Insufficiency of compound immune checkpoint blockade to overcome engineered T cell exhaustion in pancreatic cancer

Background Achieving robust responses with adoptive cell therapy for the treatment of the highly lethal pancreatic ductal adenocarcinoma (PDA) has been elusive. We previously showed that T cells engineered to express a mesothelin-specific T cell receptor (TCRMsln) accumulate in autochthonous PDA, mediate therapeutic antitumor activity, but fail to eradicate tumors in part due to acquisition of a dysfunctional exhausted T cell state. Methods Here, we investigated the role of immune checkpoints in mediating TCR engineered T cell dysfunction in a genetically engineered PDA mouse model. The fate of engineered T cells that were either deficient in PD-1, or transferred concurrent with antibodies blocking PD-L1 and/or additional immune checkpoints, were tracked to evaluate persistence, functionality, and antitumor activity at day 8 and day 28 post infusion. We performed RNAseq on engineered T cells isolated from tumors and compared differentially expressed genes to prototypical endogenous exhausted T cells. Results PD-L1 pathway blockade and/or simultaneous blockade of multiple coinhibitory receptors during adoptive cell therapy was insufficient to prevent engineered T cell dysfunction in autochthonous PDA yet resulted in subclinical activity in the lung, without enhancing anti-tumor immunity. Gene expression analysis revealed that ex vivo TCR engineered T cells markedly differed from in vivo primed endogenous effector T cells which can respond to immune checkpoint inhibitors. Early after transfer, intratumoral TCR engineered T cells acquired a similar molecular program to prototypical exhausted T cells that arise during chronic viral infection, but the molecular programs later diverged. Intratumoral engineered T cells exhibited decreased effector and cell cycle genes and were refractory to TCR signaling. Conclusions Abrogation of PD-1 signaling is not sufficient to overcome TCR engineered T cell dysfunction in PDA. Our study suggests that contributions by both the differentiation pathways induced during the ex vivo T cell engineering process and intratumoral suppressive mechanisms render engineered T cells dysfunctional and resistant to rescue by blockade of immune checkpoints.

[1]  Li‐Wha Wu,et al.  IL-20 antagonist suppresses PD-L1 expression and prolongs survival in pancreatic cancer models , 2020, Nature Communications.

[2]  A. Kamphorst,et al.  An intra-tumoral niche maintains and differentiates stem-like CD8 T cells , 2019, Nature.

[3]  S. Berger,et al.  TCF-1-Centered Transcriptional Network Drives an Effector versus Exhausted CD8 T Cell-Fate Decision. , 2019, Immunity.

[4]  Myung Ah Lee,et al.  Durvalumab With or Without Tremelimumab for Patients With Metastatic Pancreatic Ductal Adenocarcinoma: A Phase 2 Randomized Clinical Trial. , 2019, JAMA oncology.

[5]  Xianjun Yu,et al.  A PD-L2-based immune marker signature helps to predict survival in resected pancreatic ductal adenocarcinoma , 2019, Journal of Immunotherapy for Cancer.

[6]  Adam L. Burrack,et al.  Combination PD-1 and PD-L1 Blockade Promotes Durable Neoantigen-Specific T Cell-Mediated Immunity in Pancreatic Ductal Adenocarcinoma , 2019, Cell reports.

[7]  R. Gottardo,et al.  Engineered Adoptive T-cell Therapy Prolongs Survival in a Preclinical Model of Advanced-Stage Ovarian Cancer , 2019, Cancer Immunology Research.

[8]  M. Delorenzi,et al.  TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection , 2019, Nature.

[9]  Yong Liu,et al.  TOX is a critical regulator of tumour-specific T cell differentiation , 2019, Nature.

[10]  S. Berger,et al.  TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion , 2019, Nature.

[11]  Howard Y. Chang,et al.  Clonal replacement of tumor-specific T cells following PD-1 blockade , 2019, Nature Medicine.

