Different Subsets of T Cells, Memory, Effector Functions, and CAR-T Immunotherapy

This review is focused on different subsets of T cells: CD4 and CD8, memory and effector functions, and their role in CAR-T therapy––a cellular adoptive immunotherapy with T cells expressing chimeric antigen receptor. The CAR-T cells recognize tumor antigens and induce cytotoxic activities against tumor cells. Recently, differences in T cell functions and the role of memory and effector T cells were shown to be important in CAR-T cell immunotherapy. The CD4+ subsets (Th1, Th2, Th9, Th17, Th22, Treg, and Tfh) and CD8+ memory and effector subsets differ in extra-cellular (CD25, CD45RO, CD45RA, CCR-7, L-Selectin [CD62L], etc.); intracellular markers (FOXP3); epigenetic and genetic programs; and metabolic pathways (catabolic or anabolic); and these differences can be modulated to improve CAR-T therapy. In addition, CD4+ Treg cells suppress the efficacy of CAR-T cell therapy, and different approaches to overcome this suppression are discussed in this review. Thus, next-generation CAR-T immunotherapy can be improved, based on our knowledge of T cell subsets functions, differentiation, proliferation, and signaling pathways to generate more active CAR-T cells against tumors.

[1]  P. Darcy,et al.  Adoptive cell therapy (ACT) utilizing gene-modified T cells expressing chimeric antigen receptors (CAR) has emerged as a promising regimen for the treatment of a broad range of cancers including chronic lymphoid leukaemia and neuroblastoma , 2013 .

[2]  J. Trapani,et al.  Adoptive transfer of gene-engineered CD4+ helper T cells induces potent primary and secondary tumor rejection. , 2005, Blood.

[3]  J. Rathmell,et al.  Cutting Edge: Distinct Glycolytic and Lipid Oxidative Metabolic Programs Are Essential for Effector and Regulatory CD4+ T Cell Subsets , 2011, The Journal of Immunology.

[4]  A. Schmidt,et al.  CD28 costimulation Impairs the efficacy of a redirected t-cell antitumor attack in the presence of regulatory t cells which can be overcome by preventing Lck activation. , 2011, Molecular therapy : the journal of the American Society of Gene Therapy.

[5]  Martin Pule,et al.  Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. , 2011, Blood.

[6]  Gisen Kim,et al.  Interleukin 10 acts on regulatory T cells to maintain expression of the transcription factor Foxp3 and suppressive function in mice with colitis , 2009, Nature Immunology.

[7]  E. Yang,et al.  Transcriptional insights into the CD8+ T cell response to infection and memory T cell formation , 2013, Nature Immunology.

[8]  S. Sleijfer,et al.  T cell receptor-engineered T cells to treat solid tumors: T cell processing toward optimal T cell fitness. , 2014, Human gene therapy methods.

[9]  H. Abken,et al.  Of CARs and TRUCKs: chimeric antigen receptor (CAR) T cells engineered with an inducible cytokine to modulate the tumor stroma , 2014, Immunological reviews.

[10]  Z. Eshhar,et al.  Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. , 1989, Proceedings of the National Academy of Sciences of the United States of America.

[11]  H. Abken Adoptive therapy with CAR redirected T cells: the challenges in targeting solid tumors. , 2015, Immunotherapy.

[12]  Z. Eshhar,et al.  The emergence of T-bodies/CAR T cells. , 2014, Cancer journal.

[13]  S. Gottschalk,et al.  CAR T Cells for Solid Tumors: Armed and Ready to Go? , 2014, Cancer journal.

[14]  S. Heimfeld,et al.  Anti-CD19 Chimeric Antigen Receptor-Modified T Cell Therapy for B Cell Non-Hodgkin Lymphoma and Chronic Lymphocytic Leukemia: Fludarabine and Cyclophosphamide Lymphodepletion Improves In Vivo Expansion and Persistence of CAR-T Cells and Clinical Outcomes , 2015 .

[15]  Peng Qiu,et al.  Individual Motile CD4+ T Cells Can Participate in Efficient Multikilling through Conjugation to Multiple Tumor Cells , 2015, Cancer Immunology Research.

[16]  D. Maloney,et al.  Accepted Article Preview : Published ahead of advance online publication , 2016 .

[17]  Marcela V Maus,et al.  Antibody-modified T cells: CARs take the front seat for hematologic malignancies. , 2014, Blood.

[18]  M. Sadelain,et al.  Tumor-Targeted Human T Cells Expressing CD28-Based Chimeric Antigen Receptors Circumvent CTLA-4 Inhibition , 2015, PloS one.

