) pts; 18 (28.1%) pts experienced Grade ≥3 TRAEs. Treatment related SAEs were reported in 6 (9.4 %) pts.

Table. Confirmed tumor response per RECIST v1.1 (efficacy evaluable analysis set*, n=62)
IRC Investigator review
Objective Response Rate, % (95% CI) 38.7 (26.6, 51.9) 41.9 (29.5, 55.2)
Best Overall Response, n (%)
Complete Response 0 (0.0) 1 (1.6)
Partial Response 24 (38.7) 25 (40.3)
Stable Disease 32 (51.6) 27 (43.5)
Progressive Disease 5 (8.1) 8 (12.9)
Not Assessable 1 (1.6) 1 (1.6)
Disease Control Rate, % (95% CI) 90.3 (80.1, 96.4) 85.5 (74.2, 93.1)
*Include pts with measurable disease at baseline per RECIST v1.1 who had ≥1 dose of TIS or LEN, and had ≥1 post-baseline tumor assessment (included 1 patient who died with confirmed clinical disease progression before the first radiological assessment).

Conclusions

TIS plus LEN showed promising antitumor activity with acceptable safety profile as first-line treatment for uHCC.

Clinical trial identification

NCT04401800

Collapse
Poster Display (ID 17) Poster Display

166P - Combination of 5-fluorouracil (FU), interferon (IFN)-alpha2, and nivolumab in unresectable fibrolamellar liver cancer (ID 531)

Presentation Number
166P
Lecture Time
12:30 - 12:30
Speakers
  • S. Lee (Houston, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Lee (Houston, United States of America)
  • F. Duan (Houston, United States of America)
  • S. Jindal (Houston, United States of America)
  • S. Basu (Houston, United States of America)
  • S. Yadav (Houston, United States of America)
  • J. Allison (Houston, United States of America)
  • P. Sharma (Houston, United States of America)
  • Y. Mohamed (Houston, United States of America)
  • A. Kaseb (Houston, United States of America)

Abstract

Background

Fibrolamellar liver cancer (FLC) is a rare malignancy involving multidisciplinary approaches including surgery, liver-directed, and systemic treatment. Combination therapy with 5-fluorouracil (FU) and interferon (IFN)-a2 has long been used for FLC. We hypothesized that addition of anti-PD1 to 5-FU+IFN-a2 may further improve clinical outcome and lead to a favorable modulation of tumor microenvironment.

Methods

We treated 3 patients with unresectable FLC with 5-FU+IFN-a2+nivolumab (N): 5-FU 200 mg/m2 7-day-on/7-day-off, IFN-a2 (4 million U/m2 3 times/week with 5-FU) for 8 weeks, followed by on-treatment biopsy and addition of N 480 mg every 4 weeks to 5-FU+IFN-a2. Tumor tissues were collected under an IRB-approved protocol.

Results

Two patients (26, 29 years) initially had unresectable FLC with a 6.1 cm tumor in seg IV liver invading the common hepatic duct, and a 19.5 cm tumor, peritoneal nodules, and abdominal lymph nodes. After 9 and 5 months of therapy, unresectable tumors were converted to resectable ones: the former had L hepatectomy and bile duct resection; the latter, L hepatectomy, resection of the omentum, spleen, L diaphragm, and lymph node dissection. Both patients completed adjuvant therapy with the same regimen, having no recurrence. The third patient (20 years) had multifocal liver tumors with peritoneal carcinomatosis and lymph nodes. He received treatment for 5 months and achieved stable disease, followed by hepatic tumor bleeding leading to treatment discontinuation. All 3 patients had an increase of CD8+T cells in on-treatment (5-FU+IFN-a2, week 8), compared to pre-treatment biopsies. The available surgical tumor tissues (s/p 5-FU+IFN-a2+N) were also evaluated in patients who underwent surgery with conversation of unresectable to resectable disease, showing a further increase of CD8+T cells.

Conclusions

This clinical outcome of converting an unresectable disease to a resectable one with tumors significantly decreasing in size suggests synergy of 5-FU+IFN-a2 combined with anti-PD1. The robust increased infiltration of CD8+T cells on 5-FU+IFN-a2+N may explain this clinical efficacy. Detailed immune analyses with gene expression profiling will be presented at ESMO Immuno-Oncology Meeting.

Collapse
Poster Display (ID 17) Poster Display

167P - TisCRT-LAPC: A phase Ⅱ clinical trial of Tislelizumab plus chemotherapy(nab-paclitaxel and gemcitabine, AG)followed by consolidative radiotherapy in locally advanced pancreatic cancer (LAPC) (ID 429)

Presentation Number
167P
Lecture Time
12:30 - 12:30
Speakers
  • X. Guo (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • X. Guo (Shanghai, China)
  • Y. Zhou (Shanghai, Changning, China)
  • Y. Ji (Shanghai, China)
  • W. Lou (Shanghai, Xuhui, China)
  • L. Wu (Shanghai, China)

Abstract

Background

Previously, we have demonstrated that consolidative chemoradiotherapy following induced chemotherapy is an optimal regimen for LAPC. This study aimed to evaluate the clinical benefit of tislelizumab (anti-PD-1) plus AG chemotherapy sequenced by consolidative radiotherapy for therapy-naïve LAPC.

Methods

Patients were given tislelizumab 200mg Q3W, nab-paclitaxel 125 mg/m2 and gemcitabine 1,000 mg/m2 on days 1 and 8 every three weeks for 4-6 cycles. Those who did not progress continued to sequence radiotherapy and tislelizumab 200 mg Q3W. Then, maintenance chemotherapy with anti-PD-1 was ongoing until progression. The primary endpoint was 12-month progression-free survival rate, and secondary endpoints included objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and safety. The Data cutoff date for this interim analysis was August 30, 2022.

Results

From April 2021 to August 2022, 15 patients were enrolled. Twelve patients completed at least one efficacy assessment. 4 of 12 patients had an optimal partial response (PR), 7 patients had stable disease (SD), and 1 had progressive disease (PD). ORR was 33.3%, and DCR was 91.7%, respectively. Most patients (10/12) showed serum CA19-9 decline with different degrees, but only PR patients (3 cases) fell within normal ranges and declines exceeding 93.7%.The median follow-up time was 283 days, and the median PFS was 266 days (95%CI,176-not reached). The median OS was not reached. 6-month and 12-month PFS rates were 70.7% and 44.2%, separately. Notably, the12-month OS rate was 100%. The most common treatment-emergent adverse events (TEAEs) were myelosuppression, rash, and fatigue. Grade 3/4 TEAEs were neutropenia in 4 patients, anemia in 1 patient, and rash in 1 patient. Immune-related AEs included 1 Grade-1 myocarditis and 1 Grade-1 pneumonia, and no deaths occurred.

Conclusions

Our data suggest that tislelizumab combined with AG chemotherapy sequenced by radiotherapy for LAPC provided encouraging efficacy with reasonable tolerability. Significant CA19-9 decline is predictive of therapeutic response.

Clinical trial identification

ChiCTR2000040872

Collapse
Poster Display (ID 17) Poster Display

168P - Liver metastases (mets) and treatment effect of cemiplimab-based therapy: an analysis from three Phase 3 trials (EMPOWER-Lung 1, EMPOWER-Lung 3 Part 2, and EMPOWER-Cervical 1) (ID 575)

Presentation Number
168P
Lecture Time
12:30 - 12:30
Speakers
  • A. Baramidze (Tbilisi, Georgia)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Baramidze (Tbilisi, Georgia)
  • C. Gessner (Leipzig, Germany)
  • M. Gogishvili (Tbilisi, Georgia)
  • A. Sezer (Adana, Turkey)
  • T. Makharadze (Tbilisi, Georgia)
  • S. Kilickap (Ankara, Turkey)
  • M. Gumus (Istanbul, Turkey)
  • K. Tewari (Orange, United States of America)
  • B. Monk (Edgewater, United States of America)
  • A. De Melo (Rio de Janeiro, Brazil)
  • A. Oaknin (Barcelona, Spain)
  • S. Li (Tarrytown, United States of America)
  • B. Gao (Tarrytown, United States of America)
  • M. Mathias (Sleepy Hollow, United States of America)
  • G. Gullo (Tarrytown, United States of America)
  • M. Salvati (New York, United States of America)
  • F. Seebach (Tarrytown, United States of America)
  • I. Lowy (Tarrytown, United States of America)
  • M. Fury (New York, NY, United States of America)
  • P. Rietschel (Tarrytown, United States of America)

Abstract

Background

Emerging data suggest that liver mets are immune privileged and can induce systemic loss of tumour specific CD8+ T cells, leading to reduced anti-tumour immunity. Liver mets were shown to negatively predict the success of immunotherapy (as opposed to chemotherapy [chemo] or targeted therapy) in several cancer types. However, limited prospective data exist on the effect of immunotherapy in patients (pts) with liver mets.

Methods

This post-hoc analysis included pts with baseline liver mets from three Phase 3 trials of cemiplimab (cemi; anti-PD-1) in non-small cell lung cancer (NSCLC) and cervical cancer: EMPOWER-Lung 1, EMPOWER-Lung 3 Part 2, and EMPOWER-Cervical 1. Overall survival (OS), progression free survival (PFS), and objective response rate (ORR) were analysed.

Results

Patients with baseline liver metastases represented 17% of pts in EMPOWER-Lung 1, 15% in EMPOWER-Lung 3 Part 2, and 24% in EMPOWER-Cervical 1. As expected, OS, PFS, and ORR were generally poorer in pts with liver mets regardless of treatment arms. Nonetheless, in EMPOWER-Lung 1 (1L treatment for advanced NSCLC with PD-L1 ≥50%), among pts with liver mets, cemiplimab monotherapy demonstrated notably longer OS (not reached vs 7.4 months; hazard ratio [HR] 0.38) and PFS (6.2 vs 4.2 months; HR 0.51), as well as higher ORR (29% vs 15%) vs chemo (Table 1). Similar improvement was observed for EMPOWER-Lung 3 Part 2 (1L cemiplimab+chemo vs placebo+chemo for advanced NSCLC with all PD-L1 levels). In EMPOWER-Cervical 1 (2L treatment of recurrent cervical cancer with all PD-L1 levels), cemiplimab showed comparable OS, PFS, and ORR vs chemo in the liver mets subgroup.

Table 1. OS, PFS, and ORR in patients with baseline liver mets

EMPOWER-Lung1
n=563

EMPOWER-Lung3 Part 2
n=466

EMPOWER-Cervical1
n=608

Liver mets n=95


Cemi n=48
vs
chemo n=47

Liver mets n=70


Cemi+chemo n=47
vs
Pbo+chemo n=23

Liver mets n=143


Cemi n=78
vs
chemo n=65

OS median, months

NR vs 7.4

14.4 vs 8.9

5.2 vs 6.5

OS HR, (95% CI)

0.38
(0.19–0.75)

0.61
(0.31–1.20)

0.92
(0.62–1.36)

PFS median, months

6.2 vs 4.2

5.4 vs 4.2

1.4 vs 1.9

PFS HR, (95% CI)

0.51
(0.30–0.88)

0.64
(0.34–1.17)

0.89
(0.62–1.28)

ORR, %

29 vs 15

34 vs 13

9 vs 3

PD-L1≥50% population

CI, confidence interval; Pbo, placebo.

Conclusions

Despite a lower efficacy of Cemiplimab in patients with liver mets from primary lung/cervix cancer than those without, the relative benefit over prior standard-of-care regimens was preserved, and for some patients, even enhanced.

Clinical trial identification

NCT03088540 (EMPOWER-Lung 1), NCT03409614 (EMPOWER-Lung 3), NCT03257267 (EMPOWER-Cervical 1)

Editorial acknowledgement

Medical writing support was provided by Qing Zhou, PhD, ELS from Regeneron Pharmaceuticals, Inc. with editing supporting from Prime, Knutsford, UK, funded by Regeneron Pharmaceuticals, Inc.

Collapse
Poster Display (ID 17) Poster Display

169P - Immunomodulatory effects of RBS2418, an oral ENPP1 inhibitor in combination with pembrolizumab in checkpoint-refractory metastatic adrenal cancer (ID 350)

Presentation Number
169P
Lecture Time
12:30 - 12:30
Speakers
  • J. Glenn (Sunnyvale, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Glenn (Sunnyvale, United States of America)
  • I. Csiki (Glendora, United States of America)
  • J. Schanzer (Sunnyvale, United States of America)
  • B. Tuan (San Francisco, United States of America)
  • N. Huang (Sunnyvale, United States of America)
  • A. Dong (Sunnyvale, United States of America)
  • E. John (Sunnyvale, United States of America)
  • L. O'Toole (Sunnyvale, United States of America)
  • J. Seppa (Sunnyvale, United States of America)
  • R. Hawley (Sunnyvale, United States of America)
  • C. Exon (Sunnyvale, United States of America)
  • K. Klumpp (Sunnyvale, United States of America)

Abstract

Background

ENPP1 is a type II transmembrane protein with nucleotide pyrophosphatase, and phosphodiesterase enzymatic activities and its expression is associated with poor prognosis in cancer. ENPP1 inhibition protects cGAMP and ATP from hydrolysis and reduces adenosine levels in the TME, activates APCs and increases T-cell infiltration promoting anticancer immunity. RBS2418 is a potent oral ENPP1 inhibitor. Here, we report pharmacokinetic (PK) and immunomodulatory properties of RBS2418 in combination with pembrolizumab (pembro) in a checkpoint-refractory high grade adrenal cancer with an immune-desert tumor phenotype at baseline.

Methods

Pembro was given at 200mg IV every 3 weeks with escalating doses of RBS2418 weekly, at 100 mg p.o. BID, followed by 200 and 400 mg p.o BID. Blood samples were collected at each dose level to determine plasma RBS2418 concentration and serum ENPP1 inhibition. Peripheral blood immune cell subpopulations were analyzed by flow cytometry and TCR/RNA sequencing.

Results

All dose levels of RBS2418 with pembro were safe, well tolerated with no DLTs. Plasma concentrations (Ctrough) of RBS2418 corresponded to > 15-fold and > 30-fold above the 90% inhibition level of ENPP1 (EC90) for the 100 and 200 mg dose, respectively. cGAMP was fully stable in serum. Peripheral blood analyses showed a 2.1-fold increase from baseline in conventional dendritic cells (cDCs) and 2.5-fold expansion of proliferating CD4/Ki67 T cells. Furthermore, CD8/Ki67 proliferating T cells increased 19.5-fold from baseline. TCR/RNA sequencing showed a > 50-fold expansion in TCR CD3β clonotypes, an increase in hyperexpanded TCR clonotypes and upregulation of granzyme B, perforin and granulysin gene expression.

Conclusions

RBS2418 with pembro was safe, well tolerated with no DLTs. RBS2418 PK data showed excellent oral bioavailability with plasma levels leading to complete ENPP1 inhibition. RBS2418 induced increases in peripheral cDCs, proliferation of CD4 and CD8 T cells and expansion of TCR clonotypes as well as upregulation of T cell cytotoxic granule protein gene expression supporting clinical development of this novel first-in-class immunotherapy agent in an ongoing clinical trial (NCT05270213).

Collapse
Poster Display (ID 17) Poster Display

170P - EO2401 microbiome derived therapeutic vaccine + nivolumab +/- bevacizumab, in neoadjuvant, adjuvant and non-surgery linked treatment of recurrent glioblastoma: phase 1-2 EOGBM1-18/ROSALIE study (ID 440)

Presentation Number
170P
Lecture Time
12:30 - 12:30
Speakers
  • W. Wick (Heidelberg, Germany)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • W. Wick (Heidelberg, Germany)
  • A. IdBaih (Paris, France)
  • M. Vieito Villar (Barcelona, La Coruña, Spain)
  • G. Tabatabai (Tübingen, Germany)
  • A. Stradella (L'Hospitalet de Llobregat, Spain)
  • F. Ghiringhelli (Dijon, France)
  • M. Burger (Frankfurt am Main, Germany)
  • I. Mildenberger (Mannheim, Germany)
  • U. Herrlinger (Bonn, Germany)
  • M. Touat (Villejuif, Cedex, France)
  • P. Wen (Boston, United States of America)
  • A. Wick (Heidelberg, Germany)
  • C. Gouttefangeas (Tuebingen, Germany)
  • A. Maia (Porto Salvo, -- select, Portugal)
  • C. Bonny (Paris, France)
  • J. Paillarse (Paris, France)
  • J. Fagerberg (Prague, Czech Republic)
  • D. Reardon (Boston, MA, United States of America)

Abstract

Background

EO2401 was designed to activate memory T cells cross-reacting with tumor associated antigens and includes synthetically produced HLA-A2 peptides with molecular mimicry to antigens (IL13Rα2, BIRC5 and FOXM1) upregulated in glioblastoma, and the CD4 helper peptide UCP2.

Methods

Patients with glioblastoma at first progression after radiotherapy/temozolomide received EO2401 (300 µg/peptide, q2w x4 then q4w) with nivolumab (3 mg/kg q2w; EN), or EN with bevacizumab (10 mg/kg q2w; ENB). In study part 2, low-dose bevacizumab (5 mg/kg q2w) could be used as symptom directed time-limited treatment of edema. In study part 3, 6 evaluable patients are to be treated with neoadjuvant EN, and 15 further patients with ENB (enrollment ongoing).

Results

Part 1 included 40 patients (EN = 29, ENB = 11). Part 2 enrolled 36 patients treated with EN.

EN and ENB (n=76) were well tolerated with EO2401 associated tox limited to local administration site reactions (45% of patients; events 70% Grade 1, 26% Grade 2, and 4% Grade 3). The frequency and severity of nivolumab-/bevacizumab-tox was consistent with historical single-agent data.

Immune monitoring demonstrated T cell responses against the 3 microbiome-derived peptides for 97% of patients investigated ex vivo or after in vitro stimulation (IVS). Cross-reactivity against the pool of 3 human target peptides was demonstrated in 96% of patients by IFN-γ ELISpot. The strength of the immune response correlated with clinical outcome (progression-free survival [PFS] and objective responses [OR]).

For part 1, median PFS, and median survival for EN (n=29, median follow-up 20.1 months) were 1.8 and 10.6 months. Patients on ENB (n=11, median follow-up 13.1 months) had median PFS of 5.5 months and 55% of patients were alive beyond 12 months. OR rate / Disease Control Rate (DCR) (OR rate + stable disease) for EN and ENB were 14%/34% and 55%/82%.

