Immune profiling and clinical outcomes in patients treated with ramucirumab and pembrolizumab in phase I study JVDF.

2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 3089-3089 ◽  
Author(s):  
Roy S. Herbst ◽  
Hendrik-Tobias Arkenau ◽  
Emiliano Calvo ◽  
Johanna C. Bendell ◽  
Nicolas Penel ◽  
...  

3089 Background: In Study JVDF (NCT02443324), we combined ramucirumab (VEGFR2 antagonist) and pembrolizumab (PD-1 antagonist) to simultaneously target the tumor microenvironment and immune checkpoints in patients with advanced non-small cell lung cancer (NSCLC), gastric or gastroesophageal junction adenocarcinoma (G/GEJ), urothelial carcinoma (UC) or biliary tract cancer (BTC). We reported that this combination was associated with increased antitumor activity in patients with PD-L1 positive tumors by immunohistochemistry (IHC) compared with PD-L1 negative tumors.1) Here we explore the association between baseline gene expression profiles and clinical outcomes. Methods: JVDF was a nonrandomized phase 1a/b trial that treated patients with intravenous ramucirumab at 8 mg/kg on days 1 and 8 (G/GEJ, BTC) or 10 mg/kg on day 1 (G/GEJ, NSCLC, UC) plus pembrolizumab (200 mg day 1) every 3 weeks.1 Baseline tumor samples from 53 patients across 7 study cohorts were analyzed with the NanoString PanCancer Immune Profiling Panel for RNA expression and the DAKO PD-L1 IHC 22C3 pharmDx assay for PD-L1 protein expression. Clinical outcomes included progression-free survival (PFS), overall survival (OS), and objective response rate (ORR). Results: Across cohorts, PD-L1 gene expression was correlated with increased IFNγ gene expression and immune-related gene signatures (T effector, T cell-inflamed (TIS)), and trended with PD-L1 protein expression. Expression of immune checkpoint-related genes and myeloid-derived suppressor cell /regulatory T cell markers was increased in the NSCLC TPS≥50% PD-L1 IHC subgroup (N=8), while no clear pattern of expression was observed in other cohorts. Higher T effector and TIS scores appeared associated with better survival and response in NSCLC cohorts (mean TIS: 1.21±0.80 in responders (N=7) vs -0.13±0.57 in non-responders (N=7); p=0.004), and a trend was observed in G/GEJ cohorts (mean TIS: -0.18±0.30 (N=5) vs -0.39±0.21 (N=13); p=0.207). Of note, partial responses and stable disease were also observed in NSCLC and G/GEJ patients with a low baseline inflammatory signature score. Additional analyses are ongoing and will be presented. Conclusions: Baseline PD-L1 gene and protein expression tends to correlate with immune-related gene expression, and an inflamed tumor microenvironment may be associated with better clinical outcomes with ramucirumab and pembrolizumab. However, interpretation is limited by lack of control arm and sample size.1) Herbst et al. Lancet Oncol. 2019 Aug; 20 (8):1109-1123. Clinical trial information: NCT02443324 .

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A91-A91
Author(s):  
Jennifer Chew ◽  
Cedric Uytingco ◽  
Rapolas Spalinskas ◽  
Yifeng Yin ◽  
Joe Shuga ◽  
...  

