Mechanisms of T cell exhaustion guiding next-generation immunotherapy

Trends in Cancer - Tập 8 - Trang 726-734 - 2022
Caitlin C. Zebley1,2, Ben Youngblood1
1Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
2Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA

Tài liệu tham khảo

Zajac, 1998, Viral immune evasion due to persistence of activated T cells without effector function, J. Exp. Med., 188, 2205, 10.1084/jem.188.12.2205 Zinkernagel, 1993, Effector T-cell induction and T-cell memory versus peripheral deletion of T cells, Immunol. Rev., 133, 199, 10.1111/j.1600-065X.1993.tb01517.x Wherry, 2004, Antigen-independent memory CD8 T cells do not develop during chronic viral infection, Proc. Natl. Acad. Sci. U. S. A., 101, 16004, 10.1073/pnas.0407192101 Utzschneider, 2013, T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion, Nat. Immunol., 14, 603, 10.1038/ni.2606 Shin, 2007, CD8 T cell dysfunction during chronic viral infection, Curr. Opin. Immunol., 19, 408, 10.1016/j.coi.2007.06.004 Man, 2017, Transcription factor IRF4 promotes CD8+ T cell exhaustion and limits the development of memory-like T cells during chronic infection, Immunity, 47, 1129, 10.1016/j.immuni.2017.11.021 Martinez, 2015, The transcription factor NFAT promotes exhaustion of activated CD8(+) T cells, Immunity, 42, 265, 10.1016/j.immuni.2015.01.006 Alfei, 2019, TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection, Nature, 571, 265, 10.1038/s41586-019-1326-9 Khan, 2019, TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion, Nature, 571, 211, 10.1038/s41586-019-1325-x Scott, 2019, TOX is a critical regulator of tumour-specific T cell differentiation, Nature, 571, 270, 10.1038/s41586-019-1324-y Frias, 2021, Epigenetic regulation of T cell adaptive immunity, Immunol. Rev., 300, 9, 10.1111/imr.12943 Shields, 2017, Indicators of responsiveness to immune checkpoint inhibitors, Sci. Rep., 7, 807, 10.1038/s41598-017-01000-2 Morad, 2021, Hallmarks of response, resistance, and toxicity to immune checkpoint blockade, Cell, 184, 5309, 10.1016/j.cell.2021.09.020 Ghoneim, 2017, De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation, Cell, 170, 142, 10.1016/j.cell.2017.06.007 Pauken, 2016, Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade, Science, 354, 1160, 10.1126/science.aaf2807 Liu, 2021, LSD1 inhibition sustains T cell invigoration with a durable response to PD-1 blockade, Nat. Commun., 12, 6831, 10.1038/s41467-021-27179-7 Prinzing, 2021, Deleting DNMT3A in CAR T cells prevents exhaustion and enhances antitumor activity, Sci. Transl. Med., 13, 10.1126/scitranslmed.abh0272 Abdel-Hakeem, 2021, Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation, Nat. Immunol., 22, 1008, 10.1038/s41590-021-00975-5 Yates, 2021, Epigenetic scars of CD8+ T cell exhaustion persist after cure of chronic infection in humans, Nat. Immunol., 22, 1020, 10.1038/s41590-021-00979-1 Im, 2016, Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy, Nature, 537, 417, 10.1038/nature19330 Brummelman, 2018, High-dimensional single cell analysis identifies stem-like cytotoxic CD8(+) T cells infiltrating human tumors, J. Exp. Med., 215, 2520, 10.1084/jem.20180684 Gettinger, 2018, A dormant TIL phenotype defines non-small cell lung carcinomas sensitive to immune checkpoint blockers, Nat. Commun., 9, 3196, 10.1038/s41467-018-05032-8 Utzschneider, 2020, Early precursor T cells establish and propagate T cell exhaustion in chronic infection, Nat. Immunol., 21, 1256, 10.1038/s41590-020-0760-z Sade-Feldman, 2018, Defining T cell states associated with response to checkpoint immunotherapy in melanoma, Cell, 175, 998, 10.1016/j.cell.2018.10.038 Hudson, 2019, Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1+ stem-like CD8+ T cells during chronic infection, Immunity, 51, 1043, 10.1016/j.immuni.2019.11.002 Pace, 2018, The epigenetic control of stemness in CD8+ T cell fate commitment, Science, 359, 177, 10.1126/science.aah6499 Lee, 2021, Tet2 inactivation enhances the antitumor activity of tumor-infiltrating lymphocytes, Cancer Res., 81, 1965, 10.1158/0008-5472.CAN-20-3213 Henning, 2018, Epigenetic control of CD8+ T cell differentiation, Nat. Rev. Immunol., 18, 340, 10.1038/nri.2017.146 Zhong, 2022, Hierarchical regulation of the resting and activated T cell epigenome by major transcription