Conclusions

EO2401 was well tolerated and generated rapid and durable immune responses correlating with clinical outcome. Addition of standard bevacizumab to EN improved efficacy. Updated data from all parts of the trial will be presented. NCT04116658.

Clinical trial identification

NCT04116658

Collapse
Poster Display (ID 17) Poster Display

171P - Pharmacodynamic (PD) biomarker analysis from CheckMate (CM) 8KX: a multitumor study of a subcutaneous (SC) formulation of nivolumab (NIVO) monotherapy (ID 441)

Presentation Number
171P
Lecture Time
12:30 - 12:30
Speakers
  • C. Jackson (Dunedin, New Zealand)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Jackson (Dunedin, New Zealand)
  • D. Perumal (Princeton, United States of America)
  • I. Lugowska (Warsaw, Poland)
  • A. O’donnell (Wellington, New Zealand)
  • R. North (Tauranga, New Zealand)
  • P. Calvo Ferrandiz (Madrid, Spain)
  • L. Latten-Jansen (Maastricht, Netherlands)
  • C. Sánchez (Santiago, Chile)
  • L. Medina Rodríguez (Malaga, Spain)
  • A. Santoro (Rozzano, (MI), Italy)
  • L. Li (Summit, United States of America)
  • K. Sidik (Summit, United States of America)
  • T. Tang (Plainsboro, New Jersey, United States of America)
  • J. Deutsch (Baltimore, United States of America)
  • J. Taube (Baltimore, United States of America)
  • C. Horak (Cambridge, United States of America)
  • S. Ravimohan (Cambridge, United States of America)
  • S. Lonardi (Padova, Italy)

Abstract

Background

CM 8KX is a phase 1/2 study of an SC NIVO formulation with a permeation enhancer, recombinant human hyaluronidase PH20 enzyme (rHuPH20), developed to allow rapid delivery and reduced treatment burden compared with intravenous (IV) NIVO administration. We report PD biomarker changes in the tumor microenvironment and peripheral blood from CM 8KX and key PD biomarker comparisons between SC and IV NIVO.

Methods

Patients were checkpoint inhibitor–naive, ≥ 18 years of age, ECOG performance status 0–1, with metastatic/unresectable solid tumors and measurable disease. Patients received SC NIVO 720 mg ± rHuPH20, SC NIVO 960 mg ± rHuPH20, or SC NIVO 1200 mg + rHuPH20. Paired baseline and on-treatment tumor biopsies were assessed for CD8+ tumor-infiltrating lymphocytes (TILs) and programmed death ligand 1 expression on tumor cells (TC PD-L1) by immunohistochemistry. Soluble PD-1 (sPD-1), interferon gamma (IFNγ), CXCL9, and CXCL10 were assessed in serum from cycle (C) 1 day (D) 1 to C2D1. sPD-1 changes were compared with unpublished data from IV NIVO 3 mg/kg Q2W administration in CM 038; other biomarkers were compared with published data.

Results

As previously reported, SC NIVO ± rHuPH20 resulted in numerical mean increases from baseline to C1D15 in CD8+ TILs (5.0% [95% CI, −17.5 to 27.5]) and TC PD-L1 (2.8% [95% CI, −12.8 to 18.4]), which are surrogates for tumoral IFNγ activity. These observations were consistent with those published for IV NIVO in several indications. sPD-1 increased significantly following SC NIVO across dose cohorts, similar to increases observed in CM 038. Early increases in IFNγ and CXCL9 were observed following SC NIVO ± rHuPH20, with significant cycle-day effects but no apparent dose dependence. Numerical increases in CXCL10 were also observed. These increases are consistent with published IV NIVO data from CM 038.

Conclusions

Increases in tumor biomarkers and changes in key peripheral PD markers demonstrate immune activation post-SC NIVO. Despite the limitations of cross-trial comparisons, PD effects are consistent with historical IV NIVO data from CM 038. The results support further evaluation of SC NIVO + rHuPH20 in phase 3 studies.

Clinical trial identification

Clinicaltrials.gov NCT03656718 and NCT01621490.

Editorial acknowledgement

Editorial support was provided by Sandra Page, PhD, of Spark Medica Inc.

Collapse
Poster Display (ID 17) Poster Display

172P - Phase 1 Study of JTX-8064, a LILRB2 (ILT4) inhibitor, as monotherapy and combination with pimivalimab (pimi), a PD-1 inhibitor (PD-1i), in patients (pts) with advanced solid tumors (ID 476)

Presentation Number
172P
Lecture Time
12:30 - 12:30
Speakers
  • K. Papadopoulos (San Antonio, TX, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • T. Li (Sacramento, United States of America)
  • N. Lakhani (Grand Rapids, United States of America)
  • J. Powderly (Huntersville, United States of America)
  • T. George (Gainesville, United States of America)
  • D. Teoh (Minneapolis, United States of America)
  • D. Kilari (Milwaukee, United States of America)
  • G. Giaccone (Thousand Oaks, MD, United States of America)
  • R. Sanborn (Portland, United States of America)
  • S. Ghamande (Augusta, United States of America)
  • P. Lorusso (New Haven, MI, United States of America)
  • G. Gibney (Washington DC, United States of America)
  • V. Ma (Madison, United States of America)
  • K. Yalamanchili (Lubbock, United States of America)
  • J. Brown (Cleveland, United States of America)
  • N. Mota (Cambridge, United States of America)
  • C. Tasillo Kadra (Cambridge, United States of America)
  • B. Umiker (Cambridge, United States of America)
  • X. Xiao (Cambridge, United States of America)
  • E. Trehu (Cambridge, United States of America)

Abstract

Background

JTX-8064, a highly selective and potent IgG4 inhibitor of myeloid-specific Leukocyte Immunoglobulin Like Receptor B2 (LILRB2), leads to macrophage reprogramming from an immunosuppressive to an immune activated state, resulting in T cell activation. INNATE (NCT04669899) is a phase (P) 1/2 dose escalation/expansion study of two investigational agents, JTX-8064 monotherapy (mono) and combination (combo) with pimi. P1 data defining Recommended P2 Dose (RP2D) are presented.

Methods

Pts with advanced solid tumors who progressed after all available therapy were treated at 7 dose levels of JTX-8064 mono IV q3w (50, 150, 300, 450, 600, 900, 1200 mg) and 2 dose levels of JTX-8064 (700, 1200 mg) + pimi 500 mg IV q3w using Bayesian design. 1o objectives: safety, tolerability, RP2D. 2 o objectives: receptor occupancy (RO), PK, immunogenicity. Objective response rate (RECIST 1.1) was exploratory.

Results

31 pts were treated in dose escalation, 22 JTX-8064 mono, 9 JTX-8064 + pimi. No dose limiting toxicities; maximum tolerated dose not reached.

Safety: Mono: Median age 67. Eleven pts (50%) had treatment-related adverse events (TRAE). Most common were fatigue (n= 5), upper abdominal pain, arthralgia, flushing, nausea, and pyrexia (n= 2 each). The only Grade 3 (G3) TRAE and only serious related AE (SRAE) was tumor flare at 1200 mg. Combo: Median age 63. Six pts (66.7%) had TRAE, most common fatigue (n=4) and pyrexia (n=2). No ≥ G3 TRAEs, no SRAEs.

PK was linear. Full RO thru 21 days was achieved at ≥ 300 mg. RP2D of 700 mg q3w was selected for JTX-8064 +/- pimi to optimize RO in tumor, with Cmin at steady state 63.4 (26.0-139.7) ug/mL. Treatment induced antibodies to JTX-8064 occurred in 1 mono and no combo pts.

P1 Efficacy: Mono: 0 PR, 7 SD with 2 durable SD (appendiceal cancer 8.3, ovarian cancer 12.2 mo). Combo: 1 PR (6.2 mo) at 700 mg in PD-1i resistant cholangiocarcinoma with resolution of both bone pain and cachexia, 3 SD with 1 durable SD (post-PD-1i NSCLC 6 mo).

Conclusions

JTX-8064 was well-tolerated as mono and with pimi, with 700 mg q3w selected as the preliminary RP2D. Enrollment is ongoing in P2, including cohorts in renal and ovarian cancer that have met Simon’s 2-stage response criteria to expand.

Clinical trial identification

NCT04669899

Collapse
Poster Display (ID 17) Poster Display

173P - First-in-human study to evaluate the safety and clinical activity of FS222, a tetravalent bispecific antibody targeting PD-L1 and CD137, in patients with advanced solid tumors (ID 470)

Presentation Number
173P
Lecture Time
12:30 - 12:30
Speakers
  • G. De Velasco Oria (Madrid, MA, Spain)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • G. De Velasco Oria (Madrid, MA, Spain)
  • E. Garralda (Barcelona, Spain)
  • V. Moreno Garcia (Madrid, Spain)
  • I. Melero (Pamplona, Spain)
  • I. Victoria Ruiz (Barcelona, Spain)
  • A. Indacochea (Barcelona, Barcelona, Spain)
  • A. Cervantes (Valencia, Valencia, Spain)
  • A. Oberoi (Barcelona, Spain)
  • D. Jones (Cambridge, United Kingdom)
  • M. Lakins (Cambridge, United Kingdom)
  • L. Kayitalire (Cambridge, United States of America)

Abstract

Background

FS222 is a novel tetravalent bispecific antibody targeting PD-L1 and CD137 that aims to overcome the limitations of current PD-(L)1 and CD137 (4-1BB) monotherapies via a unique mechanism of action that elicits PD-L1-dependent CD137 activation. This has the potential to deliver strong antitumor clinical activity, even in PD-L1 low conditions as seen in preclinical models, with good tolerability.

Methods

FS222 is being evaluated in an open-label, multi-center study in patients with advanced treatment-refractory solid tumors. Part A consists of single patient cohorts in an accelerated dose titration followed by a 3+3 design and pharmacokinetic/pharmacodynamic (PK/PD) expansion cohorts. Patients received FS222 intravenously every 4 weeks until disease progression or unacceptable toxicity. Adverse events and dose-limiting toxicities (DLTs) were primary endpoints. Efficacy (RECIST 1.1), PK, immunogenicity and PD biomarkers were secondary endpoints. Efficacy (iRECIST) and further PD biomarkers were assessed as exploratory endpoints.

Results

As of 20th July, 2022, 33 patients (median age: 58 years) were treated with FS222 at doses of 300 μg (n=1), 1 mg (n=1), 3 mg (n=1), 10 mg (n=1), 30 mg (n=5), 0.75 mg/kg (n=15) and 1 mg/kg (n=9). 24 patients (73%) were checkpoint inhibitor-naïve. Median time on study was 58 days (range 19-359) with 11 ongoing. One patient experienced a DLT of grade-3 febrile neutropenia and one patient experienced grade-3 transaminase elevation without total bilirubin increase, both at 1 mg/kg. Maximum tolerated dose was not reached and dose escalation is ongoing. Pharmacological activity was demonstrated by increased peripheral soluble target receptors and proliferating CD4+ and CD8+ T cells. At 8 weeks, one patient experienced a complete response (CR), 6 had disease stabilization, 14 had progressive disease (RECIST 1.1), 3 discontinued before week 8, and 9 were awaiting their week 8 evaluation. The CR occurred in a non-squamous NSCLC patient who was naïve to PD-(L)1 therapy, dosed at 1 mg/kg. The CR remained persistent 10 months later.

Conclusions

Thus far, FS222 has demonstrated manageable tolerability and early signs of antitumor activity.

Clinical trial identification

NCT04740424

Collapse
Poster Display (ID 17) Poster Display

174P - First-in-human Study of NGM707, An ILT2/ILT4 Dual Antagonist Antibody in Advanced or Metastatic Solid Tumors: Preliminary Monotherapy Dose Escalation Data (ID 587)

Presentation Number
174P
Lecture Time
12:30 - 12:30
Speakers
  • A. Naing (Houston, TX, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Naing (Houston, TX, United States of America)
  • J. Wang (Lake Mary, United States of America)
  • M. Sharma (Grand Rapids, United States of America)
  • D. Sommerhalder (San Antonio, United States of America)
  • L. Gandhi (New York, United States of America)
  • D. Oh (Seoul, Korea, Republic of)
  • Y. Jiang (Baltimore, United States of America)
  • J. Michalski (Omaha, United States of America)
  • J. Lee (Seoul, Korea, Republic of)
  • K. Zhou (South San Francisco, United States of America)
  • N. Taylor (Davis, United States of America)
  • L. Yan (South San Francisco, United States of America)
  • J. Roda (South San Francisco, United States of America)
  • L. Blum (South San Francisco, United States of America)
  • L. Ling (South San Francisco, United States of America)
  • I. Mikaelian (South San Francisco, United States of America)
  • A. Depaoli (South San Francisco, United States of America)
  • V. Hanes (South San Francisco, United States of America)
  • D. Kaplan (South San Francisco, United States of America)
  • H. Lieu (South San Francisco, United States of America)

Abstract

Background

NGM707 is a humanized monoclonal antibody that binds the immune inhibitory receptors ILT2 and ILT4 and blocks interactions with their HLA ligands.

Methods

NGM707-IO-101 is a Phase 1/2, dose escalation/expansion study evaluating NGM707 as a monotherapy and in combination with pembrolizumab. The monotherapy dose escalation enrolled eligible pts with advanced or metastatic solid tumors into escalating dose cohorts of NGM707 (6-1800 mg, Q3W iv). Primary objectives are to assess safety and tolerability of NGM707 and to identify phase 2 doses. Secondary/exploratory objectives include assessment of PK/biomarker correlation, immunogenicity and preliminary antitumor activity per RECIST v1.1.

Results

As of 21 Sep 2022, 33 pts have been enrolled in the monotherapy dose escalation at dose levels up to 1800 mg. Pts had received a median of 4 prior therapies (range 1-11) and all had metastatic disease. Treatment (Tx)-related adverse events (any grade/grade ≥3) occurred in 44%/9% of pts. One dose-limiting toxicity of pneumonitis (G5) in a pt with pulmonary metastasis was observed at 600 mg. A maximum tolerated dose was not reached and the maximum administered dose was 1800 mg. Linear PK was observed at doses ≥200 mg and analysis of peripheral immune cells demonstrated dose-dependent receptor occupancy (RO), with doses ≥200 mg maintaining ILT2 and ILT4 RO for the entire dosing interval. Preliminary evidence of myeloid reprogramming was observed in tumor biopsies, with reduction of the M2 macrophage marker CD163 observed post-treatment. Of 20 response-evaluable pts, best overall responses to date are stable disease in 6 pts and non-complete response/non-progressive disease in 1 pt. Five pts had reduced target lesion size with a maximum decrease in 1 pt of 28%. Seven pts remain on study with a maximum ongoing tx duration of 5 mo.

Conclusions

NGM707 monotherapy appears to be well tolerated at all dose levels. In this advanced, metastatic solid tumor cohort, early signals of efficacy were observed. Preliminary evidence of myeloid reprogramming was seen in tumor biopsies. Monotherapy tx is ongoing and enrollment of pembrolizumab combination cohorts has been initiated.

Clinical trial identification

NCT04913337

Collapse
Poster Display (ID 17) Poster Display

175P - Safety and Preliminary Efficacy of GEN1042 (DuoBody®-CD40x4-1BB) Combination Therapy in Patients With Advanced Solid Tumors (ID 692)

Presentation Number
175P
Lecture Time
12:30 - 12:30
Speakers
  • I. Melero (Pamplona, Spain)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • I. Melero (Pamplona, Spain)
  • E. Grande Pulido (Madrid, Spain)
  • M. De Miguel Luken (Madrid, Spain)
  • M. Johnson (Nashville, TN, United States of America)
  • J. Bauman (Philadelphia, United States of America)
  • V. Moreno Garcia (Madrid, Spain)
  • A. Walter (Mainz, Germany)
  • H. Adams III (Princeton, United States of America)
  • Ö. Türeci (Mainz, Germany)
  • G. Russo (Plainsboro, United States of America)
  • U. Sahin (Mainz, Germany)
  • J. Steinberg (Princeton, United States of America)
  • T. Ahmadi (Princeton, United States of America)
  • E. Felip (Barcelona, Spain)

Abstract

Background

GEN1042 (DuoBody-CD40x4-1BB) is an investigational, novel, agonistic bispecific antibody that combines targeting and conditional activation of CD40 and 4-1BB on immune cells. GEN1042 demonstrated biologic and early clinical activity with a manageable safety profile in patients (pts) with advanced solid tumors in the dose escalation part of the ongoing phase 1/2 trial (NCT04083599). Previously presented preclinical data suggest that the immune response generated by GEN1042 treatment (Tx) can be amplified by co-administration of a PD-1 inhibitor. Combination with chemotherapy (CTx) is hypothesized to further enhance antitumor responses by increasing antigen release and PD-L1 expression. We present data from the safety run-in (SRI) and expansion cohorts investigating GEN1042 + pembrolizumab (PEM) ± standard of care (SoC) CTx.

Methods

Tx-naive pts with advanced, metastatic/unresectable/recurrent non-CNS tumors received GEN1042 + PEM ± CTx (nab-paclitaxel + gemcitabine or cis/carboplatin + 5-FU) until disease progression or unacceptable toxicity. Primary endpoint was dose-limiting toxicity (DLT) for SRI and objective response rate (ORR) for the expansion cohorts. Secondary endpoints included AEs, PK, antitumor activity (RECIST v1.1), and pharmacodynamic biomarkers.

Results

As of July 27, 2022, 20 pts (median age, 70.5 y; 12 pts with NSCLC, 3 pts with melanoma, 5 pts with HNSCC) had received GEN1042 + PEM; 17 pts (median age, 64.0 y; 5 pts with HNSCC, 12 pts with PDAC) had received GEN1042 + PEM + CTx. Tx-related AEs in ≥15% of pts (all grades; grade ≥3) were pruritus (20%; 0%), rash (15%; 5%), and pyrexia (15%; 0%) in the GEN1042 + PEM cohorts, and transaminase elevation (23.5%; 5.9%), rash (17.6%; 5.9%), diarrhea (17.6%; 0%), fatigue (23.5%; 0%), and nausea (17.6%; 0%) in the GEN1042 + PEM + CTx cohorts. No DLTs were observed in either group, and irAEs were as expected and manageable. Encouraging preliminary activity was observed in all patients with HNSCC treated with GEN1042 + PEM + CTx; updated results will be presented.

Conclusions

GEN1042 + PEM ± SoC CTx was overall well tolerated and showed initial antitumor activity in pts with advanced/metastatic disease.