[19]  T. Waldmann,et al.  IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T Cells , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[20]  Z. Eshhar,et al.  Therapeutic Potential of T Cell Chimeric Antigen Receptors (CARs) in Cancer Treatment: Counteracting Off-Tumor Toxicities for Safe CAR T Cell Therapy. , 2016, Annual review of pharmacology and toxicology.

[21]  Hao Shen,et al.  Requirement for CD4 T Cell Help in Generating Functional CD8 T Cell Memory , 2003, Science.

[22]  C. Klebanoff,et al.  Paths to stemness: building the ultimate antitumour T cell , 2012, Nature Reviews Cancer.

[23]  G. V. D. Windt,et al.  Metabolic switching and fuel choice during T‐cell differentiation and memory development , 2012, Immunological reviews.

[24]  S. Rosenberg,et al.  Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells , 2005, The Journal of experimental medicine.

[25]  Brian Keith,et al.  Distinct Signaling of Coreceptors Regulates Specific Metabolism Pathways and Impacts Memory Development in CAR T Cells. , 2016, Immunity.

[26]  A. Rudensky,et al.  Control of the Inheritance of Regulatory T Cell Identity by a cis Element in the Foxp3 Locus , 2014, Cell.

[27]  H. Abken,et al.  CAR T cells transform to trucks: chimeric antigen receptor–redirected T cells engineered to deliver inducible IL-12 modulate the tumour stroma to combat cancer , 2012, Cancer Immunology, Immunotherapy.

[28]  yang-xin fu,et al.  Immunotherapy and tumor microenvironment. , 2016, Cancer letters.

[29]  G. Ashton,et al.  Differential Role of Th1 and Th2 Cytokines in Autotoxicity Driven by CD19-Specific Second-Generation Chimeric Antigen Receptor T Cells in a Mouse Model , 2014, The Journal of Immunology.

[30]  Sing Sing Way,et al.  Regulatory T cell memory , 2015, Nature Reviews Immunology.

[31]  F. Marincola,et al.  Mitochondrial Membrane Potential Identifies Cells with Enhanced Stemness for Cellular Therapy. , 2016, Cell metabolism.

[32]  Chih-Hao Chang,et al.  Fueling Immunity: Insights into Metabolism and Lymphocyte Function , 2013, Science.

[33]  Raphael Sandaltzopoulos,et al.  Chimeric Antigen Receptor T Cells with Dissociated Signaling Domains Exhibit Focused Antitumor Activity with Reduced Potential for Toxicity In Vivo , 2013, Cancer Immunology Research.

[34]  M. Smyth,et al.  Anti-PD-1 Antibody Therapy Potently Enhances the Eradication of Established Tumors By Gene-Modified T Cells , 2013, Clinical Cancer Research.

[35]  S. Rosenberg,et al.  Modulating the differentiation status of ex vivo-cultured anti-tumor T cells using cytokine cocktails , 2013, Cancer Immunology, Immunotherapy.

[36]  D. Maloney,et al.  Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo , 2015, Leukemia.

[37]  T. Forsthuber,et al.  T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. , 2015, Cytokine.

[38]  M. Brown,et al.  Different cytokine and stimulation conditions influence the expansion and immune phenotype of third-generation chimeric antigen receptor T cells specific for tumor antigen GD2. , 2015, Cytotherapy.

[39]  C. Creighton,et al.  Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. , 2014, Blood.

[40]  G. Coukos,et al.  Redirected antitumor activity of primary human lymphocytes transduced with a fully human anti-mesothelin chimeric receptor. , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.

[41]  S. Rosenberg,et al.  IL-2 administration increases CD4+ CD25(hi) Foxp3+ regulatory T cells in cancer patients. , 2006, Blood.

[42]  H. Abken,et al.  IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively Muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. , 2011, Cancer research.

[43]  Mike Gough,et al.  Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. , 2008, The Journal of clinical investigation.

[44]  S. Berger,et al.  Cutting Edge: Persistently Open Chromatin at Effector Gene Loci in Resting Memory CD8+ T Cells Independent of Transcriptional Status , 2011, The Journal of Immunology.

[45]  S. Steinberg,et al.  Levels of peripheral CD4(+)FoxP3(+) regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. , 2012, Blood.

[46]  C. Klebanoff,et al.  Sorting Through Subsets: Which T-Cell Populations Mediate Highly Effective Adoptive Immunotherapy? , 2012, Journal of immunotherapy.

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