factor families, Nat. Immunol., 23, 122, 10.1038/s41590-021-01086-x Abdelsamed, 2020, Beta cell-specific CD8+ T cells maintain stem cell memory-associated epigenetic programs during type 1 diabetes, Nat. Immunol., 21, 578, 10.1038/s41590-020-0633-5 Zebley, 2021, CD19-CAR T cells undergo exhaustion DNA methylation programming in patients with acute lymphoblastic leukemia, Cell Rep., 37, 10.1016/j.celrep.2021.110079 Gerhard, 2021, Tumor-infiltrating dendritic cell states are conserved across solid human cancers, J. Exp. Med., 218, 10.1084/jem.20200264 Di Pilato, 2021, CXCR6 positions cytotoxic T cells to receive critical survival signals in the tumor microenvironment, Cell, 184, 4512, 10.1016/j.cell.2021.07.015 Flynn, 2014, Stem memory T cells (TSCM)—their role in cancer and HIV immunotherapies, Clin. Transl. Immunol., 3, 10.1038/cti.2014.16 Gattinoni, 2011, A human memory T cell subset with stem cell-like properties, Nat. Med., 17, 1290, 10.1038/nm.2446 Sabatino, 2016, Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies, Blood, 128, 519, 10.1182/blood-2015-11-683847 Klebanoff, 2012, Sorting through subsets: which T-cell populations mediate highly effective adoptive immunotherapy?, J. Immunother., 35, 651, 10.1097/CJI.0b013e31827806e6 Chen, 2021, Integrative bulk and single-cell profiling of pre-manufacture T-cell populations reveals factors mediating long-term persistence of CAR T-cell therapy, Cancer Discov., 11, 2186, 10.1158/2159-8290.CD-20-1677 Qin, 2018, Murine pre-B-cell ALL induces T-cell dysfunction not fully reversed by introduction of a chimeric antigen receptor, Blood, 132, 1899, 10.1182/blood-2017-12-815548 Palma, 2017, T cells in chronic lymphocytic leukemia display dysregulated expression of immune checkpoints and activation markers, Haematologica, 102, 562, 10.3324/haematol.2016.151100 Sommermeyer, 2016, Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo, Leukemia, 30, 492, 10.1038/leu.2015.247 Fraietta, 2018, Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia, Nat. Med., 24, 563, 10.1038/s41591-018-0010-1 Summers, 2021, Hematopoietic cell transplantation after CD19 chimeric antigen receptor T cell-induced acute lymphoblastic lymphoma remission confers a leukemia-free survival advantage, Transplant. Cell. Ther., 28, 21, 10.1016/j.jtct.2021.10.003 Lynn, 2019, c-Jun overexpression in CAR T cells induces exhaustion resistance, Nature, 576, 293, 10.1038/s41586-019-1805-z Seo, 2021, BATF and IRF4 cooperate to counter exhaustion in tumor-infiltrating CAR T cells, Nat. Immunol., 22, 983, 10.1038/s41590-021-00964-8 Wei, 2019, Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy, Nature, 576, 471, 10.1038/s41586-019-1821-z Shifrut, 2018, Genome-wide CRISPR screens in primary human T cells reveal key regulators of immune function, Cell, 175, 1958, 10.1016/j.cell.2018.10.024 Fraietta, 2018, Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells, Nature, 558, 307, 10.1038/s41586-018-0178-z Wang, 2021, Low-dose decitabine priming endows CAR T cells with enhanced and persistent antitumour potential via epigenetic reprogramming, Nat. Commun., 12, 409, 10.1038/s41467-020-20696-x Ley, 2010, DNMT3A mutations in acute myeloid leukemia, N. Engl. J. Med., 363, 2424, 10.1056/NEJMoa1005143 Kramer, 2017, Dnmt3a regulates T-cell development and suppresses T-ALL transformation, Leukemia, 31, 2479, 10.1038/leu.2017.89 Tovy, 2020, Tissue-biased expansion of DNMT3A-mutant clones in a mosaic individual is associated with conserved epigenetic erosion, Cell Stem Cell, 27, 326, 10.1016/j.stem.2020.06.018 van den Akker, 2020, Dynamic clonal hematopoiesis and functional T-cell immunity in a supercentenarian, Leukemia, 35, 2125, 10.1038/s41375-020-01086-0 Gibson, 2021, Donor clonal hematopoiesis and recipient outcomes after transplantation, J. Clin. Oncol., 40, 189, 10.1200/JCO.21.02286 Zebley, 2020, Rewriting history: epigenetic reprogramming of CD8+ T cell differentiation to enhance immunotherapy, Trends Immunol., 41, 665, 10.1016/j.it.2020.06.008 Topper, 2020, The emerging role of epigenetic therapeutics in immuno-oncology, Nat. Rev. Clin. Oncol., 17, 75, 10.1038/s41571-019-0266-5 Zebley, 2021, Proinflammatory cytokines promote TET2-mediated DNA demethylation during CD8 T cell effector differentiation, Cell Rep., 37, 10.1016/j.celrep.2021.109796