Clinical trial identification

NCT04083599

Study Start Date: September 17, 2019

Editorial acknowledgement

Medical writing and editorial assistance were provided by Peloton Advantage, LLC, an OPEN Health company, Parsippany, NJ, USA, and funded by Genmab.

Collapse
Poster Display (ID 17) Poster Display

176P - Phase 1 Study of INCB106385 Alone or in Combination with Immunotherapy in Advanced Solid Tumors (ID 436)

Presentation Number
176P
Lecture Time
12:30 - 12:30
Speakers
  • A. Italiano (Bordeaux, CEDEX, France)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Italiano (Bordeaux, CEDEX, France)
  • O. Saavedra Santa Gadea (Barcelona, Spain)
  • K. Papadopoulos (San Antonio, TX, United States of America)
  • A. Naing (Houston, TX, United States of America)
  • C. Gomez-Roca (Toulouse, Haute-Garonne, France)
  • M. Mita (Los Angeles, CA, United States of America)
  • M. Stein (New York, United States of America)
  • I. Rybicka (Epalinges, Switzerland)
  • M. Van der Velden (Geneva, Switzerland)
  • P. Zhang (Wilmington, United States of America)
  • M. Smith (Wilmington, United States of America)
  • O. Ivanova (Morges, Switzerland)
  • Y. Loriot (Villejuif, Cedex, France)

Abstract

Background

A2A/A2B receptors drive adenosine-mediated immunosuppression in the tumor microenvironment; blockade has shown clinical activity in advanced solid tumors. This ongoing, 2-part (dose-escalation/expansion) phase 1 study is investigating the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of INCB106385, an oral, potent, selective dual A2A/A2B receptor antagonist, alone or combined with an anti-PD-1 antibody retifanlimab for advanced solid tumors (NCT04580485). Part 1 (dose-escalation) monotherapy results are reported herein.

Methods

Eligible adults had confirmed advanced solid tumors by histology/cytology, disease progression after available therapy (including PD-[L]1 therapy, if applicable), measurable disease (RECIST v1.1), and ECOG PS 0-1. INCB106385 dosing started at 40 mg QD and was escalated using a BOIN design. PK parameters were calculated from INCB106385 plasma concentrations using non-compartmental, model-independent methods. PD was evaluated by measuring the reversal of adenosine-dependent inhibition of T cell and monocyte inflammatory cytokine production.

Results

As of July 16, 2022, 18 pts (male, n=16; age ≥65 years, n=17) had received INCB106385 monotherapy (40 mg QD, n=4; 40 mg BID, n=4; 80 mg BID, n=6; 160 mg BID, n=4); 16 discontinued therapy (progressive disease, n=14). The median (range) duration of treatment was 9.5 (0.9–40) weeks. Treatment-emergent AEs (TEAEs) occurred in 17 pts, most commonly asthenia, constipation, and diarrhea (each n=4). One pt had a grade ≥3 TEAE (grade 3 neutropenia; 40 mg BID); 2 had serious TEAEs (tumor pain; encephalopathy; both 40 mg BID); none were treatment-related. There were no dose-limiting toxicities or discontinuations due to TEAEs. INCB106385 plasma concentration remained above EC90 (~300 nM) for all BID doses during the inter-dose 0-12 h. In PD analysis, doses ≥80 mg BID approached EC90 at steady state in most pts.

Conclusions

INCB106385 was generally well tolerated in pts with advanced solid tumors, with asthenia and gastrointestinal events being the most common TEAEs. Doses ≥80 mg BID were sufficient for target inhibition in most pts. Further dose escalation is ongoing in parallel with dose expansion.

Clinical trial identification

ClinicalTrials.Gov ID: NCT04580485

Editorial acknowledgement

Medical writing assistance was provided by Simon J. Slater, PhD, CMPP, of Envision Pharma Group (Philadelphia, PA, USA), and funded by Incyte Corporation.

Collapse
Poster Display (ID 17) Poster Display

177P - Targeting of immune regulatory proteins through locoregional delivery of mAbs augments CD8 T cell mediated anti-tumour responses in vivo. (ID 639)

Presentation Number
177P
Lecture Time
12:30 - 12:30
Speakers
  • Q. Wright (St Lucia, Australia)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Q. Wright (St Lucia, Australia)
  • D. Sinha (St Lucia, Australia)
  • J. Gonzalez-Cruz (St Lucia, Australia)
  • I. Frazer (St Lucia, Australia)
  • G. Leggatt (St Lucia, Australia)

Abstract

Background

Immune checkpoint blockade (ICB) has resulted in impressive clinical response rates in the treatment of melanoma and squamous cell carcinoma. Unfortunately, systemic administration of ICB therapy is associated with inconsistent patient responses alongside severe immune-related adverse events (iRAEs), resulting in discontinuation of treatment, especially when anti-PD-1 and anti-CTLA-4 mAb therapies are used in combination. Preclinical studies indicate that locoregional administration of ICB to internal solid tumours enhances immune responses with minimal toxicities, but this approach has not been fully explored in cutaneous tumours.

Methods

A triple combination (TC) of mAbs targeting (4-1bb/PD-1/VISTA) were injected peritumourally (10ug/mAb) or systemically (100ug/mAB) to C57BL/6J mice bearing TC-1 ears tumours, 3x over 9 days and compared to isotype control mAb treated mice. Tumour volumes, resistance to secondary tumour rechallenge, IFN-g ELISPOT and serum ALT assays and flow cytometry analysis of tumour bearing ears of therapy and isotype mAb treated mice was undertaken to characterize the mechanism, efficacy and safety following therapy administration.

Results

TC administered locally to ear tumours, induced potent anti-tumour responses and had superior efficacy to monotherapy. An abscopal effect was observed after one of two tumour bearing ears were administered TC therapy, resulting in 66% of treated mice regressing tumours in both ears. Surviving treated mice were resistant to subcutaneous TC-1 tumour rechallenge, implying the later induction of systemic immune memory. Mechanistically, the central role of CD8 T cells in driving therapy mediated anti-tumour responses was confirmed using systemic CD8 depletion. Increased proportions of Granzyme B+ CD8+ T cells and lower CD4+ Tregs were concurrently observed in TC treated animals compared to isotype control. While i.v. therapy resulted in elevated serum ALT and liver immune cell infiltrates, locally administered TC mAbs were well tolerated.

Conclusions

Our results demonstrate a novel, safe, dose-sparing strategy to administer combinations of checkpoint therapies for immune targeting of advanced, local cutaneous tumours.

Collapse
Poster Display (ID 17) Poster Display

178P - Antibody engineering to evaluate binding, internalisation, and intracellular routing of recombinant immunotherapeutic fusion proteins. (ID 183)

Presentation Number
178P
Lecture Time
12:30 - 12:30
Speakers
  • M. Karaan (Cape Town, South Africa)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Karaan (Cape Town, South Africa)
  • S. Barth (Cape Town, South Africa)

Abstract

Background

Recombinant immunotherapeutic fusion proteins (rIFPs) were designed to target triple-negative breast cancer (TNBC). This is a heterogeneous and aggressive subset of breast cancer accounting for 15-20% of all diagnosed breast cancer cases, with women of premenopausal age and African descent inordinately predisposed. Based on previous studies, we hypothesised that the ability to bind to two identical antigens through a bivalent antibody increases the total strength of the reaction and that increasing the affinity and valency of tumour-targeting antibodies results in improved tumour uptake. Furthermore, the rate of internalisation and intracellular routing of rIFPs may be significant for their therapeutic efficacy. Understanding these factors could impact the use of such biopharmaceuticals for targeted treatment with relevant cell surface biomarkers.

Methods

Mono- and bivalent rIFPs targeting a tumour-associated antigen were transiently expressed in mammalian cell culture and purified using ion metal affinity chromatography (IMAC). Following SDS-PAGE and Western Blot protein analysis, the proteins were fluorescently labelled and the differences between the mono- and bivalent rIFP formats were evaluated in vitro using confocal imaging. The efficacy in binding to targeted cell surface receptors, rate of internalisation, and intracellular routing of internalised rIFPs were evaluated to determine such differences. Differences in rIFP-mediated cytotoxicity were evaluated in vitro using XTT-based cell viability assays.

Results

The rIFPs were successfully expressed, purified, and characterised. Imaging results indicate that the bivalent rIFPs display increased binding affinity and faster internalisation rate compared to the monovalent counterpart when applied to a confirmed antigen-positive TNBC cell line. This is correlated with an enhanced cytotoxic effect of the bivalent rIFP.

Conclusions

The results of this study may have implications for the future designs of rIFPs, however, in vivo pharmacokinetic studies are needed to further elucidate the effect of valency on the efficacy of recombinant immunotherapeutic fusion proteins.

Collapse
Poster Display (ID 17) Poster Display

179P - Early Proof of Concept of Safety and Clinical Activity of Clonal Neoantigen Reactive T Cells (cNeT) (ID 722)

Presentation Number
179P
Lecture Time
12:30 - 12:30
Speakers
  • M. Forster (London, London, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Forster (London, London, United Kingdom)
  • J. Cave (Southampton, United Kingdom)
  • A. Greystoke (Newcastle-upon-Tyne, Tyne and Wear, United Kingdom)
  • R. Plummer (Newcastle-upon-Tyne, Tyne and Wear, United Kingdom)
  • J. Spicer (London, United Kingdom)
  • F. Thistlethwaite (Manchester, United Kingdom)
  • S. Turajlic (London, United Kingdom)
  • A. Craig (Stevenage, United Kingdom)
  • K. Newton (Stevenage, United Kingdom)
  • M. Saggese (Stevenage, United Kingdom)
  • S. Quezada (London, United Kingdom)
  • K. Peggs (London, United Kingdom)

Abstract

Background

cNeT therapy addresses limitations of TIL therapies including the requirement for high dose lymphodepletion and IL-2 which restricts application to fitter patients, and limited understanding of the active component that limits product optimisation and potency assay development. We have initiated trials evaluating cNeT in advanced, heavily pre-treated patients with NSCLC (CHIRON, NCT04032847) or melanoma (THETIS, NCT03997474) utilising lower dose lymphodepletion and IL-2 (1M IU/m2 sc x10d).

Methods

14 patients (8 CHIRON, 6 THETIS) received a single dose of cNeT characterised using cytokine-based antigen-specific assays, flow cytometry and single cell RNA/TCRseq, that were also used to track cNeT post-infusion. These form the safety data set. Median cNeT dose was 18M cells (0.1-287M), with a median clonal reactivity of 16% (0.2-77%).

Results

There were 28 G≥3 treatment-related AEs with lymphopenia and neutropenia the most common. One cNeT related SAE (ICANS) and 3 episodes of cytokine release syndrome (G≤2) were reported with no high-grade toxicities associated with IL-2. 6 patients have died on study, all due to disease progression.

With 15-115 wk follow up 7 NSCLC patients have ≥2 restaging scans (1 pending). Best response was PR in 1 (33+wk), SD in 5 (ongoing in 2 at 15+, 26+ wk), and PD in 1, with an overall durable clinical benefit ≥18 wk in 3/7 (43%). The PR continues to deepen at 24 wk with a 76% total tumour reduction. Flow cytometry revealed early expansion of CD8+KI67+ T cells previously associated with response to checkpoint inhibitors. Peak expansion of cNeT was seen at d21 coincident with a peak in IL-6. In depth characterisation of the cNeT product using single cell RNA and TCRseq suggests the active component is polyfunctional in nature with reactive T cell clusters bearing signatures of T cell proliferation, IL-2 sensitivity, cytokine secretion and tissue migration.

Conclusions

We demonstrate an encouraging early safety and tolerability profile consistent with further application to a broader patient population along with the potential for deep and durable clinical responses in NSCLC despite low doses of cNeT and reduced dose lymphodepletion and IL-2.

Clinical trial identification

NCT04032847

NCT03997474

Collapse
Poster Display (ID 17) Poster Display

180P - Recalling pre-existing microbiota-specific T cells to target tumors (ID 520)

Presentation Number
180P
Lecture Time
12:30 - 12:30
Speakers
  • J. Carpier (Paris, France)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Carpier (Paris, France)
  • A. Talpin (Paris, France)
  • G. Kulakowski (Paris, France)
  • A. Dos Santos Leite (Tuebingen, Germany)
  • K. Laviolette (Toulouse Cedex 3, France)
  • J. Noguerol (Toulouse Cedex 3, France)
  • T. Mersceman (Paris, France)
  • A. GARNIER (Paris, France)
  • C. Gaal (Paris, France)
  • C. Pereira Oliveira (Paris, France)
  • C. Calderon (Paris, France)
  • L. Boullerot (Besancon, France)
  • M. Malfroy (Besancon, France)
  • F. Strozzi (Paris, France)
  • C. Gouttefangeas (Tuebingen, Germany)
  • O. Adotevi (Besancon, CEDEX, France)
  • O. Joffre (Toulouse Cedex 3, France)
  • J. Gamelas Magalhaes (Paris, France)
  • L. Chêne (Paris, France)
  • C. Bonny (Paris, France)

Abstract

Background

The gut microbiome has the singular property of shaping the immune system through a multitude of antigens that educate the T cell repertoire. Since microbial peptides can share similarities with tumor antigens, cross-reactive memory T cells that recognize both bacterial and tumor-derived peptides can thus emerge from the imprinting of the T cell repertoire by gut commensals. Harnessing the full potential of cross-reactive CD8+ T cells against tumor cells could therefore be achieved through the rational design of a microbiota-derived peptide vaccine.

Methods

Enterome developed a novel peptide-based immunotherapy built on the principle of T cell cross-reactivity and molecular mimicry between bacterial-derived peptides called OncoMimicsTM peptides (OMP) and Tumor-Associated Antigens (TAA). HLA-A2-restricted OMP candidates targeting human TAA were first selected in silico from our proprietary metagenomic bank of human gut microbiota and then confirmed through in vitro binding assays, and in vivo evaluations of their immunogenicity and capacity to induce cross-reactive T cell responses against TAA in HLA-A2/DR1 humanized mice. Finally, the prevalence and functionality of OMP-specific T cells were assessed in human blood from healthy donors using tetramers, in vitro stimulation, and cytotoxicity assays.

Results

Selected OMPs could elicit cross-reactive cytotoxic T cell (CTL) responses against human TAA. This was demonstrated by the propensity of these CTL to secrete IFN-γ upon recognition of OMP or TAA peptides, reject TAA peptide-pulsed splenocytes as well as tumors ectopically expressing the selected TAA targets. We finally demonstrated that OMP-specific memory CD8+ T cells pre-exist in human blood, with high prevalence (>80% in the population), and harbor strong CTL functions against TAA when activated ex vivo with OMPs.

Conclusions

We show that gut microbiota-derived peptides mimicking TAAs can elicit strong immune responses against tumors. The pre-existence of these cross-reactive CD8+ T cells in healthy donors exemplifies the strong promises of this innovative approach to develop efficient therapeutic cancer immunotherapies. To date, two OMP-based immunotherapies are evaluated in 3 clinical trials (Glioblastoma, Adrenal cancers, B lymphomas).

Collapse
Poster Display (ID 17) Poster Display

181P - SELECT: A phase 2 randomized trial evaluating 2 doses of vopratelimab (V) + pimivalimab (P) vs P in TISvopra selected patients (pts) (ID 388)

Presentation Number
181P
Lecture Time
12:30 - 12:30
Speakers
  • M. Gumus (Istanbul, Turkey)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Gumus (Istanbul, Turkey)
  • A. Sukalinskaya (Minsk, Belarus)
  • Z. Andric (Belgrade, Serbia)
  • V. Cheshuk (Kiev, Ukraine)
  • T. Ciuleanu (Cluj-Napoca, Romania)
  • S. Sezgin Goksu (Antalya, Turkey)
  • T. Cil (Adana, TR, Turkey)
  • I. Cicin (Edirne, Turkey)
  • I. Bulat (Chisinau, Moldova)
  • Y. Ostapenko (Kiev, Ukraine)
  • K. Penkov (Saint-Petersburg, Russian Federation)
  • C. Hart (Cambridge, United States of America)
  • M. Lai (Cambridge, United States of America)
  • B. Chaao (Cambridge, United States of America)
  • J. Jimenez (Cambridge, United States of America)
  • A. Sepahi (Cambridge, United States of America)
  • G. Shi (Cambridge, United States of America)
  • S. Trott (Cambridge, United States of America)
  • E. Hooper (Cambridge, United States of America)

Abstract

Background

SELECT is a Phase 2 randomized study of P, an investigational PD-1 inhibitor (PD-1i) plus V, an investigational ICOS agonist, vs P in TISvopra selected, immunotherapy naïve, 2nd line non-small cell lung cancer (NSCLC). Tumor inflammation signature (TIS) at and above a designated cut-off, termed TISvopra, was previously associated with improved clinical outcomes in patients treated with V +/- nivolumab. SELECT was designed to demonstrate superiority of P + V vs P in TISvopra selected pts and evaluate 2 doses of V with different pulsatile target engagement (TE) profiles to reduce T cell exhaustion.

Methods

69 TISvopra positive pts with metastatic NSCLC after 1 prior platinum-containing regimen were randomized 2:1:1 to P 1000 mg (MC1, n=36), P+V 0.1 mg/kg (CC1, n=18) or P+V 0.03 mg/kg (CC2, n=15) q6w. Primary (1°) endpoint was mean % change from baseline size of all measurable lesions averaged over 9 & 18 weeks by independent central radiology review (ICR); the study was powered to compare MC1 vs pooled CC1+CC2. Overall response rate (ORR) per RECIST v1.1 by ICR, progression free survival (PFS), and overall survival (OS) were secondary (2°) endpoints. Safety, TE and association between PD-L1 and efficacy were evaluated.

Results

1° endpoint was numerically better in CC1+CC2 vs MC1 but not statistically significant. CC2 trended favorably for 1° endpoint, ORR, and PFS. ORR (95% confidence interval [CI]) was 40% (16, 68) for CC2, 25% (12, 42) for MC1, 17% (4, 41) for CC1. 6 month (mo) PFS rate (95% CI) was 80% (50, 93) for CC2, 33% (16, 50) for MC1, 29% for CC1 (10, 52). Except for CC2 6 mo PFS rate data are not mature. PD-L1 was evenly distributed across TISvopra pts but not associated with efficacy. Study treatment was well tolerated with 7.2% of pts reporting Grade ≥3 treatment related adverse events (TRAEs). Most common TRAEs were Grade 1/2 hyperthyroidism, hypothyroidism, rash. The pulsatile pattern of V 0.03 mg showed shorter duration of TE vs 0.1 mg.

