Improving CAR-T immunotherapy: Overcoming the challenges of T cell exhaustion
Tóm tắt
Từ khóa
Tài liệu tham khảo
Tan, 2020, Adoptive T-cell therapy for HBV-associated HCC and HBV infection, Antivir Res, 176, 10.1016/j.antiviral.2020.104748
Laskowski, 2020, Adoptive cell therapy: living drugs against cancer, J Exp Med, 217, e20200377, 10.1084/jem.20200377
Zhou, 2021, Challenges and opportunities of using adoptive T-cell therapy as part of an HIV cure strategy, J Infect Dis, 223, 38, 10.1093/infdis/jiaa223
Fong, 2002, Immunotherapy in autoimmune diseases, Ann Acad Med Singap, 31, 702
Duffy, 2019, Adoptive transfer of regulatory T cells as a promising immunotherapy for the treatment of multiple sclerosis, Front Neurosci, 13, 1107, 10.3389/fnins.2019.01107
Eshhar, 1993, 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, Proc Natl Acad Sci USA, 90, 720, 10.1073/pnas.90.2.720
Marofi, 2021, CAR T cells in solid tumors: challenges and opportunities, Stem Cell Res Ther, 12, 81, 10.1186/s13287-020-02128-1
Zmievskaya, 2021, Application of CAR-T cell therapy beyond oncology: autoimmune diseases and viral infections, Biomedicines, 9, 59, 10.3390/biomedicines9010059
Finney, 2019, CD19 CAR T cell product and disease attributes predict leukemia remission durability, J Clin Invest, 129, 2123, 10.1172/JCI125423
Hu, 2021, Anti-CD19 CAR-T cell therapy bridge to HSCT decreases the relapse rate and improves the long-term survival of R/R B-ALL patients: a systematic review and meta-analysis, Ann Hematol, 100, 1003, 10.1007/s00277-021-04451-w
Xu, 2020, Challenges and clinical strategies of CAR T-cell therapy for acute lymphoblastic leukemia: overview and developments, Front Immunol, 11
Majzner, 2018, Tumor antigen escape from CAR T-cell therapy, Cancer Discov, 8, 1219, 10.1158/2159-8290.CD-18-0442
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
Rodriguez-Garcia, 2020, CAR-T cells hit the tumor microenvironment: strategies to overcome tumor escape, Front Immunol, 11, 1109, 10.3389/fimmu.2020.01109
Lesch, 2020, Determinants of response and resistance to CAR T cell therapy, Semin Cancer Biol, 65, 80, 10.1016/j.semcancer.2019.11.004
Sterner, 2021, CAR-T cell therapy: current limitations and potential strategies, Blood Cancer J, 11, 69, 10.1038/s41408-021-00459-7
Liu, 2021, Enhancing CAR-T cell efficacy in solid tumors by targeting the tumor microenvironment, Cell Mol Immunol, 18, 1085, 10.1038/s41423-021-00655-2
Carmenate, 2018, Blocking IL-2 signal in vivo with an IL-2 antagonist reduces tumor growth through the control of regulatory T cells, J Immunol, 200, 3475, 10.4049/jimmunol.1700433
Louis, 2011, Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma, Blood, 118, 6050, 10.1182/blood-2011-05-354449
Majzner, 2019, Clinical lessons learned from the first leg of the CAR T cell journey, Nat Med, 25, 1341, 10.1038/s41591-019-0564-6
Bucks, 2009, Chronic antigen stimulation alone is sufficient to drive CD8+ T cell exhaustion, J Immunol, 182, 6697, 10.4049/jimmunol.0800997
Pauken, 2015, Overcoming T cell exhaustion in infection and cancer, Trends Immunol, 36, 265, 10.1016/j.it.2015.02.008
Fraietta, 2021, Author correction: determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia, Nat Med, 27, 561, 10.1038/s41591-021-01248-2
Wang, 2019, In vitro tumor cell rechallenge for predictive evaluation of chimeric antigen receptor T cell antitumor function, J Vis Exp, e59275
Yang, 2020, TIGIT expression is associated with T-cell suppression and exhaustion and predicts clinical outcome and anti-PD-1 response in follicular lymphoma, Clin Cancer Res, 26, 5217, 10.1158/1078-0432.CCR-20-0558
Lindner, 2020, Chimeric antigen receptor signaling: functional consequences and design implications, Sci Adv, 6, eaaz3223, 10.1126/sciadv.aaz3223
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
Saeidi, 2018, T-cell exhaustion in chronic infections: reversing the state of exhaustion and reinvigorating optimal protective immune responses, Front Immunol, 9, 2569, 10.3389/fimmu.2018.02569
Wherry, 2015, Molecular and cellular insights into T cell exhaustion, Nat Rev Immunol, 15, 486, 10.1038/nri3862