BackgroundThe tumor microenvironment (TME) is composed of highly heterogeneous extracellular structures and cell types such as endothelial cells, immune cells, and fibroblasts that dynamically influence and communicate with each other. The constant interaction between a tumor and its microenvironment plays a critical role in cancer development and progression and can significantly affect a tumor’s response to therapy and capacity for multi-drug resistance. High resolution analyses of gene and protein expression with spatial context can provide deeper insights into the interactions between tumor cells and surrounding cells within the TME, where a better understanding of the underlying biology can improve treatment efficacy and patient outcomes. Here, we demonstrated the ability to perform streamlined multi-omic tumor analyses by utilizing the 10X Genomics Visium Spatial Gene Expression Solution for FFPE with multiplex protein enablement. This technique simultaneously assesses gene and protein expression to elucidate the immunological profile and microenvironment of different breast cancer samples in conjunction with standard pathological methods.MethodsSerial (5 µm) sections of FFPE human breast cancer samples were placed on Visium Gene Expression (GEX) slides. The Visium GEX slides incorporate ~5,000 molecularly barcoded, spatially encoded capture spots onto which tissue sections are placed, stained, and imaged. Following incubation with a human whole transcriptome, probe-based RNA panel and an immuno-oncology oligo-tagged antibody panel, developed with Abcam conjugated antibodies, the tissues are permeabilized and the representative probes are captured. Paired GEX and protein libraries are generated for each section and then sequenced on an Illumina NovaSeq at a depth of ~50,000 reads per spot. Resulting reads from both libraries are aligned and overlaid with H&E-stained tissue images, enabling analysis of both mRNA and protein expression. Additional analyses and data visualizations were performed on the Loupe Browser v4.1 desktop software.ConclusionsSpatial transcriptomics technology complements pathological examination by combining histological assessment with the throughput and deep biological insight of highly-multiplexed protein detection and RNA-seq. Taken together, our work demonstrated that Visium Spatial technology provides a spatially-resolved, multi-analyte view of the tumor microenvironment, where a greater understanding of cellular behavior in and around tumors can help drive discovery of new biomarkers and therapeutic targets.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A612-A612
Author(s):  
Yoshinori Narita ◽  
Mika Kamata-Sakurai

BackgroundAgonistic antibodies targeting CD137 in clinic have failed due to severe hepatotoxicity, leading to the development of bispecific approaches that must rely on high tumor-associated antigen expression to crosslink CD137. Here we report on STA551, a novel anti-CD137 agonist antibody which binds to CD137 only in the presence of ATP. Extracellular ATP concentration is well-known to be elevated in tumor tissue while remaining tightly regulated in non-tumor tissue, suggesting that STA551 can activate immune cells only in tumor tissue and not elsewhere. Thus, STA551 has great potential to overcome the limitations of conventional CD137-targeted antibodies.MethodsWe evaluated in vitro STA551’s effect on IFN-γ production from human CD8+ T cells. We also evaluated in vivo STA551’s effect on tumor growth, RNAseq-based immune-related gene expression, immunohistochemistry, and T cell activation in tumor and non-tumor tissues in human CD137 knock-in mice treated with mouse surrogate STA551 (Sta-MB) or urelumab (Ure-MB) in combination with anti-PD-L1 antibody.ResultsIn a human T cell assay, STA551 induced IFN-γ only in the presence of ATP. In contrast, urelumab induced IFN-γ regardless of ATP concentration. In mice with Colon 38 tumors, Sta-MB inhibited tumor growth at least as strongly as Ure-MB, but whereas Ure-MB elicited systemic immune responses in draining lymph node, spleen, and liver, Sta-MB appeared to evade such responses. To confirm immune responses in tumors, we evaluated immune-related gene expression and found changes after treatment with Sta-MB or Ure-MB. These results suggest that STA551 works only in tumor tissue. Furthermore, Sta-MB with anti-PD-L1 antibody synergistically inhibited tumor growth and dramatically changed immune-related gene expression, CD8+ T cell infiltration, and PD-L1 expression without systemic immune responses. Also, it was well-tolerated in cynomolgus monkey in a repeated-dose toxicity study*.ConclusionsSTA551 is a novel anti-CD137 agonist antibody that exerts agonistic activity selectively in tumors without on-target toxicity in non-tumor tissues, regardless of tumor-associated antigen expression. These results strongly support the clinical testing of STA551 for the treatment of solid tumors. STA551 is currently being tested in a phase 1 clinical study.Ethics ApprovalAll animal studies were reviewed and approved by the Institutional Animal Care and Use Committee (IACUC).


2020 ◽  
Author(s):  
Yi-Jun Kim ◽  
Kwangsoo Kim ◽  
Soo Yeon Seo ◽  
Juyeon Yu ◽  
Il Han Kim ◽  
...  

Abstract The time-sequential change in immune-related gene expression of the glioblastoma cell line after irradiation was evaluated to speculate the effect of combined immunotherapy with radiotherapy. The U373 MG glioblastoma cell line was irradiated with 6 Gy single dose. Next-generation sequencing (NGS) transcriptome data was generated before irradiation (control), and at 6, 24, and 48 hours post-irradiation. Immune-related pathways were analyzed at each time period. The same analyses were also performed for A549 lung cancer and U87 MG glioblastoma cell lines. Western blotting confirmed the programmed death-ligand 1 (PD-L1) expression levels over time. In the U373 MG cell line, neutrophil-mediated immunity, type I interferon signaling, antigen cross-presentation to T cell, and interferon-γ signals began to increase significantly at 24 hours and were upregulated until 48 hours after irradiation. The results were similar to those of the A549 and U87 MG cell lines. Without T cell infiltration, PD-L1 did not increase even with upregulated interferon-γ signaling in cancer cells. In conclusions, In the glioblastoma cell line, immune-related signals were significantly upregulated at 24 hours after irradiation. Therefore, the time interval between daily radiotherapy might not be enough to expect full immune responses by combined immune checkpoint inhibitors and newly infiltrating immune cells after irradiation.


2020 ◽  
Vol 59 (4) ◽  
pp. 669-676 ◽  
Author(s):  
Pedro Negri ◽  
Leonor Ramirez ◽  
Silvina Quintana ◽  
Nicolas Szawarski ◽  
Matías D. Maggi ◽  
...  

2021 ◽  
Vol 23 (1) ◽  
Author(s):  
Matthew Mannarino ◽  
Hosni Cherif ◽  
Li Li ◽  
Kai Sheng ◽  
Oded Rabau ◽  
...  

Abstract Background There is an increased level of senescent cells and toll-like teceptor-1, -2, -4, and -6 (TLR) expression in degenerating intervertebral discs (IVDs) from back pain patients. However, it is currently not known if the increase in expression of TLRs is related to the senescent cells or if it is a more general increase on all cells. It is also not known if TLR activation in IVD cells will induce cell senescence. Methods Cells from non-degenerate human IVD were obtained from spine donors and cells from degenerate IVDs came from patients undergoing surgery for low back pain. Gene expression of TLR-1,2,4,6, senescence and senescence-associated secretory phenotype (SASP) markers was evaluated by RT-qPCR in isolated cells. Matrix synthesis was verified with safranin-O staining and Dimethyl-Methylene Blue Assay (DMMB) confirmed proteoglycan content. Protein expression of p16INK4a, SASP factors, and TLR-2 was evaluated by immunocytochemistry (ICC) and/or by enzyme-linked immunosorbent assay (ELISA). Results An increase in senescent cells was found following 48-h induction with a TLR-2/6 agonist in cells from both non-degenerate and degenerating human IVDs. Higher levels of SASP factors, TLR-2 gene expression, and protein expression were found following 48-h induction with TLR-2/6 agonist. Treatment with o-vanillin reduced the number of senescent cells, and increased matrix synthesis in IVD cells from back pain patients. Treatment with o-vanillin after induction with TLR-2/6 agonist reduced gene and protein expression of SASP factors and TLR-2. Co-localized staining of p16INK4a and TLR-2 demonstrated that senescent cells have a high TLR-2 expression. Conclusions Taken together our data demonstrate that activation of TLR-2/6 induce senescence and increase TLR-2 and SASP expression in cells from non-degenerate IVDs of organ donors without degeneration and back pain and in cells from degenerating human IVD of patients with disc degeneration and back pain. The senescent cells showed high TLR-2 expression suggesting a link between TLR activation and cell senescence in human IVD cells. The reduction in senescence, SASP, and TLR-2 expression suggest o-vanillin as a potential disease-modifying drug for patients with disc degeneration and back pain.


Sign in / Sign up

Export Citation Format

Share Document