Conclusions

P +/- V was well tolerated. The 1° endpoint was not met. P+V 0.03 mg/kg trends toward improved clinical benefit in 1° and 2° endpoints and responses are durable. P is active and continues to be used in combination trials. Shorter TE duration of V 0.03 mg/kg may contribute to clinical activity.

Clinical trial identification

NCT04549025

Collapse
Poster Display (ID 17) Poster Display

182P - Cancer vaccines based on whole-tumor-lysate or neoepitopes with validated HLA-binding outperform those with predicted HLA-binding affinity (ID 524)

Presentation Number
182P
Lecture Time
12:30 - 12:30
Speakers
  • L. Kandalaft (Lausanne, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • H. Fritah (Lausanne, Switzerland)
  • M. Graciotti (Lausanne, Switzerland)
  • C. Chiang (Lausanne, Switzerland)
  • R. Petremand (Lausanne, Switzerland)
  • P. Guillaume (Lausanne, Switzerland)
  • J. Schmidt (Lausanne, Switzerland)
  • B. Stevenson (Billerica, United States of America)
  • D. Gfeller (Lausanne, Switzerland)
  • A. Harari (Lausanne, Switzerland)

Abstract

Background

Vaccines represent attractive components of the tumor immunotherapy armamentarium. Although whole tumor-based vaccines and private neoantigens-based vaccines; each have pros and cons, their relative efficacy has never been comprehensively investigated. Furthermore, neoantigens prioritization to enrich for tumor-rejection antigens remains challenging.

Methods

In this study, we used a murine lung cancer model (LLC1) to investigate the effect of immunization with DCs pulsed with oxidized whole tumor lysate (WTL_DC vax) or neoantigen peptides (Neo_DC vax) on tumor growth. Distinct algorithms were used to predict and prioritize LLC1 candidate neoepitopes. Finally, we retrospectively meta-analyzed neoepitopes selection in a cohort of vaccine-treated ovarian and lung patients to improve correlates of immunogenicity and efficacy.

.

Results

WTL-DC vax was more effective in suppressing LLC1 tumor growth than Neo_DC vax (when neoepitopes were selected with any of the commonly-used in silico predictors based on peptide-MHC affinity). In contrast, vaccine efficacy significantly improved when immunogens were selected on the basis of the effective in vitro binding of candidate neoepitopes to cognate MHC alleles that was experimentally determined. Vaccine efficacy also significantly improved when immunogens had pre-validated or predicted (using PRIME1.0) immunogenicity. Finally, as opposed to their in silico scores, the effective in vitro binding efficacy of candidate immunogens better correlated with vaccine efficacy (clinical responses) in cohorts of lung cancer patients and with immunogenicity in cohorts of ovarian cancer patients

Conclusions

Vaccine based on private neoepitopes predicted using common in silico predictors of peptide-MHC affinity are outperformed by WTL-based vaccines. Parameters such as in vitro peptide-MHC binding and peptide immunogenicity significantly improve the prioritization of tumor-rejecting neoepitopes and should be taken into consideration to increase vaccines’ clinical efficacy.

Collapse
Poster Display (ID 17) Poster Display

183P - COMMANDER-001: Initial safety data from a phase I/II dose escalation/expansion study of SQZ-eAPC-HPV, a cell-based mRNA therapeutic cancer vaccine for HPV16+ solid tumors (ID 315)

Presentation Number
183P
Lecture Time
12:30 - 12:30
Speakers
  • J. Moser (Scottsdale, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Moser (Scottsdale, United States of America)
  • M. Pelster (Nashville, United States of America)
  • A. Jimeno (Aurora, United States of America)
  • J. Park (Boston, United States of America)
  • W. Iams (Nashville, United States of America)
  • T. Wise-Draper (Cincinnati, United States of America)
  • J. Jennings (Watertown, United States of America)
  • N. Miselis (Watertown, United States of America)
  • R. Ji (Watertown, United States of America)
  • S. Loughhead (Watertown, United States of America)
  • R. Zwirtes (Watertown, United States of America)
  • M. Warren (Watertown, United States of America)
  • A. Sharei (Watertown, United States of America)
  • H. Bernstein (Watertown, United States of America)
  • M. Gordon (Scottsdale, United States of America)

Abstract

Background

SQZ-eAPC-HPV (eAPC) is an autologous, cell based, HLA-genotype agnostic HPV therapeutic cancer vaccine that leverages clinical and manufacturing experience from the SQZ-PBMC-HPV clinical candidate (NCT04084951). eAPC is engineered from peripheral blood mononuclear cells by using Cell Squeeze® technology to simultaneously deliver 5 mRNAs encoding for full length HPV16 E6 and E7 proteins, CD86, and membrane-bound (mb) IL-2 and mbIL-12 cytokines. By driving co-localized MHC-I antigen presentation (E6/E7), costimulation (CD86), and cytokine signaling (IL-2/IL-12), the eAPCs may enable more potent T cell activation and proliferation while limiting known toxicity of interleukin therapy.

Methods

Part 1A assesses eAPC as a monotherapy with intravenous dosing every 3 weeks (Q3W). In Cohort 1 enrolled patients (pts) receive 0.5 x 106eAPC/kg Q3W for up to 1 year. Following a Bayesian optimal interval design, 3 - 12 pts may be enrolled per cohort. Eligible pts undergo a single leukapheresis. Preconditioning is not required and eAPCs are administered outpatient after the first dose. Adverse events were assessed by CTCAEv5.0. Tumor response was assessed by RECIST1.1. Baseline and on-treatment pharmacokinetic and pharmacodynamic samples were also collected.

Results

4 pts with HPV16+ solid tumors (cervical, 2; oropharyngeal, 1; vaginal, 1) were dosed in Cohort 1. Cell collection to product release was ~1 week; adequate eAPC doses were manufactured for all pts. All pts completed the 28-day dose limiting toxicity (DLT) period without experiencing a DLT. No related serious adverse events were reported. Only one eAPC related treatment emergent adverse event was reported (Grade 1 fall). Enrollment in Cohort 2 has commenced at a dose of 2.5 x 106 eAPC/kg. The presentation may include safety, preliminary efficacy, and pharmacokinetic/pharmacodynamic data from a cut-off proximal to presentation.

Conclusions

eAPC explores the impact of multiplexed mRNA-based engineering of a cancer vaccine. As of the data cut-off for this abstract (17Aug22), it is thus far well tolerated.

Clinical trial identification

NCT05357898

Collapse
Poster Display (ID 17) Poster Display

184P - A Phase I Study of the cancer-specific vaccine FMPV-1 in Healthy Male Subjects to Assess Safety and Immune Response (ID 514)

Presentation Number
184P
Lecture Time
12:30 - 12:30
Speakers
  • N. Singh (Nottingham, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • N. Singh (Nottingham, United Kingdom)
  • R. Miller (Stevenage, Hertfordshire, United Kingdom)
  • S. Arbe-Barnes (Stevenage, United Kingdom)
  • H. Kvalheim Eriksen (Oslo, Norway)
  • E. Inderberg (Oslo, Norway)
  • B. Iverson (Oslo, Norway)
  • J. Eriksen (OSLO, Norway)

Abstract

Background

FMPV-1 is a novel injectable vaccine to generate anti-cancer T cell responses. The vaccine is an immunogenic frameshift mutated transforming growth factor β receptor 2 (TGFβR2) peptide commonly occurring in microsatellite instability cancers (MSI-H). FMPV-1 is administered in combination with the adjuvant granulocyte macrophage colony stimulating factor (GM-CSF). This is the first clinical study to assess the safety and immunogenicity of FMPV-1/GM-CSF in man.

Methods

This was a single centre, open-label study in 16 healthy male subjects. Primary safety endpoints were adverse events (AEs), laboratory tests, vital signs, electrocardiograms (ECGs) and physical examination. Immunogenicity was assessed by FMPV-1-specific delayed type hypersensitivity (DTH) responses and FMPV-1-specific T-cell proliferation. All subjects received FMPV-1 (0.15 mg/injection) plus GM-CSF (0.03 mg/injection) as two separate injections on days 1, 8, 15, 29, and 43. DTH skin reactivity test was performed with FMPV-1 only on days 1, 29 & 43 with DTH response assessment 2 days later. T cell analyses were performed. Subjects were followed to Day 80 with further immunogenicity to be assessed at 6 and 12 months.

Results

16 subjects were enrolled and completed day 80 per protocol. FMPV-1/GM-CSF was well tolerated with no dose limiting toxicities reported, no major protocol deviations and no serious adverse events. Of the 17 treatment-emergent AEs occurring in 11 subjects, all were Grade 1 except 1 unrelated to IMP Grade 2 rise of creatine kinase. Six related AEs occurred in 6 subjects, being Grade 1 pruritus at injection site. No other clinically significant changes to ECG, vital signs or laboratory measures were seen. All subjects were DTH negative at baseline before dosing; after 3 FMPV-1/GM-CSF vaccinations, 8/16 (50%) subjects were positive at Day 31 with 15/16 (94%) at Day 45 after 4 vaccinations. In vitro T cell proliferation post-vaccination increased compared to pre-vaccination in subjects tested, with analysis ongoing.

Conclusions

The excellent safety profile of FMPV-1/GM-CSF and vaccine-specific immune responses in healthy subjects support further clinical development in a patient population for a Phase 2 study in MSI-high colorectal cancer.

Clinical trial identification

NCT05238558

Collapse
Poster Display (ID 17) Poster Display

185P - Interim analysis of the EOGBM1-18 study: Strong immune response to therapeutic vaccination with EO2401 microbiome derived therapeutic vaccine + nivolumab (ID 650)

Presentation Number
185P
Lecture Time
12:30 - 12:30
Speakers
  • W. Wick (Heidelberg, Germany)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • W. Wick (Heidelberg, Germany)
  • J. Gamelas Magalhaes (Paris, France)
  • A. Dos Santos Leite (Tuebingen, Germany)
  • A. IdBaih (Paris, France)
  • M. Vieito Villar (Barcelona, La Coruña, Spain)
  • G. Tabatabai (Tübingen, Germany)
  • A. Stradella (L'Hospitalet de Llobregat, Spain)
  • F. Ghiringhelli (Dijon, France)
  • M. Burger (Frankfurt am Main, Germany)
  • I. Mildenberger (Mannheim, Germany)
  • U. Herrlinger (Bonn, Germany)
  • M. Touat (Villejuif, Cedex, France)
  • P. Wen (Boston, United States of America)
  • A. Wick (Heidelberg, Germany)
  • H. Toussaint (Paris, France)
  • C. Gouttefangeas (Tuebingen, Germany)
  • C. Bonny (Paris, France)
  • J. Paillarse (Paris, France)
  • D. Reardon (Boston, MA, United States of America)

Abstract

Background

EO2401 is a therapeutic vaccine designed to activate memory commensal-specific T cells cross-reacting with tumor-associated antigens (TAAs). EO2401 includes synthetically produced HLA-A2 peptides with molecular mimicry to TAAs (IL13Rα2, BIRC5 and FOXM1) upregulated in glioblastoma, and the CD4 helper peptide UCP2.

Methods

Patients with glioblastoma at first progression after radiotherapy/temozolomide received EO2401 (300µg/peptide, q2w x4 then q4w) with nivolumab (3 mg/kg q2w; EN), or EN with bevacizumab (10 mg/kg q2w; ENB) (NCT04116658). Blood collection was performed at baseline and then every 2-4 weeks. Immune responses were investigated on PBMCs ex vivo or in vitro stimulation (IVS) using tetramer staining, IFN-γ ELISpot and intracellular cytokine staining.

Results

Vaccination with EO2401 induces a strong and durable CD8+ T cell response against bacterial peptides and human TAAs.

Immune monitoring using ELISpot demonstrated T cell responses against the 3 microbiome-derived peptides for 97% of patients investigated ex vivo or after IVS. Cross-reactivity against theTAA target peptides was demonstrated in 96% of patients by IFN-γ ELISpot. Frequency of CD8+ T cells against bacterial peptides after IVS is extremely high, with max value above 40% for EO2317 and EO2318. Ex vivo, CD8+ T cell against at least one of the EO2401 peptides or human peptides are detected in almost all evaluable patients with some patients exhibiting up to 5% of circulating specific CD8+ T cells. Polyfunctionality of CD8+ T cell induce upon vaccination was demontrated in most patients (CD107a,IFN-γ and TNF-α production after stimulation). Memory-specific CD8+ T cell response is found as early as 2 weeks after the 1st vaccination and maintenance of a strong immune response could be detected for more than 10 months.

Conclusions

EO2401 vaccine demonstrates ability to generate fast and durable immune responses in patients treated with E02401/nivolumab +/- bevacizumab. Activation of specific T cells cross-reacting with commensal antigens and TAAs is thereby validated as an efficient approach to activate strong immune responses in a difficult to treat tumor. Updated immuno-monitoring data will be presented.

Clinical trial identification

Trial EOGBM1-18 (NCT04116658)

Collapse
Poster Display (ID 17) Poster Display

186P - Synergistic antitumor effect of gemcitabine combined with Nyeso1-vaccine treatment evaluated in mouse PDAC organoids (ID 712)

Presentation Number
186P
Lecture Time
12:30 - 12:30
Speakers
  • N. Ferreira (Göttingen, Germany)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • N. Ferreira (Göttingen, Germany)

Abstract

Background

Pancreatic adenocarcinoma (PDAC) is projected to significantly increase and become the second cause of cancer-related deaths before 2030. Immunotherapy has revolutionized the treatment of cancer, however, it has failed to show benefits in PDAC treatment. Recent efforts to synergize chemotherapy and immunotherapy have shown promising results in clinical trials. With this rationale, we tested the anti-tumor effect of the combination of the chemotherapeutic agent gemcitabine with a chitosan-based nanoparticle vaccine that contains the immunogenic cancer germline antigen Ny-eso1. Nyeso1-nanovaccine formulation alone or combined with gemcitabine was tested in vitro in mouse PDAC organoids.

Methods

Mouse peripheral blood mononuclear cells (PBMCs) were obtained and stimulated in vitro with Nyeso1- nanovaccine for 48 hours. Mouse PDAC organoids from KPC-orthotopic injected tumors were directly stimulated with Nyeso1-peptide nanovaccine, with respective nanovaccine controls, or indirectly with the Nyeso1- or nanovaccine controls-stimulated PBMCs or with unstimulated PBMCs. Two different gemcitabine concentrations, 0.38 µM and 0.84 µM, were added to the stimulated-PBMCs and the PDAC organoid co-culture system. During 10 days of co-culture, PDAC organoids were imaged using an Incucyte system, and the organoid areas were measured over time in response to treatment.

Results

The addition of Nyeso1-nanovaccine-stimulated PBMCs to the co-culture did not result in an alteration of PDAC organoids growth. However, the combination of Nyeso1-nanovaccine-stimulated PBMCs with gemcitabine led to a total reduction in sizes of PDAC organoids in comparison to PDAC organoids co-cultured with unstimulated- or nanovaccine-formulation stimulated PBMCs. Administration of gemcitabine in the medium of PDAC organoids alone only induced a slight decrease in the size of the PDAC organoids.

Conclusions

Gemcitabine treatment together with Nyeso1-nanovaccine stimulated PBMCs resulted in a synergistic anti-tumor effect in mouse PDAC organoids. Further studies will be carried out to potentiate the antitumor effect of the PBMCs and to analyze antigen-specific response upon nanovaccine treatment.

Collapse
Poster Display (ID 17) Poster Display

187P - Phase 1/2 study of the oral CCR4 antagonist, FLX475, as monotherapy and in combination with pembrolizumab in advanced cancer (ID 309)

Presentation Number
187P
Lecture Time
12:30 - 12:30
Speakers
  • C. Lin (Taipei City, Taiwan)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Lin (Taipei City, Taiwan)
  • T. Kim (Seoul, Korea, Republic of)
  • P. Desai (Saint Petersburg, United States of America)
  • K. Lee (Cheongju, Korea, Republic of)
  • Y. Feng (Tainan City, Taiwan)
  • N. Ngamphaiboon (Bangkok, Thailand)
  • S. Kim (Seoul, Korea, Republic of)
  • M. Yang (Taipei City, Taiwan)
  • J. Muzaffar (tampa, United States of America)
  • B. Chmielowski (Los Angeles, United States of America)
  • P. Swiecicki (Ann Arbor, United States of America)
  • S. Bowyer (Perth, WA, Australia)
  • J. Brahmer (Baltimore, MD, United States of America)
  • M. Chisamore (Whitehouse Station, United States of America)
  • R. Goyal (South San Francisco, United States of America)
  • N. Nasrah (South San Francisco, United States of America)
  • W. Ho (South San Francisco, United States of America)
  • B. Cho (Seoul, Korea, Republic of)

Abstract

Background

Preclinical studies with oral CCR4 antagonists have demonstrated inhibition of suppressive regulatory T cell (Treg) migration into the tumor microenvironment and antitumor efficacy. The FLX475-02 trial is a Phase 1/2, open-label, dose-escalation and cohort expansion study to determine the safety and antitumor activity of the oral CCR4 antagonist FLX475 as monotherapy and in combination with pembrolizumab in subjects with several types of advanced cancer.

Methods

A standard 3+3 design was used in Phase 1 testing FLX475 given orally once daily as monotherapy or in combination with pembrolizumab (200 mg IV Q3 weeks). In Phase 2, expansion cohorts of subjects both naïve to and pretreated with checkpoint inhibitor with tumor types predicted to be enriched for Teff, Treg, and CCR4 ligand expression (i.e. “charged tumors”) – including EBV+ tumors and NSCLC– are being enrolled using a Simon 2-stage design.

Results

Phase 1 dose escalation has been completed and a recommended Phase 2 dose of 100 mg once daily was selected for FLX475. The safety profile of FLX475 was consistent with that previously described in healthy volunteers, and there has been no evidence of increased severity or frequency of adverse events in combination therapy vs either FLX475 or pembrolizumab given alone. In Phase 2 expansion, FLX475 monotherapy has induced complete responses in the first two subjects of five evaluable enrolled with EBV+ NK/T cell lymphoma. In addition, enrollment of the Stage 1 portion has been completed in several Phase 2 expansion cohorts for the combination of FLX475 and pembrolizumab. In a cohort enrolling subjects with non-small-cell lung cancer (NSCLC) not previously treated with checkpoint inhibitor, 4/13 subjects (31%) have had confirmed partial responses (PRs), including several ongoing for over 6 months, meeting criteria to proceed to Stage 2 enrollment.