Dong, 2019, CD4+ T cell exhaustion revealed by high PD-1 and LAG-3 expression and the loss of helper T cell function in chronic hepatitis B, BMC Immunol, 20, 27, 10.1186/s12865-019-0309-9
Han, 2010, Role of antigen persistence and dose for CD4+ T-cell exhaustion and recovery, Proc Natl Acad Sci USA, 107, 20453, 10.1073/pnas.1008437107
Gerlach, 2010, One naive T cell, multiple fates in CD8+ T cell differentiation, J Exp Med, 207, 1235, 10.1084/jem.20091175
Cornberg, 2013, Clonal exhaustion as a mechanism to protect against severe immunopathology and death from an overwhelming CD8 T cell response, Front Immunol, 4, 475, 10.3389/fimmu.2013.00475
Obar, 2010, Memory CD8+ T cell differentiation, Ann N Y Acad Sci, 1183, 251, 10.1111/j.1749-6632.2009.05126.x
Cosma, 2019, Impact of epitope density on CD8+ T cell development and function, Mol Immunol, 113, 120, 10.1016/j.molimm.2019.03.010
Kroger, 2007, Cutting edge: CD8+ T cell clones possess the potential to differentiate into both high- and low-avidity effector cells, J Immunol, 179, 748, 10.4049/jimmunol.179.2.748
Kroger, 2008, Cutting edge: dendritic cells prime a high avidity CTL response independent of the level of presented antigen, J Immunol, 180, 5784, 10.4049/jimmunol.180.9.5784
Sharma, 2011, Increased sensitivity to antigen in high avidity CD8+ T cells results from augmented membrane proximal T-cell receptor signal transduction, Immunology, 133, 307, 10.1111/j.1365-2567.2011.03440.x
Curdy, 2019, Regulatory mechanisms of inhibitory immune checkpoint receptors expression, Trends Cell Biol, 29, 777, 10.1016/j.tcb.2019.07.002
McLane, 2019, CD8 T cell exhaustion during chronic viral infection and cancer, Annu Rev Immunol, 37, 457, 10.1146/annurev-immunol-041015-055318
Khan, 2019, TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion, Nature, 571, 211, 10.1038/s41586-019-1325-x
Ostroumov, 2021, Transcriptome profiling identifies TIGIT as a marker of T-cell exhaustion in liver cancer, Hepatology, 73, 1399, 10.1002/hep.31466
Jiang, 2020, Exhausted CD8+T cells in the tumor immune microenvironment: new pathways to therapy, Front Immunol, 11
Beltra, 2020, Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms, Immunity, 52, 825, 10.1016/j.immuni.2020.04.014
Chu, 2020, Charting the roadmap of T cell exhaustion, Immunity, 52, 724, 10.1016/j.immuni.2020.04.019
Philip, 2017, Chromatin states define tumour-specific T cell dysfunction and reprogramming, Nature, 545, 452, 10.1038/nature22367
Chen, 2019, TCF-1-centered transcriptional network drives an effector versus exhausted CD8 T cell-fate decision, Immunity, 51, 840, 10.1016/j.immuni.2019.09.013
Zander, 2019, CD4+ T cell help is required for the formation of a cytolytic CD8+ T cell subset that protects against chronic infection and cancer, Immunity, 51, 1028, 10.1016/j.immuni.2019.10.009
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
Kim, 2020, Single-cell transcriptome analysis reveals TOX as a promoting factor for T cell exhaustion and a predictor for anti-PD-1 responses in human cancer, Genome Med, 12, 22, 10.1186/s13073-020-00722-9
Liang, 2021, TOX as a potential target for immunotherapy in lymphocytic malignancies, Biomark Res, 9, 20, 10.1186/s40364-021-00275-y
Seo, 2021, Transcriptional regulatory network for the establishment of CD8+ T cell exhaustion, Exp Mol Med, 53, 202, 10.1038/s12276-021-00568-0
Seo, 2019, TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion, Proc Natl Acad Sci USA, 116, 12410, 10.1073/pnas.1905675116
Chen, 2019, NR4A transcription factors limit CAR T cell function in solid tumours, Nature, 567, 530, 10.1038/s41586-019-0985-x
Scott, 2019, TOX is a critical regulator of tumour-specific T cell differentiation, Nature, 571, 270, 10.1038/s41586-019-1324-y
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
Yao, 2019, Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection, Nat Immunol, 20, 890, 10.1038/s41590-019-0403-4
McLane, 2021, Role of nuclear localization in the regulation and function of T-bet and Eomes in exhausted CD8 T cells, Cell Rep, 35, 10.1016/j.celrep.2021.109120