[12]  E. Wherry,et al.  CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. , 2019, Annual review of immunology.

[13]  R. Pierce,et al.  Differential Effects of Depleting versus Programming Tumor-Associated Macrophages on Engineered T Cells in Pancreatic Ductal Adenocarcinoma , 2019, Cancer Immunology Research.

[14]  M. Sadelain,et al.  CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape , 2019, Nature.

[15]  Joonsoo Kang Faculty Opinions recommendation of Intratumoral Tcf1+PD-1+CD8+ T Cells with Stem-like Properties Promote Tumor Control in Response to Vaccination and Checkpoint Blockade Immunotherapy. , 2019, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.

[16]  F. Hodi,et al.  Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade , 2019, Nature Immunology.

[17]  Jianjun Hu,et al.  The Transcription Factor TCF1 Preserves the Effector Function of Exhausted CD8 T Cells During Chronic Viral Infection , 2019, Front. Immunol..

[18]  Daniel E. Speiser,et al.  Intratumoral Tcf1+PD‐1+CD8+ T Cells with Stem‐like Properties Promote Tumor Control in Response to Vaccination and Checkpoint Blockade Immunotherapy , 2019, Immunity.

[19]  J. Wolchok,et al.  Acquired resistance to immunotherapy in MMR-D pancreatic cancer , 2018, Journal of Immunotherapy for Cancer.

[20]  Monika S. Kowalczyk,et al.  Induction and transcriptional regulation of the co-inhibitory gene module in T cells , 2018, Nature.

[21]  G. D. Di Liberto,et al.  Expression of the DNA‐Binding Factor TOX Promotes the Encephalitogenic Potential of Microbe‐Induced Autoreactive CD8+ T Cells , 2018, Immunity.

[22]  A. Rogel,et al.  PD-1 Blockade and CD27 Stimulation Activate Distinct Transcriptional Programs That Synergize for CD8+ T-Cell–Driven Antitumor Immunity , 2018, Clinical Cancer Research.

[23]  R. Pierce,et al.  T-cell Localization, Activation, and Clonal Expansion in Human Pancreatic Ductal Adenocarcinoma , 2017, Cancer Immunology Research.

[24]  W. Shen,et al.  TGF-β in pancreatic cancer initiation and progression: two sides of the same coin , 2017, Cell & Bioscience.

[25]  I. Pastan,et al.  Comprehensive immunohistochemical study of mesothelin (MSLN) using different monoclonal antibodies 5B2 and MN-1 in 1562 tumors with evaluation of its prognostic value in malignant pleural mesothelioma , 2017, Oncotarget.

[26]  A. Elkahloun,et al.  The TCF1-Bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness , 2016, Science Immunology.

[27]  I. Pastan,et al.  Mesothelin Immunotherapy for Cancer: Ready for Prime Time? , 2016, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[28]  Jeffrey J Delrow,et al.  Tumor-Specific T Cell Dysfunction Is a Dynamic Antigen-Driven Differentiation Program Initiated Early during Tumorigenesis. , 2016, Immunity.

[29]  Sandra P. Calderon-Copete,et al.  T Cell Factor 1-Expressing Memory-like CD8(+) T Cells Sustain the Immune Response to Chronic Viral Infections. , 2016, Immunity.

[30]  Matheus C. Bürger,et al.  Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy , 2016, Nature.

[31]  Shohei Koyama,et al.  Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints , 2016, Nature Communications.

[32]  Thomas M. Schmitt,et al.  T Cells Engineered against a Native Antigen Can Surmount Immunologic and Physical Barriers to Treat Pancreatic Ductal Adenocarcinoma. , 2015, Cancer cell.

[33]  E. Wherry,et al.  Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells , 2015, The Journal of experimental medicine.

[34]  P. Greenberg,et al.  Stromal reengineering to treat pancreas cancer. , 2014, Carcinogenesis.