Conclusions

In this ongoing Phase 1/2 trial of the oral CCR4 antagonist, FLX475, as monotherapy and in combination with pembrolizumab, antitumor activity including complete responses to FLX475 monotherapy and encouraging combination activity have been observed with an acceptable safety profile.

Clinical trial identification

NCT03674567

Collapse
Poster Display (ID 17) Poster Display

188P - IPH5201 as Monotherapy or in Combination with Durvalumab (D) in Advanced Solid Tumours (ID 472)

Presentation Number
188P
Lecture Time
12:30 - 12:30
Speakers
  • M. Imbimbo (Lausanne, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Imbimbo (Lausanne, Switzerland)
  • A. Hollebecque (Villejuif, Cedex, France)
  • A. Italiano (Bordeaux, CEDEX, France)
  • M. McKean (Nashville, United States of America)
  • T. Macarulla (Barcelona, Spain)
  • E. Castanon Alvarez (Las Palmas de Gran Canaria, Spain)
  • B. Carneiro (Providence, United States of America)
  • R. Mager (Phoenix, United States of America)
  • V. Barnhart (Gaithersburg, United States of America)
  • E. Murtomaki (Cambridge, United Kingdom)
  • Y. He (Phoenix, United States of America)
  • Z. Cooper (Phoenix, United States of America)
  • E. Tu (Phoenix, United States of America)
  • A. Linke (South San Francisco, United States of America)
  • C. Fan (Gaithersburg, United States of America)
  • D. Zhou (Gaithersburg, United States of America)
  • A. Boyer Chammard (Marseille, France)
  • C. Paturel (Marseille, France)
  • P. Fraenkel (Phoenix, United States of America)
  • J. Powderly (Huntersville, United States of America)

Abstract

Background

IPH5201 is an anti-CD39 monoclonal antibody that may promote antitumour immunity by increasing immunostimulatory ATP and reducing immunosuppressive adenosine levels in the tumour microenvironment. This first-in-human, multicentre, non-randomised, open-label, phase 1 study (NCT04261075) assessed the safety, efficacy, pharmacokinetics (PK) and pharmacodynamics (PD) of IPH5201 in patients (pts) with advanced solid tumours.

Methods

The study consisted of 2 dose-escalation parts: ascending doses of IPH5201 (100, 300, 1000 and 3000 mg) IV Q3W, and ascending doses of IPH5201 + D 1500 mg IV Q3W. Primary endpoints were safety and tolerability; key secondary endpoints were preliminary efficacy and PK. Biomarkers were assessed as exploratory endpoints.

Results

Overall, 57 pts received treatment (IPH5201, n=38; IPH5201 + D, n=19). Median age was 62 years (range, 41–78); 54.4% were male. Pts had a median of 3 prior therapies (range, 2–11). As of 7 Jul 2022, all pts had discontinued treatment, 89.5% due to disease progression. Treatment-emergent adverse events (AEs) occurred in 96.5% of pts. Treatment-related AEs (TRAEs) occurred in 66.7% of pts (37/57 [64.9%] related to IPH5201; 11/19 [57.9%] related to D); 10.5% had grade 3 TRAEs, with no grade 4 or 5 TRAEs. The most common TRAEs were infusion-related reactions (21.1%), fatigue (17.5%), and nausea, arthralgia, tumour pain and pruritus (each 8.8%). Median duration of exposure to IPH5201 was 9.3 wks (range, 3.0–66.1) and to D was 8.7 wks (range, 3.0–66.1). 22/57 pts (38.6%) had stable disease; there were no partial or complete responses. Median progression-free survival was 1.4 mo (range, 0–15.2); median overall survival was 8.2 mo (range, 1.0–22.1). IPH5201 saturated soluble CD39 at ≥300 mg and CD39 on immune cells at 3000 mg and decreased tumoural enzymatic activity in 5/8 pts. IPH5201 PK were non-linear at ≤300 mg and linear at ≥1000 mg. An indirect response PD model describing the relationship between IPH5201 concentration and free membrane CD39 on monocytes proposed 3000 mg Q3W or Q4W as the recommended phase 2 dose.

Conclusions

IPH5201 was well tolerated when used alone or in combination with D, with no new safety signals identified. PD were consistent with the mechanism of action.

Clinical trial identification

NCT04261075

Editorial acknowledgement

Medical writing support for this abstract, under the direction of the authors, was provided by Werner Gerber of Ashfield MedComms (Kimberley, South Africa), an Inizio company, and was funded by AstraZeneca.

Collapse
Poster Display (ID 17) Poster Display

189P - IL-10 enhances macrophage phagocytosis of cancer cells in response to CD47 blockade and opsonizing antibodies (ID 202)

Presentation Number
189P
Lecture Time
12:30 - 12:30
Speakers
  • A. Maoz (Boston, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Maoz (Boston, United States of America)
  • J. Allen (Cambridge, United States of America)
  • K. Vaccaro (Cambridge, United States of America)
  • J. Velarde (Cambridge, United States of America)
  • K. Weiskopf (Cambridge, CA, United States of America)

Abstract

Background

There is an urgent need to improve the safety and efficacy of immunotherapy, which has focused primarily on the adaptive immune system. Macrophages are the most common infiltrating immune cells in tumors and they can be directed to kill cancer cells by targeting macrophage-specific immune checkpoints, such as the CD47/SIRPa axis. CD47 serves as a “don’t eat me” signal on cancer cells and binds to SIRPa on macrophages to inhibit phagocytosis. Cytokines can influence macrophage polarization states, but how cytokines influence macrophage activation, particularly in the setting of macrophage checkpoint blockade, remains unknown.

Methods

To study how cytokines modulate macrophage anti-tumor function, we conducted an unbiased screen of 113 recombinant human cytokines using a novel co-culture assay that measures long-term interactions between primary human macrophages and human cancer cells. We tested how each cytokine alters macrophage-mediated cytotoxicity at baseline or in the presence of CD47 blockade. In validation studies, we tested more generally if candidate cytokines could enhance the macrophage response to tumor-opsonizing antibodies such as cetuximab (anti-EGFR mAb). We also confirmed the cross-species relevance of these findings in a murine in vitro system.

Results

Surprisingly, we found that despite not having single-agent activity, interleukin 10 (IL-10) synergizes with anti-CD47 therapy to enhance human macrophage anti-tumor function in long-term co-culture with non-small cell lung cancer (NSCLC) cells. These findings were conserved in a murine model of NSCLC in vitro. IL-10 also synergizes with tumor opsonizing antibodies, such as cetuximab, to enhance macrophage killing of cancer cells. IL-10 priming of macrophages enhanced antibody-dependent phagocytosis of multiple cancer cell lines, likely underlying its mechanism of action.

Conclusions

We discovered an unexpected role for IL-10 in enhancing macrophage anti-tumor functions in response to CD47 blockade and tumor-opsonizing antibodies. These findings illuminate a novel strategy to harness macrophages for cancer immunotherapy and provides preclinical rationale for testing this therapeutic combination in patients with cancer.

Collapse
Poster Display (ID 17) Poster Display

190P - Combination of IPH5201, a blocking antibody targeting the CD39 immunosuppressive pathway, with durvalumab and chemotherapies: Preclinical rationale (ID 384)

Presentation Number
190P
Lecture Time
12:30 - 12:30
Speakers
  • C. Paturel (Marseille, France)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • N. Anceriz (Marseille, France)
  • J. Eyles (Northolt, United Kingdom)
  • J. Lapointe (Northolt, United Kingdom)
  • C. Denis (Marseille, France)
  • V. Breso (Marseille, France)
  • R. Courtois (Marseille, France)
  • S. Augier (Marseille, France)
  • L. Brown (Northolt, United Kingdom)
  • N. Luheshi (Macclesfield, United Kingdom)
  • A. Watkins (Northolt, United Kingdom)
  • Z. Cooper (Phoenix, United States of America)
  • E. Tu (Phoenix, United States of America)
  • E. Vivier (Marseille, France)
  • P. Fraenkel (Phoenix, United States of America)

Abstract

Background

CD39 is an extracellular ectonucleotidase highly expressed in the tumor microenvironment (TME) that, sequentially with CD73, contributes to the production of adenosine (Ado), via hydrolysis of adenosine triphosphate (ATP). IPH5201 is a blocking anti-CD39 monoclonal antibody (mAb) that may promote antitumor immunity by accumulating immunostimulatory ATP released by necrotic cells and reducing immunosuppressive Ado levels in the TME. Targeting the Ado pathway has recently been reported to improve Durvalumab (D) efficacy in early-stage NSCLC patients, through the use of Oleclumab, an anti-CD73 mAb. In a first-in-human study (NCT04261075), IPH5201 was well tolerated alone or in combination with D. Here we explored preclinical efficacy of IPH5201 in combination with anti-PD-L1 and chemotherapies (CT).

Methods

CD39 expression was assessed in early and late stage NSCLC biopsies. In vitro, CT were assessed for their ability to induce ATP release by dying tumor cells and IPH5201 was evaluated for its ability to accumulate the released ATP. In vivo, human (hu) CD39 Knock-In (KI) mice were used to engraft syngeneic tumors and evaluate the efficacy of a mouse (mo) version of IPH5201 (moIPH5201) alone or in combination with CT and anti-PD-L1.

Results

CD39 was expressed in both squamous and adenocarcinoma subtypes of NSCLC with no difference according to the stage of the disease. In a mouse tumor model engrafted in huCD39KI mice, moIPH5201 was able to decrease the human CD39 enzymatic activity and to lower the Ado level in situ. In vitro, CT induced ATP release by tumor cells and IPH5201 was able to accumulate the released ATP. Finally, in vivo, in a mouse tumor model engrafted in huCD39KI mice, moIPH5201 improved the anti-tumor efficacy of gemcitabine and anti-PD-L1 combination.

Conclusions

IPH5201 was shown to block CD39 enzymatic activity, to lower Ado and increase ATP levels in the TME and finally to improve anti-tumor efficacy in preclinical models. Altogether, the expression profile of CD39 in early stage NSCLC and preclinical combination data support the clinical evaluation of IPH5201 in combination with D and CT in early stage NSCLC patients.

Collapse
Poster Display (ID 17) Poster Display

191P - Preliminary biomarker and safety results of SQZ-PBMC-HPV at RP2D in monotherapy and combination with checkpoint inhibitors in HLA A*02+ patients with recurrent, locally advanced, or metastatic HPV16+ solid tumors (ID 574)

Presentation Number
191P
Lecture Time
12:30 - 12:30
Speakers
  • A. Jimeno (Aurora, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Jimeno (Aurora, United States of America)
  • N. Miselis (Watertown, United States of America)
  • J. Park (Boston, United States of America)
  • J. Jennings (Watertown, United States of America)
  • N. Dhani (Toronto, Canada)
  • U. Holtick (Cologne, Germany)
  • W. Iams (Nashville, United States of America)
  • K. Rodabaugh (Omaha, United States of America)
  • N. Nair (Basel, Switzerland)
  • M. Kornacker (Basel, Switzerland)
  • S. Loughhead (Watertown, United States of America)
  • H. Bernstein (Watertown, United States of America)
  • R. Zwirtes (Watertown, United States of America)
  • R. Ji (Watertown, United States of America)
  • M. Warren (Watertown, United States of America)
  • A. Sharei (Watertown, United States of America)

Abstract

Background

Cancer vaccines strive to produce robust, antigen-targeted, T cell-mediated anti-tumor responses, but have struggled to induce tumor regression in patients. The Cell Squeeze® system achieves efficient antigen presentation by delivering target antigens directly into the cytosol of a patient’s PBMCs. SQZ-PBMC-HPV is an autologous cell vaccine that presents epitopes of HPV16 viral oncoproteins on HLA-A*02 to induce CD8 T cell activation. Monotherapy data showed tumor inflammation markers that may indicate correlation with clinical response motivating expansion of the monotherapy and combination with immune checkpoint inhibitors (ICIs).

Methods

SQZ-PBMC-HPV-101 enrolls patients (pts) with advanced HPV16+ cancers who have progressed after standard therapy, have an ECOG 0-1, adequate organ function, and measurable lesion(s) per RECIST1.1. Manufacturing of the cell product takes <24 hours with a collection-to-release time of about 1 week. The RP2D dose of SQZ-PBMC-HPV is 5x106 live cells/kg double prime, given IV q3w. Pts also receiving atezolizumab or ipilimumab are dosed IV q3w. Blood samples and paired biopsies are collected for pharmacodynamic (PD) analyses.

Results

As of Aug 17, 2022, 14 pts have been treated at the RP2D +/- ICI [head and neck (10), cervical (2), vulvar (1), and anal (1)]. These pts have received a median of 3 prior lines of systemic cancer therapy. Treatment was well-tolerated with no dose limiting toxicities, Grade (G) >2 related SAEs, related G >3 AEs, or cytokine release syndrome. In addition to assessment by RECIST1.1 criteria, 8 pts in the monotherapy cohort provided paired tumor biopsies for assessment of changes in the tumor microenvironment including CD8 influx, Treg presence, antigen expression, and evasion mechanisms (MHCI and PD-L1). This report will be the first to summarize the PD and safety endpoints for pts receiving SQZ-PBMC-HPV + ICI therapy.

Conclusions

Building on encouraging earlier monotherapy response data with SQZ-PBMC-HPV, this monotherapy and ICI combination data will further elucidate the impact of this therapeutic vaccine’s mechanism and evaluate its place in patient treatment.

Clinical trial identification

NCT04084951

Collapse
Poster Display (ID 17) Poster Display

192P - Safety and clinical activity of IOA-244, a highly selective phosphoinositide 3-kinase inhibitor delta (PI3Kδ), in a Phase I First in Human (FIH) study. (ID 499)

Presentation Number
192P
Lecture Time
12:30 - 12:30
Speakers
  • A. Di Giacomo (Siena, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Di Giacomo (Siena, Italy)
  • F. Santangelo (Siena, Italy)
  • G. Amato (Siena, Italy)
  • E. Simonetti (Siena, Italy)
  • J. Graham (Glasgow, United Kingdom)
  • M. Lahn (Geneva, Switzerland)
  • L. Van der Veen (Geneva, Switzerland)
  • T. Hammett (Geneva, Switzerland)
  • C. Pickering (Geneva, Switzerland)
  • M. Durini (Milan, Italy)
  • T. Ziyang (Stockholm, Sweden)
  • T. Lakshmikanth (Stockholm, Sweden)
  • P. Brodin (Stockholm, Sweden)
  • M. Occhipinti (Liège, Belgium)
  • M. Simonelli (Milan, Italy)
  • C. Carlo-Stella (Milan, Italy)
  • A. Santoro (Rozzano, (MI), Italy)
  • P. Spiliopoulou (Toronto, Canada)
  • T. Evans (Glasgow, United Kingdom)
  • M. Maio (Siena, Italy)

Abstract

Background

The highly selective inhibitor of PI3Kδ, IOA-244, has a unique safety and pharmacodynamic profile showing immune modulation in patients with solid tumours.

Methods

IOA-244 was investigated in a two-part FIH study: Part A investigated the safety and pharmacokinetics of continuous daily dosing of IOA-244 at 10, 20, 40 and 80 mg. Primary objective: safety of the anticipated biologically effective dose (BED), or of the recommended phase 2 dose (RP2D). Secondary objectives: PK; PD (e.g., inhibition of CD63 expression on basophils, changes in immune cell subsets in peripheral blood); RECIST 1.1.-based responses; PFS and OS. Exploratory studies: changes in circulating immune cells by mass cytometry (Cytometry by Time of Flight; CyTOF); response assessments by radiomics. Analysis of Part B expansion cohorts will be reported at future meetings.

Results

Part A Solid Tumor: Sixteen patients (pts) were treated in 4 cohorts each with 4 pts. Pts characteristics: uveal melanoma (9/16; 56%), cutaneous melanoma (5/16; 31%) and pleural mesothelioma (2/16; 13%). There were no DLTs or treatment-emergent adverse events (TEAE) leading to study drug dose modification. 7/16 pts (44%) were treated for more than 6 months and had SD during this period. At the estimated IC90 (80 mg QD), patients had improvements of their liver enzymes unless tumour progressed in the liver. Four patients at lower dose levels were increased to 80 mg QD with no additional toxicities. 5/16 were treated after RECIST 1.1-defined PD and 2/4 are still on IOA-244 (>2 years on treatment). The median OS has not been reached. In the UM group 2/9 patients are still on IOA-244 (>18 months on treatment; Median OS: 5.4 - not reached)

Part A Follicular Lymphoma: Eight patients were treated at 20 mg (4/4 pts) and 80 mg QD (4/4). No DLT. At 80 mg, there was one PR for 4 months and 1 transient clinical response for 6 weeks. All patients had low circulating blood Tregs during the treatment.

Conclusions

IOA-244 at the 80 mg dose shows less than 5% Grade 3/4 toxicities. In contrast to other PI3Kδ inhibitors, long-term administration of IOA-244 (>6 months) was not associated with diarrhoea or hepatic toxicity.

Clinical trial identification

NCT: NCT04328844.

Collapse
Poster Display (ID 17) Poster Display

193P - CLAUDIN-1 Targeting Antibodies in Solid Tumors: from ALE.C04 to CLAUDIN-1 Oncology Platform (ID 667)

Presentation Number
193P
Lecture Time
12:30 - 12:30
Speakers
  • A. Toso (Allschwil, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Toso (Allschwil, Switzerland)
  • G. Teixiera (Allschwil, Switzerland)
  • T. Zimmermann (Allschwil, Switzerland)
  • D. Schmitter (Allschwil, Switzerland)
  • M. Meyer (Allschwil, Switzerland)
  • M. Muller (STRASBOURG, France)
  • L. Mailly (Strasbourg, France)
  • T. Baumert (Strasbourg, France)
  • R. Iacone (Basel, Switzerland)

Abstract

Background

Claudin 1 (CLDN1) is a protein confined within the normal epithelial tight junctions of different tissues. Upon malignant transformation, CLDN1 is overxpressed and epitopes become exposed outside the tight junctions (non-Junctional CLDN1). ALE.C04 is a highly specific humanized monoclonal antibody that recognizes a unique CLDN1 exposed epitope in different solid tumors.