Li, 2018, High levels of Eomes promote exhaustion of anti-tumor CD8+ T cells, Front Immunol, 9, 2981, 10.3389/fimmu.2018.02981
Jia, 2019, Eomes+T-bet(low) CD8+ T cells are functionally impaired and are associated with poor clinical outcome in patients with acute myeloid leukemia, Cancer Res, 79, 1635, 10.1158/0008-5472.CAN-18-3107
Riley, 2009, PD-1 signaling in primary T cells, Immunol Rev, 229, 114, 10.1111/j.1600-065X.2009.00767.x
Jubel, 2020, The role of PD-1 in acute and chronic infection, Front Immunol, 11, 487, 10.3389/fimmu.2020.00487
Zuazo, 2017, Molecular mechanisms of programmed cell death-1 dependent T cell suppression: relevance for immunotherapy, Ann Transl Med, 5, 385, 10.21037/atm.2017.06.11
Bardhan, 2016, The PD1:PD-L1/2 pathway from discovery to clinical implementation, Front Immunol, 7, 550, 10.3389/fimmu.2016.00550
Patsoukis, 2012, Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation, Sci Signal, 5, ra46, 10.1126/scisignal.2002796
Ding, 2019, IFN-gamma down-regulates the PD-1 expression and assist nivolumab in PD-1-blockade effect on CD8+ T-lymphocytes in pancreatic cancer, BMC Cancer, 19, 1053, 10.1186/s12885-019-6145-8
Yan, 2018, CX3CR1 identifies PD-1 therapy-responsive CD8+ T cells that withstand chemotherapy during cancer chemoimmunotherapy, JCI Insight, 3, e97828, 10.1172/jci.insight.97828
Yamauchi, 2021, T-cell CX3CR1 expression as a dynamic blood-based biomarker of response to immune checkpoint inhibitors, Nat Commun, 12, 1402, 10.1038/s41467-021-21619-0
Serganova, 2017, Enhancement of PSMA-directed CAR adoptive immunotherapy by PD-1/PD-L1 blockade, Mol Ther Oncolytics, 4, 41, 10.1016/j.omto.2016.11.005
John, 2013, Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells, Clin Cancer Res, 19, 5636, 10.1158/1078-0432.CCR-13-0458
Adusumilli, 2021, A phase I trial of regional mesothelin-targeted CAR T-cell therapy in patients with malignant pleural disease, in combination with the anti-PD-1 agent pembrolizumab, Cancer Discov, 11, 2748, 10.1158/2159-8290.CD-21-0407
Rafiq, 2018, Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo, Nat Biotechnol, 36, 847, 10.1038/nbt.4195
Li, 2017, Enhanced cancer immunotherapy by chimeric antigen receptor-modified T cells engineered to secrete checkpoint inhibitors, Clin Cancer Res, 23, 6982, 10.1158/1078-0432.CCR-17-0867
Chen, 2021, Secretion of bispecific protein of anti-PD-1 fused with TGF-beta trap enhances antitumor efficacy of CAR-T cell therapy, Mol Ther Oncolytics, 21, 144, 10.1016/j.omto.2021.03.014
Cherkassky, 2016, Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition, J Clin Invest, 126, 3130, 10.1172/JCI83092
Liu, 2021, A novel dominant-negative PD-1 armored anti-CD19 CAR T cell is safe and effective against refractory/relapsed B cell lymphoma, Transl Oncol, 14, 10.1016/j.tranon.2021.101085
Lin, 2021, Cytotoxic effect of CLL1 CART cell immunotherapy with PD1 silencing on relapsed/refractory acute myeloid leukemia, Mol Med Rep, 23, 208, 10.3892/mmr.2021.11847
Liu, 2021, PD-1 silencing improves anti-tumor activities of human mesothelin-targeted CAR T cells, Hum Immunol, 82, 130, 10.1016/j.humimm.2020.12.002
Zhu, 2020, EGFRvIII-CAR-T Cells with PD-1 knockout have improved anti-glioma activity, Pathol Oncol Res, 26, 2135, 10.1007/s12253-019-00759-1
Rupp, 2017, CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells, Sci Rep, 7, 737, 10.1038/s41598-017-00462-8
Guo, 2018, Disruption of PD-1 enhanced the anti-tumor activity of chimeric antigen receptor T cells against hepatocellular carcinoma, Front Pharmacol, 9, 1118, 10.3389/fphar.2018.01118
Wang, 2021, Phase I study of CAR-T cells with PD-1 and TCR disruption in mesothelin-positive solid tumors, Cell Mol Immunol, 18, 2188, 10.1038/s41423-021-00749-x
Aliahmad, 2011, TOX is required for development of the CD4 T cell lineage gene program, J Immunol, 187, 5931, 10.4049/jimmunol.1101474
Xu, 2019, The transcription factor Tox2 drives T follicular helper cell development via regulating chromatin accessibility, Immunity, 51, 826, 10.1016/j.immuni.2019.10.006
