Dynamic chromatin regulatory landscape of human CAR T cell exhaustion

Significance T cell exhaustion is a major barrier to cancer immunotherapy. T cell exhaustion is the state of T cell dysfunction after chronic stimulation, and recent studies indicate that exhaustion is epigenetically controlled and associated with unique chromatin profiles. This work reports the genome-wide map of active DNA regulatory elements and their connection to genes in the time course of human chimeric antigen receptor T cell exhaustion. Early chromatin events establish the later program of gene expression, active elements are often human-specific, and exhaustion-associated enhancers can be manipulated to precisely control the level of inhibitory receptors on T cells. Dysfunction in T cells limits the efficacy of cancer immunotherapy. We profiled the epigenome, transcriptome, and enhancer connectome of exhaustion-prone GD2-targeting HA-28z chimeric antigen receptor (CAR) T cells and control CD19-targeting CAR T cells, which present less exhaustion-inducing tonic signaling, at multiple points during their ex vivo expansion. We found widespread, dynamic changes in chromatin accessibility and three-dimensional (3D) chromosome conformation preceding changes in gene expression, notably at loci proximal to exhaustion-associated genes such as PDCD1, CTLA4, and HAVCR2, and increased DNA motif access for AP-1 family transcription factors, which are known to promote exhaustion. Although T cell exhaustion has been studied in detail in mice, we find that the regulatory networks of T cell exhaustion differ between species and involve distinct loci of accessible chromatin and cis-regulated target genes in human CAR T cell exhaustion. Deletion of exhaustion-specific candidate enhancers of PDCD1 suppress the expression of PD-1 in an in vitro model of T cell dysfunction and in HA-28z CAR T cells, suggesting enhancer editing as a path forward in improving cancer immunotherapy.

[1]  C. Brander,et al.  TOX is expressed by exhausted and polyfunctional human effector memory CD8+ T cells , 2020, Science Immunology.

[2]  K. Verhamme,et al.  Immune Checkpoint Inhibitor-Related Cytokine Release Syndrome: Analysis of WHO Global Pharmacovigilance Database , 2020, Frontiers in Pharmacology.

[3]  E. Wherry,et al.  Developmental Relationships of Four Exhausted CD8+ T Cell Subsets Reveals Underlying Transcriptional and Epigenetic Landscape Control Mechanisms. , 2020, Immunity.

[4]  W. Wong,et al.  Time course regulatory analysis based on paired expression and chromatin accessibility data , 2020, Genome research.

[5]  G. Vahedi,et al.  Joint profiling of chromatin accessibility and CAR-T integration site analysis at population and single-cell levels , 2020, Proceedings of the National Academy of Sciences.

[6]  Bin Zhang,et al.  T Cell Dysfunction and Exhaustion in Cancer , 2020, Frontiers in Cell and Developmental Biology.

[7]  S. Maclean,et al.  A High-Throughput Method for Characterizing Novel Chimeric Antigen Receptors in Jurkat Cells , 2020, Molecular therapy. Methods & clinical development.

[8]  Howard Y. Chang,et al.  Transient “rest” induces functional reinvigoration and epigenetic remodeling in exhausted CAR-T cells , 2020, bioRxiv.

[9]  B. Bengsch,et al.  Use of Mass Cytometry to Profile Human T Cell Exhaustion , 2020, Frontiers in Immunology.

[10]  Howard Y. Chang,et al.  c-Jun overexpression in CAR T cells induces exhaustion resistance , 2019, Nature.

[11]  Hoguen Kim,et al.  VEGF-A drives TOX-dependent T cell exhaustion in anti–PD-1–resistant microsatellite stable colorectal cancers , 2019, Science Immunology.

[12]  A. Heimberger,et al.  Shortened ex vivo manufacturing time of EGFRvIII-specific chimeric antigen receptor (CAR) T cells reduces immune exhaustion and enhances antiglioma therapeutic function , 2019, Journal of Neuro-Oncology.