Methods

More than 1200 paraffin embedded tumor biopsies from 12 different indications have been stained by Immunohistochemistry (IHC) for both CLDN1 and CD3 (T cell marker). Different tumor mouse models have been used to assess the anti-tumor activity of ALE.C04 as single agent and/or in combination with either check-point inhibitors (CPIs) /or chemotherapy

Results

Herein we show that non-Junctional CLDN1 (NJ-CLDN1) is frequently overexpressed in solid tumors. Notably, CLDN1 expression on tumor cells positively correlates with the localization of T-cells into a fibrotic tissue environment. T cell exclusion is one the mechanisms described to hinder the efficacy of Checkpoint inhibitors (CPIs). On this line, the overexpression of Cldn1 in Hepa1-6 mouse liver tumor cells promoted T cell exclusion and resistance to anti-PD1 treatment. Importantly, ALE.C04 restored both T-cell infiltration and anti-PD1 efficacy in Hepa1-6 Cldn1+ tumors. Mechanistically, NJ-CLDN1 interacts with different component involved in extracellular matrix remodeling, thus establishing a physical barrier that exclude immune cells from the tumor nest. ALE.C04 has a direct antifibrotic effect that perturbs the interface between CDLN1+ tumor cells and the stroma, thus restoring im mune cell infiltration. The rationale for developing a CDLN1-based Oncology platform will be also presented.

Conclusions

We have shown that NJ-CLDN1 is overexpressed in different solid tumor, drives T -cell exclusion and resistance to CPIs. Importantly, ALE.C04 restored T cell infiltration and CPI efficacy in a mouse model for liver tumor . Based on our pre-clinical data, rationale and safety profile, we plan a Phase1b with ALE.C04 in combination with CPIs in solid tumors.

Clinical trial identification

INT: 1011

Collapse
Poster Display (ID 17) Poster Display

194P - Thermal ablation followed by intratumoral injection of a novel immune stimulant IP-001 in patients with advanced solid tumors: Phase IB part of study SAKK 66/17 (ID 707)

Presentation Number
194P
Lecture Time
12:30 - 12:30
Speakers
  • M. Joerger (St. Gallen, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Joerger (St. Gallen, Switzerland)
  • P. Knüsel (Chur, Switzerland)
  • E. Alejandre-Lafont (St.Gallen, Switzerland)
  • Y. Metaxas (Muensterlingen, Switzerland)
  • M. Mark (Chur, Switzerland)
  • R. Von Moos (Chur, Switzerland)
  • K. Gysel (Bern, Switzerland)
  • K. Eckhardt (Bern, Switzerland)
  • J. Glaus Garzon (Bern, Switzerland)
  • K. Koster (St. Gallen, Switzerland)
  • Y. Wittwer (St.Gallen, Switzerland)
  • S. Tissot (Lausanne, Switzerland)
  • L. Flatz (Tuebingen, Germany)
  • L. Alleruzzo (Saint Louis, United States of America)
  • S. Lam (St.Louis, United States of America)
  • D. Anderson (St.Louis, United States of America)
  • W. Chen (St.Louis, United States of America)
  • E. Baskin-Bey (St.Louis, United States of America)
  • T. Hode (St.Louis, United States of America)

Abstract

Background

The induction of immunogenic cell death by thermal ablation (TA) with local immunostimulation is a novel approach to address advanced, immune “cold”, treatment-refractory cancer. Glycated N-acetylglucosamine polymer, IP-001® , is a novel immune stimulant that drives the activation of innate immune cells through multiple pathways (Stimulator of Interferon Genes STING, Inflammasome, and others) and acts as an antigen depot, which may enhance downstream adaptive anti-tumor immunity. When combined with TA, IP-001 stimulated abscopal effects at distant, untreated tumors, and immune memory in animal models. Safety of IP-001 was tested in the Phase IB part of the SAKK66/17 Phase 1b/2 trial.

Methods

Patients (pts) with advanced solid tumors who have failed standard treatment received up to 6 four-week cycles of TA using a laser device (TRANBERG Thermal Therapy System) on an accessible lesion at a time followed by intratumoral injection of IP-001 (single dose of 4 ml, with possible de-escalation). Primary endpoint: Dose limiting toxicities (DLTs). Key secondary endpoints: Disease control per RECIST/iRECIST and safety.

Results

Four pts (2F/2M) were enrolled; 2 NSCLC; 1 leimyosarcoma; 1 follicular thyroid cancer FTC; median age=66-yrs. All pts were metastatic and had received multiple prior lines of chemo- and / or immunotherapies including nivolumab (n=2), olaratumab (n=1) and lenvatinib (n=2). Treatment related AEs of grade ≥ 2, included fatigue (50%), fever (25%), hypotension (25%), maculopapular rash (25%), increased CRP (25%) and abdomial pain (25%). No DLTs were observed and no SAEs were considered related to IP-001.

Conclusions

TA followed by IP-001 at a dose of 4 mL is well tolerated in pts with advanced solid tumors who have failed standard treatment. The combined use of thermal tumor ablation and IP-001® at a dose of 4 ml can safely be administered in patients with advanced solid malignancies with the potential to stimulate an abscopal effect via immune stimulation.

Clinical trial identification

NCT03993678

Editorial acknowledgement

NA

Collapse
Poster Display (ID 17) Poster Display

195TiP - A First-In-Human, Phase 1 a/b Dose Escalation and Expansion Study to Evaluate RBS2418 as Monotherapy and in Combination with Pembrolizumab in Subjects with Advanced Unresectable, Recurrent or Metastatic Tumors (NCT05270213) (ID 349)

Presentation Number
195TiP
Lecture Time
12:30 - 12:30
Speakers
  • J. Glenn (Sunnyvale, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Glenn (Sunnyvale, United States of America)
  • I. Csiki (Glendora, United States of America)
  • R. Boccia (Bethesda, United States of America)
  • C. Chen (Palo Alto, United States of America)
  • M. Gordon (Scottsdale, United States of America)
  • J. Misleh (Phoenix, United States of America)
  • J. Powderly (Huntersville, United States of America)
  • Z. Wainberg (Los Angeles, United States of America)
  • A. Spira (San Antonio, United States of America)
  • K. Klumpp (Sunnyvale, United States of America)

Abstract

Background

ENPP1 is a type II transmembrane protein with nucleotide pyrophosphatase, and phosphodiesterase enzymatic activities and its expression is associated with poor prognosis in cancer. ENPP1 inhibition protects cGAMP and ATP from hydrolysis and reduces adenosine levels in the TME, activates APCs and increases T-cell infiltration promoting anticancer immunity. RBS2418 is a potent oral ENPP1 inhibitor. The current study evaluates RBS2418 as monotherapy and combination with pembrolizumab (pembro), supporting preclinical data and complementary mechanism of action with the aim of generating improved anti-tumor immunity and overcoming checkpoint inhibitor resistance in the combination setting. First clinical experience with RBS2418 was within an EA IND to an advanced adrenal cancer patient where the drug was safe, well tolerated and immunologically active.

Trial Design

This is a Phase 1a/b, open-label, non-randomized study in patients (pts) (≥18 years) with advanced, unresectable, recurrent or metastatic solid tumors, who have progressed on, or are ineligible for standard therapies. The study begins with dose-escalation where in a 3+3 fashion pts will receive in one cohort RBS2418 starting at 100mg bid as monotherapy and in a second cohort RBS2418 in combination with pembro (200 mg IV q3w). In the dose-escalation portion, fixed dose as the 10X OBA (OBA as the dose of RBS2418 that inhibits >90% of ENPP1 enzyme activity in human serum) or MTD will be determined, to be used in the expansion cohort which will enroll pts as monotherapy and combination therapy with pembro in tumor types where pembro is approved. The study will enroll up to 64 pts at 10 sites in the US. The primary objectives of the study are safety, tolerability, ENPP1 inhibition and cGAMP protection of RBS2418 as monotherapy and in the combination setting. Immune biomarkers predictive of anti-tumor responses will be explored as well as translational analyses will be done to explore the depth and breadth of immune activation both in tumor tissue and blood. This study is being funded by Riboscience, LLC.

Clinical trial identification

A First-In-Human, Phase 1 a/b Dose Escalation and Expansion Study to Evaluate RBS2418 as Monotherapy and in Combination with Pembrolizumab in Subjects with Advanced Unresectable, Recurrent or Metastatic Tumors (NCT05270213)

Released June, 2022

Collapse
Poster Display (ID 17) Poster Display

196TiP - A Phase 1 study of REGN6569, a GITR monoclonal antibody (mAb), in combination with cemiplimab in patients with advanced solid tumour malignancies (ID 415)

Presentation Number
196TiP
Lecture Time
12:30 - 12:30
Speakers
  • N. Lakhani (Grand Rapids, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • N. Lakhani (Grand Rapids, United States of America)
  • O. Hamid (Los Angeles, CA, United States of America)
  • I. Braña (Barcelona, Spain)
  • P. Reguera Puertas (Madrid, Spain)
  • M. Lopez Criado (Madrid, Spain)
  • P. Swiecicki (Ann Arbor, United States of America)
  • M. De Miguel Luken (Madrid, Spain)
  • M. Gil Martín (L'Hospitalet de Llobregat, Spain)
  • H. Khong (Tampa, United States of America)
  • V. Moreno Garcia (Madrid, Spain)
  • M. Lostes Bardaji (Barcelona, Spain)
  • F. Sun (Tarrytown, United States of America)
  • S. Sandigursky (Tarrytown, United States of America)
  • M. Zambrano (Tarrytown, United States of America)
  • M. Cristea (Tarrytown, United States of America)
  • M. Fury (New York, NY, United States of America)

Abstract

Background

REGN6569 is a fully human immunoglobulin G1 mAb that is highly specific for glucocorticoid-induced tumour necrosis factor receptor–related protein (GITR). GITR is expressed on several immune cell subtypes, notably regulatory T cells (Tregs). REGN6569 has demonstrated in vitro antibody-dependent cell-mediated cytotoxicity (ADCC) with greater cytotoxicity against GITR expressing Tregs than CD8+ T cells.

GITR mAbs have shown preclinical antitumour efficacy. Anti-GITR and anti–programmed cell death-1 (PD-1) combination therapy has synergistic biological activity and may optimise immune checkpoint inhibitor (ICI) treatment in patients with advanced solid tumours.

Trial Design

This ongoing first-in-human study (NCT04465487) will assess safety, tolerability, pharmacokinetics, pharmacodynamics and antitumour activity of REGN6569 in combination with cemiplimab (anti-PD-1) in patients with advanced solid tumours, for which ICI therapies have not been approved or are not available. Patients must be at least 18 years old, have Eastern Cooperative Oncology Group performance status (0 or 1), and be naive to ICIs.

The study includes dose-escalation (a “4+3” design; in advanced solid tumours) and dose-expansion (in patients with advanced head and neck squamous cell carcinoma) parts. The dose-escalation part will cover 5 dose levels (DL1-DL5) for REGN6569, administered intravenously (IV) every 3 weeks (Q3W). Patients will receive REGN6569 as monotherapy lead-in for the first dose followed by REGN6569 in combination with standard cemiplimab 350 mg IV Q3W. Dose expansion will open after review of safety, efficacy and pharmacodynamic data from the dose-escalation phase. The primary objective of the dose-escalation phase is to evaluate safety and tolerability. The co-primary objectives of the dose-expansion phase are to assess preliminary efficacy of REGN6569 in combination with cemiplimab, as measured by objective response rate, and preliminary pharmacodynamic activity of REGN6569 as lead-in monotherapy, as measured by intratumoural GITR+ Treg depletion. The study is currently open to enrolment.

Clinical trial identification

NCT04465487

Editorial acknowledgement

Medical writing support was provided by Sameen Yousaf, PhD, of Prime, Knutsford, UK, funded by Regeneron Pharmaceuticals, Inc. Responsibility for all opinions, conclusions, and data interpretation lies with the authors.

Collapse
Poster Display (ID 17) Poster Display

197TiP - First-in-human (FIH) Phase 1/2 study of ubamatamab, a MUC16xCD3 bispecific antibody, administered alone or in combination with cemiplimab in patients with recurrent ovarian cancer (OC) (ID 445)

Presentation Number
197TiP
Lecture Time
12:30 - 12:30
Speakers
  • K. Moore (Oklahoma City, Oklahoma, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • K. Moore (Oklahoma City, Oklahoma, United States of America)
  • S. Bouberhan (Boston, United States of America)
  • E. Hamilton (Nashville, United States of America)
  • J. Liu (Boston, MA, United States of America)
  • R. O'Cearbhaill (New York, NY, United States of America)
  • D. O'Malley (Columbus, United States of America)
  • K. Papadimitriou (Edegem, Belgium)
  • D. Schröder (Charleroi, Belgium)
  • E. Van Nieuwenhuysen (Leuven, Belgium)
  • S. Yoo (Sleepy Hollow, United States of America)
  • M. Peterman (Sleepy Hollow, United States of America)
  • P. Goncalves (Tarrytown, MG, United States of America)
  • T. Schmidt (Sleepy Hollow, United States of America)
  • M. Zhu (Sleepy Hollow, United States of America)
  • I. Lowy (Tarrytown, United States of America)
  • T. Uldrick (Tarrytown, United States of America)
  • E. Miller (Tarrytown, United States of America)

Abstract

Background

Mucin-16 is a cell surface glycoprotein that is overexpressed in epithelial OC. Ubamatamab (REGN4018) is a mucin-16 x cluster of differentiation 3 (MUC16xCD3) bispecific antibody that promotes T cell–mediated cytotoxicity by facilitating contact between cancer cells and T cells. Cemiplimab is an anti–programmed cell death-1 monoclonal antibody.

The Phase 1 study in patients with recurrent OC found ubamatamab monotherapy had an acceptable safety profile, durable clinical activity across a range of doses (as measured by RECIST and cancer antigen 125 [CA-125] response rates), and linear pharmacokinetics up to 800 mg weekly intravenous (IV)1. These clinical data support further evaluation of a once every 3-week (Q3W) regimen of ubamatamab alone and in combination with cemiplimab.

Trial Design

In phase 2, up to 150 patients with advanced platinum-resistant OC and elevated serum CA-125 will be randomised to three IV arms (1:1:1) to receive ubamatamab 250 mg IV Q3W or 800 mg IV Q3W as monotherapy, or ubamatamab 250 mg IV Q3W in combination with cemiplimab 350 mg Q3W. All arms will include weekly step-up dosing of ubamatamab (1 mg week 1, 20 mg week 2, and full dose weeks 3 and 4) to limit risk of cytokine release syndrome prior to proceeding to Q3W dosing. Expansion cohorts will use a Simon 2-stage study design, with an interim analysis after the first 20 patients. Any arm with ≥3 objective responses will be expanded to 50 patients.

In this dose expansion phase the primary endpoint will be the objective response rate for each arm as defined by RECIST 1.1 criteria. Secondary endpoints include evaluation of duration of response and progression-free survival as well as further evaluation of safety and pharmacokinetics. Exploratory endpoints include evaluation of baseline tumour MUC16 immunohistochemistry expression and other biomarkers as predictors of response. The impact of ubamatamab on quality of life and physical functioning will also be assessed.

Final data are pending for this TiP.

References

1. Annals of Oncology (2022) 33 (suppl_7): S235-S282. 10.1016/annonc/annonc1054

Clinical trial identification

NCT03564340

Editorial acknowledgement

Medical writing support was provided by Rachel McGrandle, BSc, of Prime, Knutsford, UK, funded by Regeneron Pharmaceuticals, Inc.

Collapse
Poster Display (ID 17) Poster Display

198TiP - DEDALUS trial: a single-arm, phase 2, multi-center study of chemo-immunotherapy followed by hypo-fractionated RT and maintenance immunotherapy in patients with unresectable stage III NSCLC (ID 516)

Presentation Number
198TiP
Lecture Time
12:30 - 12:30
Speakers
  • J. Saddi (Monza, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Saddi (Monza, Italy)
  • F. Agustoni (Pavia, Italy)
  • S. Arcangeli (Milano, Italy)
  • D. Cortinovis (Monza, Italy)
  • A. Ferrari (Pavia, Italy)
  • D. Cicognini (Pavia, Italy)
  • C. Klersy (Pavia, Italy)
  • P. Pedrazzoli (Pavia, Italy)
  • U. Malapelle (Napoli, Italy)
  • F. Grossi (Busto Arsizio, Italy)
  • A. Filippi (Pavia, Italy)

Abstract

Background

PACIFIC regimen is the gold standard for patients with unresectable locally advanced Non-Small Cell Lung Cancer (NSCLC).

Moving forward on developing the best treatment combination, this study aims to assess the safety and efficacy of a regimen of induction chemo-immunotherapy followed by de-intensified, hypo-fractionated RT given concurrently with Durvalumab followed by Durvalumab maintenance in patients with unresectable, stage III NSCLC

Trial Design

Dedalus trial is a Phase 2, open-label, single-arm, multi-center, Italian study designed to determine safety of chemo-immunotherapy followed by reduced-dose hypo-fractionated radiotherapy and maintenance immunotherapy with a fixed dose of Durvalumab monotherapy.

45 patients will be enrolled from March 2022 to March 2024.

Patients will meet the following major criteria could be included: NSCLC stage III, PD-L1 all comers, judged unresectable and ineligible for cCRT by thoracic multidisciplinary board and then candidate to sCRT.

After three cycles of Cisplatin/Carboplatin (AUC5) plus Etoposide plus Durvalumab every three weeks, responders will be receiving hypo-fractionated thoracic RT (45 Gy over 3 weeks) plus Durvalumab, followed by Durvalumab maintenance up to 12 months or progression.

Primary endpoint is safety, defined by the incidence of grade 3-4 possibly related adverse events within 6 months from the initiation of treatment.

Secondary objectives are PFS and OS (median and 12 months) and quality of life (investigated through EORTC QLQ-C30 and LC13 questionnaires).

Ancillary studies consist of radiomic and molecular analyses: correlation of radiomic signatures extracted form baseline and post-RT TC-scans with PFS; molecular alterations detected on tumor tissue specimens obtained at diagnosis will be monitored on ctDNA in plasma specimens trough liquid biopsies at 3 different time-points (at baseline, after radiotherapy and at progression).

First patient was enrolled in March 2022; at the time of submission, eight patients are included in the trial.

