scholarly journals Reversing T-cell Exhaustion in Cancer: Lessons Learned from PD-1/PD-L1 Immune Checkpoint Blockade

Author(s):  
Natalija Budimir ◽  
Graham D. Thomas ◽  
Joseph S. Dolina ◽  
Shahram Salek-Ardakani
2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e15023-e15023
Author(s):  
Brian P Riesenberg ◽  
Mingjia Li ◽  
Daniel Spakowicz ◽  
Rebecca Hoyd ◽  
Joal Beane ◽  
...  

e15023 Background: A growing body of evidence has linked platelets (plts) with immune suppression in the tumor microenvironment (TME). Our group has demonstrated, plts are the dominant source for TGF-β in TME and pharmacologic inhibition of plt function enhances multiple forms of immunotherapy (Sc Immunol 2017 PMID 28763790; J Immunol 2019 PMID 31358658; Sci Transl Med 2020 PMID 31915300). To further delineate this relationship, we examined the roles of plts on T cell exhaustion in a preclinical model and on response to immune checkpoint inhibitors (ICI) in a large cohort of patients (pts) with stage 4 cancers. Methods: The mouse MC-38 colon adenocarcinoma model was used in age-matched female C57Bl/6J mice. Antiplatelet therapy (APT) consisting of aspirin and clopidogrel (both at 30 mg/kg daily) was delivered p.o. and anti-PD-1 antibody (100 mg/mouse every 3 days) was administered i.p. starting on days 5 and 9 respectively. TME analysis via multispectral histology or flow cytometry was performed. Retrospective analysis was carried out on 826 pts who received ICI from 2011-2017 at the Ohio State University. Baseline plt count was collected within 7 days before initiating ICI. Repeat plt count was obtained prior to initiation of cycle 2. Normal plt counts were defined as 150,000 – 450,000/µl blood, and thrombocytosis as plt ≥450,000/µl blood. Kaplan Meier and log-rank analysis were performed to estimate median survival and determine the association with plt count. Results: In pre-clinical models, pretreatment with APT (1) synergized with PD-1 blockade to enhance T cell infiltration into MC-38 tumors resulting in immediate tumor reduction, and (2) decreased tumor infiltrating CD8+ T cell TOX expression, a transcription factor associated with T cell exhaustion (Nat Immunol 2019 PMID: 31427776). Among pts receiving ICIs, 46 (5.6%) pts with thrombocytosis had a significantly reduced median OS vs pts with normal plt counts: 6.0 (95 CI: 1.5—10.6) months (mos) vs 11.6 (95 CI: 9.7—13.4) mos (p = 0.002). Fluctuations ≥50,000 plt/mL in either direction between cycle 1 and 2 were associated with a significant reduction in median OS: 8.3 (95 CI: 6.4—10.1) mos vs 13.6 (95 CI: 11.2-16.1) mos (p < 0.001). Conclusions: There is a strong association between plts and failure of ICI in both the preclinical and clinical settings, likely via modifying the amount of active CD8+ T cells infiltrating into tumors. These findings merit further study to delineate the underlying mechanism for plt-mediated immune suppression and strategies to overcome it.


Cancers ◽  
2021 ◽  
Vol 13 (3) ◽  
pp. 515
Author(s):  
Sungmin Jung ◽  
Jea-Hyun Baek

T cell factor 1 (TCF1) is a transcription factor that has been highlighted to play a critical role in the promotion of T cell proliferation and maintenance of cell stemness in the embryonic and CD8+ T cell populations. The regulatory nature of TCF1 in CD8+ T cells is of great significance, especially within the context of T cell exhaustion, which is linked to the tumor and viral escape in pathological contexts. Indeed, inhibitory signals, such as programmed cell death 1 (PD-1) and cytotoxic-T-lymphocyte-associated protein 4 (CTLA-4), expressed on exhausted T lymphocytes (TEX), have become major therapeutic targets in immune checkpoint blockade (ICB) therapy. The significance of TCF1 in the sustenance of CTL-mediated immunity against pathogens and tumors, as well as its recently observed necessity for an effective anti-tumor immune response in ICB therapy, presents TCF1 as a potentially significant biomarker and/or therapeutic target for overcoming CD8+ T cell exhaustion and resistance to ICB therapy. In this review, we aim to outline the recent findings on the role of TCF1 in T cell development and discuss its implications in anti-tumor immunity.


2021 ◽  
Vol 9 (1) ◽  
pp. e001460 ◽  
Author(s):  
Xiuting Liu ◽  
Graham D Hogg ◽  
David G DeNardo

The clinical success of immune checkpoint inhibitors has highlighted the central role of the immune system in cancer control. Immune checkpoint inhibitors can reinvigorate anti-cancer immunity and are now the standard of care in a number of malignancies. However, research on immune checkpoint blockade has largely been framed with the central dogma that checkpoint therapies intrinsically target the T cell, triggering the tumoricidal potential of the adaptive immune system. Although T cells undoubtedly remain a critical piece of the story, mounting evidence, reviewed herein, indicates that much of the efficacy of checkpoint therapies may be attributable to the innate immune system. Emerging research suggests that T cell-directed checkpoint antibodies such as anti-programmed cell death protein-1 (PD-1) or programmed death-ligand-1 (PD-L1) can impact innate immunity by both direct and indirect pathways, which may ultimately shape clinical efficacy. However, the mechanisms and impacts of these activities have yet to be fully elucidated, and checkpoint therapies have potentially beneficial and detrimental effects on innate antitumor immunity. Further research into the role of innate subsets during checkpoint blockade may be critical for developing combination therapies to help overcome checkpoint resistance. The potential of checkpoint therapies to amplify innate antitumor immunity represents a promising new field that can be translated into innovative immunotherapies for patients fighting refractory malignancies.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A728-A728
Author(s):  
Shengqing Gu ◽  
Wubing Zhang ◽  
Xiaoqing Wang ◽  
Peng Jiang ◽  
Nicole Traugh ◽  
...  

BackgroundCancer immunotherapy, especially immune checkpoint blockade (ICB) therapy, is leading to a paradigm shift in cancer treatment, as a small percentage of cancer patients have obtained durable remission following ICB treatment. Successful ICB responses rely on cancer cells presenting antigens to the cell surface via the major histocompatibility complex (MHC), which activates antigen-specific T-lymphocytes to kill cancer cells. Type-I MHC (MHC-I) is wildly expressed in all cell types and mediates the interaction with cytotoxic CD8 T cells. However, over 65% of cancer patients are estimated to show defects in MHC-I-mediated antigen presentation, including downregulation of its expression that can lead to primary or acquired resistance to ICB therapy, and therapeutic strategies to effectively restore or boost MHC-I are limited.MethodsHere, we employed a CRISPR screening approach with dual-marker FACS sorting to identify factors that decouple the regulation of MHC-I and PD-L1. The experimentally validated target was used to generate a KO differential expression signature. Using this signature, we analyzed transcriptome data from drug perturbation studies to identify drugs that regulate MHC-I but not PD-L1. Finally, we validated the effect of the identified drug to enhance ICB response in a T-cell-dependent manner in vivo.ResultsCRISPR screens identified TRAF3, a suppressor of the NF-κB pathway, as a negative regulator of MHC-I but not PD-L1. The Traf3-knockout (Traf3-KO) gene expression signature is associated with better survival in ICB-naive cancer patients and better ICB response. We then screened for drugs with similar transcriptional effects as this signature and identified SMAC mimetics. We experimentally validated that the SMAC mimetic birinapant upregulates MHC-I, sensitizes cancer cells to T-cell-dependent killing, and adds to ICB efficacy. However, in cancer cells with high NF-κB activity, the effect of birinapant on MHC-I is weak, indicating context-dependent MHC-I regulation.ConclusionsIn summary, Traf3 deletion specifically upregulates MHC-I without inducing PD-L1 in response to various cytokines and sensitizes cancer cells to T-cell-driven cytotoxicity. The SMAC mimetic birinapant phenocopies Traf3-knockout and sensitizes MHC-I-low melanoma to ICB therapy. Further studies are needed to elucidate the context-dependencies of MHC-I regulation. Our findings provide preclinical rationale for treating some tumors expressing low MHC-I with SMAC mimetics to enhance sensitivity to immunotherapy. The approach used in this study can be generalized to identify other drugs that enhance immunotherapy efficacy.AcknowledgementsThis study was supported by grants from the NIH (R01CA234018 to XSL, R01AI137337 to BEG, P50CA101942-12 and P50CA206963 to GJF), Breast Cancer Research Foundation (BCRF-19-100 to XSL), Burroughs Wellcome Career Award in Medical Sciences (to BEG), and Sara Elizabeth O'Brien Trust Fellowship (to SG).We thank Drs. Kai Wucherpfennig and Deng Pan for their insightful suggestions on this study.Ethics ApprovalAll mice were housed in standard cage in Dana-Farber Cancer Institute Animal Resources Facility (ARF). All animal procedures were carried out under the ARF Institutional Animal Care and Use Committee (IACUC) protocol and were in accordance with the IACUC standards for the welfare of animals.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi95-vi95
Author(s):  
Jessica Waibl Polania ◽  
William Tomaszweski ◽  
Alexandra Hoyt-Miggelbrink ◽  
Karolina Woroniecka ◽  
Peter Fecci

Abstract BACKGROUND Glioblastoma (GBM) is the most common primary brain cancer in adults and remains universally lethal. Median survival remains a bleak 15-17 months from time of diagnosis, and current immunotherapeutic efficacy continues to be hindered by the robust immunosuppression present in the GBM microenvironment. T cells, critical for tumor clearance, are particularly affected, and many take on a functionally exhausted phenotype within the tumor. Importantly, two exhaustion states, progenitor and terminal, have been identified in models of chronic infection and cancer. This distinction is particularly relevant, as progenitor exhausted T cells can respond favorably to immune checkpoint blockade, while terminally exhausted T cells are resistant. To date, the dynamics and characteristics of these exhausted populations in GBM remain unclear. RESULTS In an orthotopic murine model of GBM, progenitor and terminal exhausted CD8 T cells were identified by flow cytometry as PD1+SLAMF6+ and PD1+TIM3+, respectively. Using a time-course approach, we detected progenitor exhaustion by day 8 in the tumor, but not in draining lymph nodes. Additionally, we show that the frequency of progenitor exhaustion is highest during early tumor progression, while terminal exhaustion is the most abundant in more advanced tumors ( &gt;14 days). Functional differences between subsets were evaluated via intracellular staining of IFNγ, TNFα, granzyme B, and Ki67. Terminally exhausted T cells displayed higher cytotoxic molecule expression than progenitor exhausted T cells, similar to what has been documented in melanoma models. CONCLUSIONS Our findings identify T cell exhaustion subsets within GBM that require further investigation and may be relevant to overcome current barriers to immunotherapeutic efficacy.


Sign in / Sign up

Export Citation Format

Share Document