[13]  Gong-Hong Wei,et al.  Enhancer Dysfunction in 3D Genome and Disease , 2019, Cells.

[14]  Yan Zhang,et al.  T Cell Dysfunction in Cancer Immunity and Immunotherapy , 2019, Front. Immunol..

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

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

[17]  Haifeng Song,et al.  A safe and potent anti-CD19 CAR T cell therapy , 2019, Nature Medicine.

[18]  P. Sachs,et al.  SMARCAD1 ATPase activity is required to silence endogenous retroviruses in embryonic stem cells , 2019, Nature Communications.

[19]  Aaron M. Newman,et al.  A functional subset of CD8+ T cells during chronic exhaustion is defined by SIRPα expression , 2019, Nature Communications.

[20]  C. Mackall,et al.  CAR T cell therapy: inroads to response and resistance , 2019, Nature Reviews Immunology.

[21]  F. Urnov,et al.  Disruption of the BCL11A Erythroid Enhancer Reactivates Fetal Hemoglobin in Erythroid Cells of Patients with β-Thalassemia Major , 2018, Molecular therapy. Methods & clinical development.

[22]  Ling Xu,et al.  T cell senescence and CAR-T cell exhaustion in hematological malignancies , 2018, Journal of Hematology & Oncology.

[23]  Hans Anton Schlößer,et al.  Cytokine release syndrome , 2018, Journal of Immunotherapy for Cancer.

[24]  Hans Bitter,et al.  Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia , 2018, Nature Medicine.

[25]  A. Shimabukuro-Vornhagen,et al.  Understanding cytokine release syndrome , 2018, Intensive Care Medicine.

[26]  D. Bigner,et al.  T-Cell Exhaustion Signatures Vary with Tumor Type and Are Severe in Glioblastoma , 2018, Clinical Cancer Research.

[27]  Howard Y. Chang,et al.  Enhancer connectome in primary human cells identifies target genes of disease-associated DNA elements , 2017, Nature Genetics.

[28]  Nigel P. Dyer,et al.  Integration of Kinase and Calcium Signaling at the Level of Chromatin Underlies Inducible Gene Activation in T Cells , 2017, The Journal of Immunology.

[29]  William J. Greenleaf,et al.  chromVAR: Inferring transcription factor-associated accessibility from single-cell epigenomic data , 2017, Nature Methods.

[30]  Christina S. Leslie,et al.  Chromatin states define tumor-specific T cell dysfunction and reprogramming , 2017, Nature.

[31]  D. Odom,et al.  Complexity and conservation of regulatory landscapes underlie evolutionary resilience of mammalian gene expression , 2017, bioRxiv.

[32]  A. Hoffmann,et al.  Exhaustion-associated regulatory regions in CD8+ tumor-infiltrating T cells , 2017, Proceedings of the National Academy of Sciences.

[33]  T. Reese,et al.  Making Mouse Models That Reflect Human Immune Responses. , 2017, Trends in immunology.

[34]  Mithat Gönen,et al.  Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection , 2017, Nature.

[35]  Todd M. Allen,et al.  The epigenetic landscape of T cell exhaustion , 2016, Science.

[36]  S. Berger,et al.  Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade , 2016, Science.

[37]  Aviv Regev,et al.  A Distinct Gene Module for Dysfunction Uncoupled from Activation in Tumor-Infiltrating T Cells , 2016, Cell.

[38]  Howard Y. Chang,et al.  HiChIP: efficient and sensitive analysis of protein-directed genome architecture , 2016, Nature Methods.

[39]  James T. Robinson,et al.  Juicebox Provides a Visualization System for Hi-C Contact Maps with Unlimited Zoom. , 2016, Cell systems.

[40]  Neva C. Durand,et al.  Juicer Provides a One-Click System for Analyzing Loop-Resolution Hi-C Experiments. , 2016, Cell systems.

[41]  M. Geyer,et al.  CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date. , 2016, Blood.

[42]  Julia V. Ponomarenko,et al.  Scalable Design of Paired CRISPR Guide RNAs for Genomic Deletion , 2016, bioRxiv.

[43]  R. Kaplan,et al.  4-1BB Costimulation Ameliorates T Cell Exhaustion Induced by Tonic Signaling of Chimeric Antigen Receptors , 2015, Nature Medicine.

[44]  J. T. Erichsen,et al.  Enhancer Evolution across 20 Mammalian Species , 2015, Cell.

[45]  Derek W Wright,et al.  Gateways to the FANTOM5 promoter level mammalian expression atlas , 2015, Genome Biology.

[46]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[47]  T. Meehan,et al.  An atlas of active enhancers across human cell types and tissues , 2014, Nature.

[48]  D. Odom,et al.  Evolution of transcription factor binding in metazoans — mechanisms and functional implications , 2014, Nature Reviews Genetics.

[49]  William Stafford Noble,et al.  Statistical confidence estimation for Hi-C data reveals regulatory chromatin contacts , 2014, Genome research.

[50]  J. Rasko,et al.  CTCF and BORIS in genome regulation and cancer. , 2014, Current opinion in genetics & development.

[51]  Thomas M. Schmitt,et al.  Re‐adapting T cells for cancer therapy: from mouse models to clinical trials , 2014, Immunological reviews.

[52]  Bernd Hauck,et al.  Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. , 2013, The New England journal of medicine.

[53]  J. Yates,et al.  MicroRNA Regulation of Molecular Networks Mapped by Global MicroRNA, mRNA, and Protein Expression in Activated T Lymphocytes , 2011, The Journal of Immunology.

[54]  Michael J. Zilliox,et al.  Phenotype, Function, and Gene Expression Profiles of Programmed Death-1hi CD8 T Cells in Healthy Human Adults , 2011, The Journal of Immunology.

[55]  J. Kirkwood,et al.  Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen–specific CD8+ T cell dysfunction in melanoma patients , 2010, The Journal of experimental medicine.

[56]  C. Glass,et al.  Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. , 2010, Molecular cell.

[57]  Cory Y. McLean,et al.  GREAT improves functional interpretation of cis-regulatory regions , 2010, Nature Biotechnology.

[58]  Xue-jie Wu,et al.  Hepatoma cells up-regulate expression of programmed cell death-1 on T cells. , 2008, World journal of gastroenterology.

[59]  E. Wherry,et al.  Molecular signature of CD8+ T cell exhaustion during chronic viral infection. , 2007, Immunity.

[60]  Shelly C. Lu,et al.  Nrf1 and Nrf2 Regulate Rat Glutamate-Cysteine Ligase Catalytic Subunit Transcription Indirectly via NF-κB and AP-1 , 2005, Molecular and Cellular Biology.

[61]  J. Lieberman,et al.  A transgenic mouse model to analyze CD8(+) effector T cell differentiation in vivo. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[62]  T. Toki,et al.  Molecular Cloning and Functional Characterization of a New Cap’n’ Collar Family Transcription Factor Nrf3* , 1999, The Journal of Biological Chemistry.

[63]  J. Altman,et al.  Viral Immune Evasion Due to Persistence of Activated T Cells Without Effector Function , 1998, The Journal of experimental medicine.

[64]  T. Baumruker,et al.  Nrf1 in a complex with fosB, c-jun, junD and ATF2 forms the AP1 component at the TNF alpha promoter in stimulated mast cells. , 1998, Nucleic acids research.

[65]  T. Curran,et al.  Maf and Nrl can bind to AP-1 sites and form heterodimers with Fos and Jun. , 1994, Oncogene.

[66]  Rolf M. Zinkernagel,et al.  Virus persistence in acutely infected immunocompetent mice by exhaustion of antiviral cytotoxic effector T cells , 1993, Nature.

[67]  Peng-Kai Wu Faculty Opinions recommendation of Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. , 2022, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.