[35]  Benjamin D. Smith,et al.  Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. , 2014, Cancer research.

[36]  P. Greenberg,et al.  Targeted depletion of an MDSC subset unmasks pancreatic ductal adenocarcinoma to adaptive immunity , 2014, Gut.

[37]  Burton E. Barnett,et al.  Progenitor and Terminal Subsets of CD8+ T Cells Cooperate to Contain Chronic Viral Infection , 2012, Science.

[38]  C. Drake,et al.  Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. , 2012, The New England journal of medicine.

[39]  Nicole R. Cunningham,et al.  T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse , 2011, The Journal of experimental medicine.

[40]  Rafael A. Irizarry,et al.  A framework for oligonucleotide microarray preprocessing , 2010, Bioinform..

[41]  S. Rosenberg,et al.  Phase 2 Trial of Single Agent Ipilimumab (Anti-CTLA-4) for Locally Advanced or Metastatic Pancreatic Adenocarcinoma , 2010, Journal of immunotherapy.

[42]  M. Bevan,et al.  Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. , 2010, Immunity.

[43]  Kendall A. Smith,et al.  Prolonged interleukin-2Ralpha expression on virus-specific CD8+ T cells favors terminal-effector differentiation in vivo. , 2010, Immunity.

[44]  P. Muranski,et al.  Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells , 2009, Nature Medicine.

[45]  W. Leonard,et al.  IL-2 and IL-21 confer opposing differentiation programs to CD8+ T cells for adoptive immunotherapy. , 2008, Blood.

[46]  E. Wherry,et al.  Molecular Signature of CD8+ T Cell Exhaustion during Chronic Viral Infection (DOI:10.1016/j.immuni.2007.09.006) , 2007 .

[47]  D. Tuveson,et al.  Dynamics of the immune reaction to pancreatic cancer from inception to invasion. , 2007, Cancer research.

[48]  J. Davis Bioinformatics and Computational Biology Solutions Using R and Bioconductor , 2007 .

[49]  M. Fraga,et al.  The Polycomb group protein EZH2 directly controls DNA methylation , 2006, Nature.

[50]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[51]  S. Rosenberg,et al.  Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. , 2005, The Journal of clinical investigation.

[52]  C. Drachenberg,et al.  TGF-β–dependent CD103 expression by CD8+ T cells promotes selective destruction of the host intestinal epithelium during graft-versus-host disease , 2005, The Journal of experimental medicine.

[53]  Yoav Benjamini,et al.  Identifying differentially expressed genes using false discovery rate controlling procedures , 2003, Bioinform..

[54]  A I Saeed,et al.  TM4: a free, open-source system for microarray data management and analysis. , 2003, BioTechniques.

[55]  J. Cameron,et al.  Mesothelin is overexpressed in the vast majority of ductal adenocarcinomas of the pancreas: identification of a new pancreatic cancer marker by serial analysis of gene expression (SAGE). , 2001, Clinical cancer research : an official journal of the American Association for Cancer Research.

[56]  Ka Yee Yeung,et al.  Validating clustering for gene expression data , 2001, Bioinform..

[57]  I. Pastan,et al.  Mesothelin Is Not Required for Normal Mouse Development or Reproduction , 2000, Molecular and Cellular Biology.

[58]  J. Aster,et al.  Notch1 expression in early lymphopoiesis influences B versus T lineage determination. , 1999, Immunity.

[59]  H. Pircher,et al.  Tolerance induction in double specific T-cell receptor transgenic mice varies with antigen , 1989, Nature.

[60]  N. Adel Current treatment landscape and emerging therapies for pancreatic cancer. , 2019, The American journal of managed care.

[61]  U. Hegde,et al.  Transcriptional Profiles of Mart-1(27-35) Epitope Specific TCReng Human CD8+ and CD4+ T Cells upon Epitope Encounter as Elucidated by RNASeq , 2014 .

[62]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .