Distinct Signaling of Coreceptors Regulates Specific Metabolism Pathways and Impacts Memory Development in CAR T Cells.

Chimeric antigen receptors (CARs) redirect T cell cytotoxicity against cancer cells, providing a promising approach to cancer immunotherapy. Despite extensive clinical use, the attributes of CAR co-stimulatory domains that impact persistence and resistance to exhaustion of CAR-T cells remain largely undefined. Here, we report the influence of signaling domains of coreceptors CD28 and 4-1BB on the metabolic characteristics of human CAR T cells. Inclusion of 4-1BB in the CAR architecture promoted the outgrowth of CD8(+) central memory T cells that had significantly enhanced respiratory capacity, increased fatty acid oxidation and enhanced mitochondrial biogenesis. In contrast, CAR T cells with CD28 domains yielded effector memory cells with a genetic signature consistent with enhanced glycolysis. These results provide, at least in part, a mechanistic insight into the differential persistence of CAR-T cells expressing 4-1BB or CD28 signaling domains in clinical trials and inform the design of future CAR T cell therapies.

[1]  S. Sleijfer,et al.  Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

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

[3]  I. Pastan,et al.  Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains , 2009, Proceedings of the National Academy of Sciences.

[4]  D. Nicholls,et al.  Spare Respiratory Capacity Rather Than Oxidative Stress Regulates Glutamate Excitotoxicity after Partial Respiratory Inhibition of Mitochondrial Complex I with Rotenone , 2007, The Journal of Neuroscience.

[5]  S. Gottschalk,et al.  Design and development of therapies using chimeric antigen receptor‐expressing T cells , 2014, Immunological reviews.

[6]  T. Brocker Chimeric Fv-ζ or Fv-ε receptors are not sufficient to induce activation or cytokine production in peripheral T cells , 2000 .

[7]  J. Zapata,et al.  T Cell Costimulation with Anti-CD137 Monoclonal Antibodies Is Mediated by K63–Polyubiquitin-Dependent Signals from Endosomes , 2013, The Journal of Immunology.

[8]  W. Wilson,et al.  B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. , 2012, Blood.

[9]  I. Pastan,et al.  Antitumor activity of SS(dsFv)PE38 and SS1(dsFv)PE38, recombinant antimesothelin immunotoxins against human gynecologic cancers grown in organotypic culture in vitro. , 2002, Clinical cancer research : an official journal of the American Association for Cancer Research.

[10]  G. Semenza,et al.  Control of TH17/Treg Balance by Hypoxia-Inducible Factor 1 , 2011, Cell.

[11]  B. Levine,et al.  Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. , 2010, Cancer research.

[12]  W. Leonard,et al.  Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. , 2013, Immunity.

[13]  D. Green,et al.  Metabolic checkpoints in activated T cells , 2012, Nature Immunology.

[14]  Matthew J. Frigault,et al.  ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells. , 2014, Blood.

[15]  S. Fu,et al.  Human T cell activation. III. Rapid induction of a phosphorylated 28 kD/32 kD disulfide-linked early activation antigen (EA 1) by 12-o- tetradecanoyl phorbol-13-acetate, mitogens, and antigens , 1986, The Journal of experimental medicine.

[16]  P. Marrack,et al.  Control of homeostasis of CD8+ memory T cells by opposing cytokines. , 2000, Science.

[17]  David L. Porter,et al.  T Cells with Chimeric Antigen Receptors Have Potent Antitumor Effects and Can Establish Memory in Patients with Advanced Leukemia , 2011, Science Translational Medicine.

[18]  Pamela A Shaw,et al.  Chimeric antigen receptor T cells for sustained remissions in leukemia. , 2014, The New England journal of medicine.

[19]  C. Thompson,et al.  The CD28 signaling pathway regulates glucose metabolism. , 2002, Immunity.

[20]  Leo Lefrançois,et al.  Cytokine control of memory T-cell development and survival , 2003, Nature Reviews Immunology.

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

[22]  David A Ferrick,et al.  Advances in measuring cellular bioenergetics using extracellular flux. , 2008, Drug discovery today.

[23]  Seth M Steinberg,et al.  T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial , 2015, The Lancet.

[24]  T. Watts,et al.  ERK-Dependent Bim Modulation Downstream of the 4-1BB-TRAF1 Signaling Axis Is a Critical Mediator of CD8 T Cell Survival In Vivo1 , 2008, The Journal of Immunology.

[25]  Sankha S. Basu,et al.  Stable Isotope Labeling by Essential Nutrients in Cell Culture for Preparation of Labeled Coenzyme A and Its Thioesters , 2011, Analytical chemistry.

[26]  M. Kalos,et al.  Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. , 2013, Immunity.

[27]  David K. Finlay,et al.  PDK1 regulation of mTOR and hypoxia-inducible factor 1 integrate metabolism and migration of CD8+ T cells , 2012, The Journal of experimental medicine.

[28]  J. Rathmell,et al.  Metabolic regulation of T lymphocytes. , 2013, Annual review of immunology.

[29]  Gang Wang,et al.  A Phase I Study on Adoptive Immunotherapy Using Gene-Modified T Cells for Ovarian Cancer , 2006, Clinical Cancer Research.

[30]  Ton N. Schumacher,et al.  Adoptive cellular therapy: A race to the finish line , 2015, Science Translational Medicine.

[31]  Michel Sadelain,et al.  Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. , 2011, Blood.

[32]  W. Wilson,et al.  Construction and Preclinical Evaluation of an Anti-CD19 Chimeric Antigen Receptor , 2009, Journal of immunotherapy.

[33]  Sankha S. Basu,et al.  SILEC: a protocol for generating and using isotopically labeled coenzyme A mass spectrometry standards , 2011, Nature Protocols.

[34]  D. Campana,et al.  Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. , 2009, Molecular therapy : the journal of the American Society of Gene Therapy.

[35]  Sankha S. Basu,et al.  Inhibition of Neuronal Cell Mitochondrial Complex I with Rotenone Increases Lipid β-Oxidation, Supporting Acetyl-Coenzyme A Levels* , 2014, The Journal of Biological Chemistry.

[36]  Qing He,et al.  CD19-Targeted T Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy-Refractory Acute Lymphoblastic Leukemia , 2013, Science Translational Medicine.

[37]  David L. Porter,et al.  Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia , 2015, Science Translational Medicine.

[38]  G. V. D. van der Windt,et al.  Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. , 2012, Immunity.

[39]  B. Faubert,et al.  CD8 memory T cells have a bioenergetic advantage that underlies their rapid recall ability , 2013, Proceedings of the National Academy of Sciences.

[40]  T. Brocker Chimeric Fv-zeta or Fv-epsilon receptors are not sufficient to induce activation or cytokine production in peripheral T cells. , 2000, Blood.

[41]  Michel Sadelain,et al.  The basic principles of chimeric antigen receptor design. , 2013, Cancer discovery.

[42]  Ajit S. Divakaruni,et al.  Mitochondrial uncoupling and lifespan , 2010, Mechanisms of Ageing and Development.

[43]  E. Pearce,et al.  Metabolic pathways in immune cell activation and quiescence. , 2013, Immunity.

[44]  D. Nicholls,et al.  Neuronal Glutamate and Gabaa Receptor Function in Health and Disease Spare Respiratory Capacity, Oxidative Stress and Excitotoxicity Glutamate as an Excitotoxin the Role of Oxidative Stress , 2022 .

[45]  D. Torigian,et al.  Mesothelin-Specific Chimeric Antigen Receptor mRNA-Engineered T Cells Induce Antitumor Activity in Solid Malignancies , 2013, Cancer Immunology Research.

[46]  Y. Kurosawa,et al.  Expression of chimeric receptor composed of immunoglobulin-derived V regions and T-cell receptor-derived C regions. , 1987, Biochemical and biophysical research communications.

[47]  B. Levine,et al.  T Cells Expressing Chimeric Antigen Receptors Can Cause Anaphylaxis in Humans , 2013, Cancer Immunology Research.

[48]  Z. Eshhar,et al.  Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. , 1993, Proceedings of the National Academy of Sciences of the United States of America.

[49]  Qicheng Ma,et al.  Activation of a metabolic gene regulatory network downstream of mTOR complex 1. , 2010, Molecular cell.

[50]  Russell G. Jones,et al.  Enhancing CD8 T-cell memory by modulating fatty acid metabolism , 2009, Nature.

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

[52]  B. Levine,et al.  Adoptive immunotherapy for cancer or viruses. , 2014, Annual review of immunology.

[53]  A. Scott,et al.  Construction and characterisation of a functional CD19 specific single chain Fv fragment for immunotherapy of B lineage leukaemia and lymphoma. , 1997, Molecular immunology.