Clinical trial identification

NCT05128630

Collapse
Poster Display (ID 17) Poster Display

199TiP - A first-in-human phase I study of IPH5301, an anti-CD73 monoclonal antibody, alone or in combination with chemotherapy and trastuzumab, in patients with advanced solid tumors (CHANCES) (ID 298)

Presentation Number
199TiP
Lecture Time
12:30 - 12:30
Speakers
  • A. Gonçalves (Marseille, Cedex 09, France)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Vicier (Marseille, Cedex 09, France)
  • M. Guerin (Marseille, Cedex 09, France)
  • J. Pignon (Marseille, Cedex 09, France)
  • B. Chanez (Marseille, CEDEX 9, France)
  • M. Provansal Gross (Marseille, France)
  • N. Penel (Lille, CEDEX, France)
  • N. Kotecki (Brussels, CEDEX, Belgium)
  • D. Marie (Marseille, France)
  • A. Boyer Chammard (Marseille, France)
  • C. Paturel (Marseille, France)
  • P. André (Marseille, France)
  • J. Pakradouni (Marseille, Cedex 09, France)
  • J. Boher (Paris, Cedex 13, France)
  • F. Awada (Brussels, Belgium)

Abstract

Background

CD73 is an extracellular ectonucleotidase overexpressed in the tumor microenvironment (TME) of multiple cancers. Extracellular adenosine triphosphate (ATP) released by necrotic cells is hydrolyzed into adenosine monophosphate (AMP), which is subsequently degraded by CD73 into immunosuppressive adenosine. CD73 overexpression has been associated with poor prognosis, conferring resistance to chemotherapy and anti-HER2 therapy. IPH5301 is a humanized IgG1 monoclonal antibody (mAb) with a functionally silent Fc domain, specifically inhibiting both soluble and membrane CD73 enzymatic activity, and restoring proliferation of immune T cells more effectively than other anti-CD73 mAbs in clinical development. IPH5301 has the potential to release TME from adenosine-mediated immune suppression, thereby leading to increased anti-tumor immunity.

Trial Design

CHANCES is a first-in-human, open label, European, multi cohort Phase I study (NCT05143970). The dose escalation cohort will evaluate 4 dose levels of IPH5301 administered alone IV every 2 weeks until progression or toxicity. The escalation will be guided by an adaptive Bayesian model, enrolling cohorts of 2 to 3 patients with advanced and/or metastatic cancer. A maximum of 15 patients is planned. In the expansion cohort, IPH5301 will be administered at the recommended dose (RP2D) and the next lower dose (RP2D-1) in combination with trastuzumab and paclitaxel in 12 patients with advanced/metastatic HER2-positive breast and gastric/esogastric cancer. The primary objectives are to evaluate the safety profile, to determine the maximum tolerated dose (MTD) of IPH5301 alone in advanced /metastatic tumors (dose-escalation part) and to recommend a dose of IPH5301 to be combined with paclitaxel and trastuzumab in patients with advanced/metastatic HER2-positive breast and gastric/esogastric cancer (expansion part). The secondary objectives are to evaluate pharmacokinetic, immunogenicity and to assess preliminary clinical activity. Finally, exploratory objectives include pharmacodynamics such as CD73 expression, occupancy and enzymatic activity.

Clinical trial identification

NCT05143970

Initial release 11/17/2021

Collapse
Poster Display (ID 17) Poster Display

Tumour biology and tumour microenvironment (ID 805)

Lecture Time
12:30 - 12:30
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Poster Display (ID 17) Poster Display

201P - Phase II study of taminadenant (A2AR antagonist) + spartalizumab (anti PD-1 antibody) in patients with triple negative breast cancer (TNBC) (ID 297)

Presentation Number
201P
Lecture Time
12:30 - 12:30
Speakers
  • G. Jerusalem (Liège, Belgium)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • G. Jerusalem (Liège, Belgium)
  • F. De Braud (Milan, Italy)
  • M. De Jonge (Rotterdam, Netherlands)
  • P. Grell (Brno, South Moravian, Czech Republic)
  • A. Gonçalves (Marseille, Cedex 09, France)
  • T. Tan (Singapore, Singapore)
  • R. Greil (Salzburg, Austria)
  • T. Yap (Houston, Surrey, United States of America)
  • C. Lin (Taipei City, Taiwan)
  • K. Kornbluth (Cambridge, United States of America)
  • X. Yang (Cambridge, United States of America)
  • C. Vong (Basel, Switzerland)
  • S. CHOUDHURY (Cambridge, United States of America)
  • J. Mataraza (Cambridge, United States of America)
  • L. Lee (Cambridge, United States of America)
  • J. Otero (Cambridge, United States of America)
  • E. Garralda (Barcelona, Spain)

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

Collapse
Poster Display (ID 17) Poster Display

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 664)

Presentation Number
202P
Lecture Time
12:30 - 12:30
Speakers
  • A. Sibille (Liège, Belgium)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Sibille (Liège, Belgium)
  • J. Plumas (La Tronche, France)
  • I. Demedts (Roeselare, Belgium)
  • E. Pons-Tostivint (Nantes, Cedex 1, France)
  • C. Van De Kerkhove (Leuven, Belgium)
  • S. Derijcke (Kortrijk, Belgium)
  • M. Collodoro (Liège, Belgium)
  • K. Al Badawy (Liège, Belgium)
  • C. Duchayne (Liège, Belgium)
  • C. Debruyne (Liège, Belgium)
  • M. Pérol (Lyon, CEDEX, France)
  • E. Buchmeier (Köln, Germany)
  • K. Cuppens (Hasselt, Belgium)
  • J. Vansteenkiste (Leuven, Belgium)

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

Collapse
Poster Display (ID 17) Poster Display

203P - Early remodeling of systemic antitumor T cell immunity in head and neck cancer patients treated by chemoradiation (ID 669)

Presentation Number
203P
Lecture Time
12:30 - 12:30
Speakers
  • B. Lecoester (Besancon, France)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • B. Lecoester (Besancon, France)
  • E. Seffar (Besancon, France)
  • A. Marguier (Besancon, France)
  • A. Renaudin (Besancon, France)
  • T. Richard (Besancon, France)
  • M. Wespiser (Besancon, France)
  • M. Malfroy (Besancon, France)
  • L. Boullerot (Besancon, France)
  • R. Arnier (Besancon, France)
  • E. Martin (Dijon, France)
  • X. Sun (Belfort, France)
  • C. Laheurte (Besancon, France)
  • J. Boustani (Besancon, France)
  • O. Adotevi (Besancon, CEDEX, France)

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

Editorial acknowledgement

The authors would like to thank the French “Ligue contre le cancer”, the Bourgogne Franche Comté regional council, the ARC and the Cancéropôle EST for funding their work on this topic.

Collapse
Poster Display (ID 17) Poster Display

204P - Tumor immune microenvironment and immunotherapy efficacy in BRAF mutation non-small-cell lung cancer (ID 210)

Presentation Number
204P
Lecture Time
12:30 - 12:30
Speakers
  • Y. Fan (Hangzhou, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Y. Fan (Hangzhou, China)
  • H. Li (Hangzhou, China)
  • Y. Zhang (Changsha, China)
  • Y. Xu (Hangzhou, Zhejiang, China)
  • Z. Huang (Hangzhou, China)
  • G. Chen (Hangzhou, China)

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

Collapse
Poster Display (ID 17) Poster Display

205P - Pre-existing tumor host immunity characterization in resected Non-Small Cell Lung Cancer (ID 356)

Presentation Number
205P
Lecture Time
12:30 - 12:30
Speakers
  • L. Pinto (Barcelona, Spain)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. Pinto (Barcelona, Spain)
  • P. Rocha (Barcelona, Spain)
  • M. Rodrigo (Barcelona, Spain)
  • L. Moliner (Manchester, United Kingdom)
  • S. Menendez (Barcelona, Spain)
  • N. Navarro Gorro (Barcelona, Spain)
  • R. Del Rey-Vergara (Barcelona, Spain)
  • M. GALINDO CAMPOS (Barcelona, Spain)
  • A. Taus Garcia (Barcelona, Spain)
  • I. Sánchez (Barcelona, Spain)
  • D. Casadevall Aguilar (Barcelona, Spain)
  • S. Clave (Barcelona, Spain)
  • B. Bellosillo Paricio (Barcelona, Spain)
  • J. Perera (Barcelona, Spain)
  • L. Comerma (Barcelona, Spain)
  • E. Arriola (Barcelona, Spain)

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.

Collapse
Poster Display (ID 17) Poster Display

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 – An EORTC IMMUcan sub-project (ID 108)

Presentation Number
206P
Lecture Time
12:30 - 12:30
Speakers
  • A. Van der Elst (Brussels, Belgium)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Van der Elst (Brussels, Belgium)
  • D. Herrero (Paris, France)
  • M. Morfouace (Brussels, Belgium)
  • R. Galot (Woluwe-Saint-Lambert, Belgium)
  • C. Van Marcke de Lummen (Woluwe-Saint-Lambert, Belgium)
  • S. Lucas (Woluwe-Saint-Lambert, Belgium)
  • J. Machiels (Woluwe-Saint-Lambert, Belgium)

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

Collapse
Poster Display (ID 17) Poster Display

207P - Impact of Lymphatic Vessel Derived Oxysterol on anti-tumor immunity (ID 376)

Presentation Number
207P
Lecture Time
12:30 - 12:30
Speakers
  • M. Sun (Geneva, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Sun (Geneva, Switzerland)
  • L. Garnier (Geneva, Switzerland)
  • D. Brighouse (Geneva, Switzerland)
  • J. Montorfani (Geneva, Switzerland)
  • E. Cosset (Geneva, Switzerland)
  • P. Walker (Geneva, Switzerland)
  • C. Pot-Kreis (Lausanne, Switzerland)
  • T. Petrova (Lausanne, Switzerland)
  • G. Muccioli (Woluwe-Saint-Lambert, Belgium)
  • S. Hugues (Geneva, Switzerland)

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.

Collapse
Poster Display (ID 17) Poster Display

208P - Sex hormones impact the response to immunotherapy in obese male mice. (ID 656)

Presentation Number
208P
Lecture Time
12:30 - 12:30
Speakers
  • A. Pommier (Geneva, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Pommier (Geneva, Switzerland)
  • E. Dupuychaffray (Geneva, Switzerland)
  • H. Poinot (Geneva, Switzerland)
  • M. Alvarez (Geneva, Switzerland)
  • B. Taskoparan (Geneva, Switzerland)
  • C. Vögel (Bern, Switzerland)
  • C. Bourquin (Geneva, Switzerland)

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.

Collapse
Poster Display (ID 17) Poster Display

209P - Single-cell profiling of tumor-associated neutrophils in advanced non-small cell lung cancer (ID 161)

Presentation Number
209P
Lecture Time
12:30 - 12:30
Speakers
  • J. Shi (Shanghai, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Shi (Shanghai, China)
  • F. Wu (Shanghai, China)
  • Z. Zhang (Shanghai, China)
  • C. Zhou (Shanghai, China)

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.

Collapse
Poster Display (ID 17) Poster Display

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 668)

Presentation Number
210P
Lecture Time
12:30 - 12:30
Speakers
  • A. Marguier (Besancon, France)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Marguier (Besancon, France)
  • V. Perreira (Besançon, France)
  • M. Wespiser (Besancon, France)
  • B. Lecoester (Besancon, France)
  • L. Boullerot (Besancon, France)
  • M. Malfroy (Besancon, France)
  • C. Nardin (Besancon, France)
  • C. Laheurte (Besancon, France)
  • E. Orillard (Besancon, France)
  • O. Adotevi (Besancon, CEDEX, France)

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

Editorial acknowledgement

The authors would like to thank the French “Ligue contre le cancer”, the Bourgogne Franche Comté regional council, the ARC, and the Cancéropôle EST for funding their work on this topic.

Collapse
Poster Display (ID 17) Poster Display

211P - A new role for CXCL3 in shaping the metastatic tumor microenvironment (ID 649)

Presentation Number
211P
Lecture Time
12:30 - 12:30
Speakers
  • C. Wolowczyk (Trondheim, Norway)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Wolowczyk (Trondheim, Norway)
  • A. Bokil (Trondheim, Norway)
  • M. Giambelluca (Trondheim, Norway)
  • S. Hak (Trondheim, Norway)
  • G. Bjørkøy (Trondheim, Norway)

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.

Collapse
Poster Display (ID 17) Poster Display

212P - Augmenting Cancer Immunotherapy by Targeting the Fes Kinase (ID 171)

Presentation Number
212P
Lecture Time
12:30 - 12:30
Speakers
  • B. Laight (Kingston, Canada)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • B. Laight (Kingston, Canada)
  • N. Dmytryk (Kingston, Canada)
  • D. Harper (Kingston, Canada)
  • N. Shakfa (Kingston, Canada)
  • V. Hoskin (Ottawa, Canada)
  • I. Shapovalov (Kingston, Canada)
  • Y. Gao (Kingston, Canada)
  • M. Koti (Kingston, Canada)
  • P. Greer (Kingston, Canada)

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.

Collapse
Poster Display (ID 17) Poster Display

213P - Type-1 diabetes restricts melanoma growth by reprogramming intra-tumoral T cell metabolism (ID 272)

Presentation Number
213P
Lecture Time
12:30 - 12:30
Speakers
  • A. Sarkar (Kolkata, India)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Sarkar (Kolkata, India)
  • S. Dhar (Kolkata, India)
  • S. Bera (Kolkata, India)
  • M. Chakravarti (Kolkata, India)
  • A. Verma (Lucknow, India)
  • P. Prasad (Kolkata, India)
  • A. Saha (Kolkata, India)
  • A. Bhuniya (Kolkata, India)
  • I. Guha (Kolkata, India)
  • S. Roy (Kolkata, India)
  • S. Banerjee (Kolkata, India)
  • R. Baral (Kolkata, India)
  • D. Datta (Lucknow, India)
  • A. Bose (Kolkata, India)

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.

Collapse
Poster Display (ID 17) Poster Display

214P - Location matters: oral cancer cells at the tumor invasive border that express GARP exclude immune cells (ID 704)

Presentation Number
214P
Lecture Time
12:30 - 12:30
Speakers
  • R. Van de Ven (Amsterdam, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • R. Van de Ven (Amsterdam, Netherlands)
  • N. Wondergem (Amsterdam, Netherlands)
  • S. Ganzevles (Amsterdam, Netherlands)
  • C. Leemans (Amsterdam, Netherlands)
  • R. Brakenhoff (Amsterdam, Netherlands)

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.

Collapse
Poster Display (ID 17) Poster Display

215P - Proteogenomics to guide neoantigen discovery in non-small cell lung cancer (ID 589)

Presentation Number
215P
Lecture Time
12:30 - 12:30
Speakers
  • Y. Kaminskiy (Stockholm, Sweden)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Y. Kaminskiy (Stockholm, Sweden)

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.2

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.

Collapse
Poster Display (ID 17) Poster Display

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 593)

Presentation Number
216P
Lecture Time
12:30 - 12:30
Speakers
  • R. Martín Lozano (Madrid, Spain)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • R. Martín Lozano (Madrid, Spain)
  • M. Roche-Molina (Madrid, Spain)
  • E. Alvarez (Madrid, Spain)
  • M. Del Monte-Millan (Madrid, Spain)
  • Y. Jerez Gilarranz (Madrid, Spain)
  • F. Moreno Anton (Madrid, Spain)
  • J. García Saenz (Madrid, Spain)
  • I. Echavarria Diaz-Guardamino (Madrid, Spain)
  • T. Massarrah (Madrid, Spain)
  • M. Cebollero (Madrid, Spain)
  • A. Ballesteros Garcia (Madrid, Spain)
  • U. Bohn Sarmiento (Las Palmas de Gran Canaria, Canary Island, Spain)
  • H. Gomez Moreno (Lima, Peru)
  • H. Fuentes (Lima, Peru)
  • B. Herrero Lopez (Santander, Spain)
  • S. Gamez Casado (Madrid, Spain)
  • C. Bueno Muiño (Alcorcon, Madrid, Spain)
  • O. Bueno (Madrid, Spain)
  • S. Lopez-Tarruella Cobo (Madrid, Spain)
  • M. Martin Jimenez (Madrid, Spain)

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.

Collapse
Poster Display (ID 17) Poster Display

217P - Characterization of immune cells in the tumor microenvironment of advanced bladder cancer (ID 620)

Presentation Number
217P
Lecture Time
12:30 - 12:30
Speakers
  • E. Flaberg (Stockholm, Sweden)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • E. Flaberg (Stockholm, Sweden)
  • R. Turkki (Helsinki, Finland)
  • L. Paavolainen (Helsinki, Finland)
  • D. Krantz (Stockholm, Sweden)
  • K. Välimäki (Helsinki, Finland)
  • A. Schoonenberg (Helsinki, Finland)
  • E. Moussaud (Stockholm, Sweden)
  • K. Holmsten (Stockholm, Sweden)
  • F. Costa Svedman (Solna, Sweden)
  • O. Kallioniemi (Stockholm, Sweden)
  • J. Abrahamsson (Lund, Sweden)
  • F. Liedberg (Malmo, Sweden)
  • G. Sjödahl (Malmo, Sweden)
  • T. Pellinen (Helsinki, Finland)
  • A. Ullén (Stockholm, Sweden)
  • P. Östling (Stockholm, Sweden)

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.

Collapse
Poster Display (ID 17) Poster Display

218P - A symbiotic relationship between persistent and emerging resistant oligo-clones dictates resistance to immunotherapy in pancreatic cancer (ID 623)

Presentation Number
218P
Lecture Time
12:30 - 12:30
Speakers
  • C. Ragulan (Sutton, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Ragulan (Sutton, United Kingdom)
  • Y. Ikami (Sutton, United Kingdom)
  • K. Desai (Sutton, United Kingdom)
  • P. Varun Lawrence (Sutton, United Kingdom)
  • S. Mohammed Musheer Aalam (Rochester, United States of America)
  • N. Kannan (Rochester, United States of America)
  • A. Sadanandam (London, United Kingdom)

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.

Collapse
Poster Display (ID 17) Poster Display

219P - Spatial sequencing of T cell repertoire in breast cancer (ID 641)

Presentation Number
219P
Lecture Time
12:30 - 12:30
Speakers
  • A. Merhi (Gosselies, Belgium)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Merhi (Gosselies, Belgium)
  • P. Rosmaninho (Gosselies, Belgium)
  • I. Bar (Gosselies, Belgium)
  • C. Birchall (Woluwe-Saint-Lambert, Belgium)
  • S. Haussy (Brussels, Belgium)
  • D. Petrone (Gosselies, Belgium)
  • J. De Wever (Gosselies, Belgium)
  • A. Devaux (Charleroi, Belgium)
  • J. Canon (Charleroi, Belgium)
  • G. Beniuga (Gosselies, Belgium)
  • F. Duhoux (Woluwe-Saint-Lambert, Belgium)
  • P. Delree (Gosselies, Belgium)
  • J. Carrasco (Charleroi, Belgium)

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.

Collapse
Poster Display (ID 17) Poster Display

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 342)

Presentation Number
220P
Lecture Time
12:30 - 12:30
Speakers
  • K. Boo (Seoul, Korea, Republic of)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • K. Boo (Seoul, Korea, Republic of)

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.

Collapse
Poster Display (ID 17) Poster Display

221P - The role of TNF-alpha in thymus dysfunction during Acute myeloid leukemia (ID 346)

Presentation Number
221P
Lecture Time
12:30 - 12:30
Speakers
  • M. Ben Khoud (Villeneuve D'ascq, France)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Ben Khoud (Villeneuve D'ascq, France)
  • N. Jouy (Lille, France)
  • B. Quesnel (Lille, France)
  • C. Brinster (Lille, France)

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.

Collapse
Poster Display (ID 17) Poster Display

222P - Platinum-based chemotherapy attenuates the CD8 T cell proliferative response to concomitant PD-1 blockade (ID 386)

Presentation Number
222P
Lecture Time
12:30 - 12:30
Speakers
  • A. Mariniello (Orbassano, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Mariniello (Orbassano, Italy)
  • T. Nasti (Atlanta, United States of America)
  • D. Chang (Atlanta, United States of America)
  • S. Malik (Atlanta, United States of America)
  • D. McManus (Atlanta, United States of America)
  • D. McGuire Jr (Atlanta, United States of America)
  • Z. Buchwald (Atlanta, United States of America)
  • S. Novello (Orbassano, Italy)
  • D. Sangiolo (Candiolo, (TO), Italy)
  • G. Scagliotti (Orbassano, TO, Italy)
  • S. Ramalingam (Atlanta, GA, United States of America)
  • R. Ahmed (Atlanta, United States of America)

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.

Collapse
Poster Display (ID 17) Poster Display

223P - Single-cell RNA-seq dissecting the stemness of tumor cells and the tumor microenvironment of liver metastasis in lung cancer (ID 471)

Presentation Number
223P
Lecture Time
12:30 - 12:30
Speakers
  • S. Zheng (Hong Kong, Hong Kong PRC)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Zheng (Hong Kong, Hong Kong PRC)
  • X. Guan (Hong Kong, Hong Kong PRC)

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.

Collapse
Poster Display (ID 17) Poster Display

224P - Immune characterization of de novo metastatic breast cancer (ID 728)

Presentation Number
224P
Lecture Time
12:30 - 12:30
Speakers
  • M. Yernaux (Brussels, Belgium)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Yernaux (Brussels, Belgium)
  • S. Chretien (Lille, France)
  • F. Rothé (Brussels, Belgium)
  • X. Wang (Brussels, Belgium)
  • G. Rouas (Brussels, Belgium)
  • A. Boisson (Brussels, Belgium)
  • N. Kotecki (Brussels, CEDEX, Belgium)
  • A. Mailliez (Lille, France)
  • D. Larsimont (Brussels, Belgium)
  • D. Venet (Brussels, Belgium)
  • C. Sotiriou (Brussels, Belgium)
  • L. Buisseret (Brussels, Belgium)

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.

Collapse
Poster Display (ID 17) Poster Display

225P - Ovarian cancer-cell glucocorticoid receptor activity modulates cytokine secretion promoting infiltration of immunosuppressive cells into the tumor microenvironment (ID 744)

Presentation Number
225P
Lecture Time
12:30 - 12:30
Speakers
  • M. Taya (Dallas, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Taya (Dallas, United States of America)
  • L. Bennett (Dallas, United States of America)
  • S. Conzen (Dallas, United States of America)

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.

Collapse
Poster Display (ID 17) Poster Display

226P - LncRNA DILC and PD-L1 Inhibitors; opposing metastatic and anti-oxidative roles in TME of locally advanced Triple Negative Breast Cancer (ID 300)

Presentation Number
226P
Lecture Time
12:30 - 12:30
Speakers
  • A. Samir (New Cairo, Egypt)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Samir (New Cairo, Egypt)
  • R. Abdeltawab (Cairo, Egypt)
  • U. Stein (Berlin, Germany)
  • H. El Tayebi (New Cairo, Egypt)

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.

Collapse
Poster Display (ID 17) Poster Display

227P - The elusive Luminal B breast tumor and the mysterious chemokines (ID 330)

Presentation Number
227P
Lecture Time
12:30 - 12:30
Speakers
  • R. De Araujo (Uberlandia, MG, Brazil)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • R. De Araujo (Uberlandia, MG, Brazil)
  • F. Da Luz (Uberlândia, Brazil)
  • E. Marinho (Uberlândia, Brazil)
  • C. Nascimento (Uberlândia, Brazil)
  • T. Mendes (Uberlândia, Brazil)
  • E. Mosca (Uberlândia, Brazil)
  • L. Marques (Uberlândia, Brazil)
  • P. Delfino (Uberlândia, Brazil)
  • R. Antonioli (Uberlândia, Brazil)
  • A. Da Silva (Uberlândia, Brazil)
  • M. Monteiro (Uberlândia, Brazil)
  • M. Bernardino Neto (Sao Paulo, Brazil)
  • M. Silva (Uberlândia, Brazil)

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.

Collapse
Poster Display (ID 17) Poster Display

228P - Modelling T Cell-Vasculature interaction in a high-throughput microfluidic platform (ID 700)

Presentation Number
228P
Lecture Time
12:30 - 12:30
Speakers
  • L. De Haan (Oegstgeest, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • L. De Haan (Oegstgeest, Netherlands)
  • J. Suijker (Oegstgeest, Netherlands)
  • L. Van den Broek (Oegstgeest, Netherlands)
  • H. Lanz (Oegstgeest, Netherlands)
  • J. Joore (Oegstgeest, Netherlands)
  • P. Vulto (Oegstgeest, Netherlands)
  • K. Queiroz (Oegstgeest, Netherlands)

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.

Collapse
Poster Display (ID 17) Poster Display

229P - Identify the impact of SARS-CoV-2 on Lung Cancer tumorigenesis using host-pathogen interaction network analysis (ID 542)

Presentation Number
229P
Lecture Time
12:30 - 12:30
Speakers
  • R. Kumar (Kozhikode, India)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • R. Kumar (Kozhikode, India)

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.

Collapse
Poster Display (ID 17) Poster Display

230P - A novel cross-platform concordance analysis using MultiOmyx and PhenoImager multiplexed immunofluorescence (mIF) (ID 495)

Presentation Number
230P
Lecture Time
12:30 - 12:30
Speakers
  • Q. Au (Aliso Viejo, United States of America)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Q. Au (Aliso Viejo, United States of America)
  • H. Nunns (Fort Myers, United States of America)
  • E. Parnell (Aliso Viejo, United States of America)
  • J. Kuo (Fort Myers, United States of America)
  • A. Hanifi (Aliso Viejo, United States of America)
  • S. Pollan (Aliso Viejo, United States of America)
  • N. Tran (Aliso Viejo, United States of America)

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.

Collapse
Poster Display (ID 17) Poster Display

231P - Characterization of the tumor microenvironment in a cohort of KRAS- and EGFR mutant non-small cell lung cancer (ID 289)

Presentation Number
231P
Lecture Time
12:30 - 12:30
Speakers
  • O. Kindler (Graz, Austria)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • O. Kindler (Graz, Austria)
  • K. Maitz (Graz, Austria)
  • P. Moser (Graz, Austria)
  • A. Lueger (Graz, Austria)
  • M. Kienzl (Graz, Austria)
  • M. Runtsch (Graz, Austria)
  • A. Santiso (Graz, Austria)
  • Z. Mihalič (Graz, Austria)
  • J. Feichtinger (Graz, Austria)
  • X. Zhu (Seattle, United States of America)
  • A. Houghton (Seattle, United States of America)
  • J. Kargl (Graz, Austria)

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.

Collapse
Poster Display (ID 17) Poster Display

232P - The efficacy of vasohibin 2 gene expression knockdown and biological relevance in pancreatic cancer cells and tumor associated macrophages. (ID 256)

Presentation Number
232P
Lecture Time
12:30 - 12:30
Speakers
  • M. Kubczak (Lodz, Poland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Kubczak (Lodz, Poland)
  • S. Michlewska (Lodz, Poland)
  • M. Karimov (Leipzig, Germany)
  • A. Ewe (Leipzig, Germany)
  • A. Aigner (Leipzig, Germany)
  • M. Bryszewska (Lodz, Poland)
  • M. Ionov (Lodz, Poland)

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.

Collapse
Poster Display (ID 17) Poster Display

233P - Tumor characteristics of mixed response upon immune checkpoint inhibition (ICI) in advanced melanoma (ID 255)

Presentation Number
233P
Lecture Time
12:30 - 12:30
Speakers
  • J. Versluis (Amsterdam, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Versluis (Amsterdam, Netherlands)
  • E. Hoefsmit (Amsterdam, Netherlands)
  • H. Shehwana (Amsterdam, Netherlands)
  • P. DIMITRIADIS (Amsterdam, Netherlands)
  • J. Sanders (Amsterdam, Netherlands)
  • A. Broeks (Amsterdam, Netherlands)
  • C. Blank (Amsterdam, Netherlands)

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.

Collapse
Poster Display (ID 17) Poster Display

234P - Oncolytic adenovirus-based therapeutics to reprogram the glioblastoma microenvironment for improved CAR T cell therapy (ID 275)

Presentation Number
234P
Lecture Time
12:30 - 12:30
Speakers
  • J. De Sostoa Pomés (Geneva, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. De Sostoa Pomés (Geneva, Switzerland)
  • V. Widmer (Geneva, Switzerland)
  • V. Dutoit (Geneva, Switzerland)
  • D. Migliorini (Geneva, Switzerland)

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.

Collapse
Poster Display (ID 17) Poster Display

235P - Multiplexed Imaging Reveals the Immunomodulatory cGAS as a Predictor of Response in Patients With Malignant Pleural Mesothelioma (ID 313)

Presentation Number
235P
Lecture Time
12:30 - 12:30
Speakers
  • A. Laure (Zurich, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Laure (Zurich, Switzerland)
  • K. Silina (Zurich, CH, Switzerland)
  • M. Kirschner (Zurich, Switzerland)
  • I. Opitz (Zurich, Switzerland)
  • S. Hiltbrunner (Schlieren, Switzerland)
  • A. Curioni-Fontecedro (Zurich, Switzerland)

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.

Collapse
Poster Display (ID 17) Poster Display

236P - A novel microfluidic platform for understanding the role of PDAC stroma in immune response (ID 726)

Presentation Number
236P
Lecture Time
12:30 - 12:30
Speakers
  • M. Geyer (Oegstgeest, Netherlands)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Geyer (Oegstgeest, Netherlands)
  • S. D’agosto (Verona, Italy)
  • H. Lanz (Oegstgeest, Netherlands)
  • V. Corbo (Verona, Italy)
  • J. Joore (Oegstgeest, Netherlands)
  • K. Queiroz (Oegstgeest, Netherlands)

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.

Collapse
Poster Display (ID 17) Poster Display

General interest (ID 806)

Lecture Time
12:30 - 12:30
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Poster Display (ID 17) Poster Display

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 314)

Presentation Number
237P
Lecture Time
12:30 - 12:30
Speakers
  • A. Cortellini (London, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • A. Cortellini (London, United Kingdom)
  • G. Dettorre (London, United Kingdom)
  • U. Dafni (Athens, Attica, Greece)
  • J. Aguilar Company (Barcelona, Spain)
  • L. Castelo-Branco (Lugano, Switzerland)
  • M. Lambertini (Genova, Italy)
  • S. Gennatas (London, United Kingdom)
  • J. Rogado (Madrid, Spain)
  • D. Vinal Lozano (Madrid, Spain)
  • K. Harrington (London, United Kingdom)
  • Z. Tsourti (Athens, Greece)
  • O. Michielin (Lausanne, Switzerland)
  • F. Pommeret (Bordeaux, CEDEX, France)
  • J. Brunet Vidal (Girona, Spain)
  • J. Tabernero (Barcelona, Spain)
  • G. Pentheroudakis (Lugano, Switzerland)
  • A. Gennari (Novara, Italy)
  • S. Peters (Lausanne, Switzerland)
  • E. Romano (Paris, CEDEX 5, France)
  • D. Pinato (London, United Kingdom)

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

Collapse
Poster Display (ID 17) Poster Display

238P - Exploring blood immune cell dynamics to unravel the immunomodulatory effect of radiotherapy in NSCLC patients undergoing immune checkpoint inhibitors (ID 478)

Presentation Number
238P
Lecture Time
12:30 - 12:30
Speakers
  • G. Mazzaschi (Parma, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • P. Tamarozzi (Parma, Italy)
  • B. Lorusso (Parma, Italy)
  • M. Verzè (Parma, Italy)
  • M. Pluchino (Parma, Italy)
  • F. Trentini (Parma, Italy)
  • B. Dalla Valle (Parma, Italy)
  • R. Minari (Parma, Italy)
  • F. Perrone (Parma, Italy)
  • P. Bordi (Parma, Italy)
  • A. Leonetti (Parma, Italy)
  • L. Moron Dalla Tor (Parma, Italy)
  • L. Leo (Parma, Italy)
  • G. Milanese (Parma, Italy)
  • M. Balbi (Parma, Italy)
  • S. Buti (Parma, Italy)
  • G. Roti (Parma, Italy)
  • F. Quaini (Parma, Italy)
  • N. Sverzellati (Parma, Italy)
  • M. Tiseo (Parma, Italy)

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 Ist 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.

Collapse
Poster Display (ID 17) Poster Display

239P - Survival outcomes predicted by irAEs on 18F-FDG-PET in response to PD-1 antibody therapy in metastatic melanoma (ID 361)

Presentation Number
239P
Lecture Time
12:30 - 12:30
Speakers
  • S. Lewis (Melbourne, AU, Australia)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • S. Lewis (Melbourne, AU, Australia)
  • M. Cherk (Melbourne, Australia)
  • D. Nadebaum (Melbourne, Australia)
  • A. Haydon (Melbourne, VIC, Australia)

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.

Collapse
Poster Display (ID 17) Poster Display

240P - AMU: Using mRNA Embedding in Transformer Network to Predict Melanoma Immune Checkpoint Inhibitor Response (ID 234)

Presentation Number
240P
Lecture Time
12:30 - 12:30
Speakers
  • Y. Yin (Fuzhou, China)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • Y. Yin (Fuzhou, China)
  • Y. Huang (Fuzhou, China)
  • X. Xie (Fuzhou, China)
  • M. Wang (Fuzhou, China)

Abstract

Background

Melanoma Immune checkpoint inhibitor (ICI) response is related to complex biological pathways. Deep learning (DL) model has powerful fitting ability and transformer structure has interactive information extraction function. We designed a transformer neural network fed with muti-gene mRNA expression quantities to predict melanoma ICI response and explored gene interactions from 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 potential associated with melanoma, inflammation, immunity, 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 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 part, TNF-TNFRSF1A pathway were 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 biomedical field.

Collapse
Poster Display (ID 17) Poster Display

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 326)

Presentation Number
241P
Lecture Time
12:30 - 12:30
Speakers
  • M. Swaminathan (Liverpool, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • M. Swaminathan (Liverpool, United Kingdom)
  • A. Angelakas (Manchester, United Kingdom)
  • M. Baxter (Dundee, United Kingdom)
  • J. Cotton (Metropolitan Borough of Wirral, United Kingdom)
  • C. Dobeson (Newcastle-upon-Tyne, United Kingdom)
  • L. Feeney (Belfast, United Kingdom)
  • A. Gault (Newcastle-upon-Tyne, United Kingdom)
  • D. Hughes (London, United Kingdom)
  • C. Jones (Leeds, United Kingdom)
  • R. Lee (Manchester, United Kingdom)
  • S. Mughal (Wakefield, United Kingdom)
  • S. Parikh (Shrewsbury, United Kingdom)
  • M. Pritchard (Liverpool, United Kingdom)
  • L. Rodgers (Glasgow, United Kingdom)
  • M. Rowe (Plymouth, United Kingdom)
  • A. Salawu (Toronto, South Yorkshire, Canada)
  • R. Shotton (Manchester, United Kingdom)
  • N. Tinsley (Manchester, United Kingdom)
  • A. Tivey (Manchester, United Kingdom)
  • A. Olsson-Brown (Liverpool, United Kingdom)

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.

Collapse
Poster Display (ID 17) Poster Display

242P - Educational needs of healthcare professionals using immune checkpoint inhibitors across multiple tumour types: A cross-speciality survey (ID 327)

Presentation Number
242P
Lecture Time
12:30 - 12:30
Speakers
  • J. Brownsword (Stockport, United Kingdom)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • J. Brownsword (Stockport, United Kingdom)
  • A. Scott (Goring-on-Thames, United Kingdom)
  • A. Noble (Stockport, United Kingdom)
  • T. Powles (London, United Kingdom)
  • K. Harrington (London, United Kingdom)

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.

Collapse
Poster Display (ID 17) Poster Display

243P - Skin biopsies for pharmacodynamic studies of tebentafusp, a novel bispecific molecule against the melanocytic antigen gp100. (ID 597)

Presentation Number
243P
Lecture Time
12:30 - 12:30
Speakers
  • R. Stäger (Zurich, Switzerland)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • R. Stäger (Zurich, Switzerland)
  • A. Tastanova (Zurich, Switzerland)
  • E. Ramelyte (Zurich, Switzerland)
  • M. Levesque (Zurich, Switzerland)
  • R. Dummer (Zurich, Switzerland)
  • B. Meier-Schiesser (Zurich, Switzerland)

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.

Collapse
Poster Display (ID 17) Poster Display

244P - DNA-PK inhibition sustains immune activation in SCLC (ID 740)

Presentation Number
244P
Lecture Time
12:30 - 12:30
Speakers
  • C. Della Corte (Napoli, Italy)
Session Name
Poster Display (ID 17)
Room
Foyer ABC
Date
Thu, 08.12.2022
Time
12:30 - 13:15
Authors
  • C. Della Corte (Napoli, Italy)
  • V. Ciaramella (Napoli, Italy)
  • V. Nardone (Napoli, Italy)
  • G. Di Guida (Caserta, Italy)
  • F. Ciardiello (Napoli, Italy)
  • F. Morgillo (Napoli, Italy)

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

Clinical trial identification

NA

Collapse