Liu, 2019, Genome-wide analysis identifies NR4A1 as a key mediator of T cell dysfunction, Nature, 567, 525, 10.1038/s41586-019-0979-8
Li, 2019, Targeting NR4As, a new strategy to fine-tune CAR-T cells against solid tumors, Signal Transduct Target Ther, 4, 7, 10.1038/s41392-019-0041-1
Batlle, 2019, Transforming growth factor-beta signaling in immunity and cancer, Immunity, 50, 924, 10.1016/j.immuni.2019.03.024
Dahmani, 2018, TGF-beta in T cell biology: implications for cancer immunotherapy, Cancers, 10, 10.3390/cancers10060194
Ahmadzadeh, 2005, TGF-beta 1 attenuates the acquisition and expression of effector function by tumor antigen-specific human memory CD8 T cells, J Immunol, 174, 5215, 10.4049/jimmunol.174.9.5215
Gunderson, 2020, TGFbeta suppresses CD8+ T cell expression of CXCR3 and tumor trafficking, Nat Commun, 11, 1749, 10.1038/s41467-020-15404-8
Maurice, 2019, CXCR3 enables recruitment and site-specific bystander activation of memory CD8+ T cells, Nat Commun, 10, 4987, 10.1038/s41467-019-12980-2
Li, 2015, Impact of chemokine receptor CXCR3 on tumor-infiltrating lymphocyte recruitment associated with favorable prognosis in advanced gastric cancer, Int J Clin Exp Pathol, 8, 14725
Thomas, 2005, TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance, Cancer Cell, 8, 369, 10.1016/j.ccr.2005.10.012
Yang, 2014, TGF-beta upregulates CD70 expression and induces exhaustion of effector memory T cells in B-cell non-Hodgkin's lymphoma, Leukemia, 28, 1872, 10.1038/leu.2014.84
O'Neill, 2017, T cell-derived CD70 delivers an immune checkpoint function in inflammatory T cell responses, J Immunol, 199, 3700, 10.4049/jimmunol.1700380
Leigh, 2017, Host-derived CD70 suppresses murine graft-versus-host disease by limiting donor T cell expansion and effector function, J Immunol, 199, 336, 10.4049/jimmunol.1502181
Park, 2016, TGFbeta1-mediated SMAD3 enhances PD-1 expression on antigen-specific T cells in cancer, Cancer Discov, 6, 1366, 10.1158/2159-8290.CD-15-1347
Gabriel, 2021, Transforming growth factor-beta-regulated mTOR activity preserves cellular metabolism to maintain long-term T cell responses in chronic infection, Immunity, 54, 1698, 10.1016/j.immuni.2021.06.007
Tang, 2020, TGF-beta inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors, JCI Insight, 5, e133977, 10.1172/jci.insight.133977
Kloss, 2018, Dominant-negative TGF-beta receptor enhances PSMA-targeted human CAR T cell proliferation and augments prostate cancer eradication, Mol Ther, 26, 1855, 10.1016/j.ymthe.2018.05.003
Webster, 2021, Self-driving armored CAR-T cells overcome a suppressive milieu and eradicate CD19+ Raji lymphoma in preclinical models, Mol Ther, 29, 2691, 10.1016/j.ymthe.2021.05.006
Stuber, 2020, Inhibition of TGF-beta-receptor signaling augments the antitumor function of ROR1-specific CAR T-cells against triple-negative breast cancer, J Immunother Cancer, 8, e000676, 10.1136/jitc-2020-000676
Uhl, 2004, SD-208, a novel transforming growth factor beta receptor I kinase inhibitor, inhibits growth and invasiveness and enhances immunogenicity of murine and human glioma cells in vitro and in vivo, Cancer Res, 64, 7954, 10.1158/0008-5472.CAN-04-1013
Li, 2020, Arming anti-EGFRvIII CAR-T With TGFbeta trap improves antitumor efficacy in Glioma mouse models, Front Oncol, 10, 1117, 10.3389/fonc.2020.01117
Hou, 2018, TGF-beta-responsive CAR-T cells promote anti-tumor immune function, Bioeng Transl Med, 3, 75, 10.1002/btm2.10097
Hartley, 2019, Chimeric antigen receptors designed to overcome transforming growth factor-beta-mediated repression in the adoptive T-cell therapy of solid tumors, Clin Transl Immunol, 8, e1064, 10.1002/cti2.1064
Chang, 2018, Rewiring T-cell responses to soluble factors with chimeric antigen receptors, Nat Chem Biol, 14, 317, 10.1038/nchembio.2565
Pang, 2021, IL-7 and CCL19-secreting CAR-T cell therapy for tumors with positive glypican-3 or mesothelin, J Hematol Oncol, 14, 118, 10.1186/s13045-021-01128-9
Singha, 2019, Increased Smad3 and reduced Smad2 levels mediate the functional switch of TGF-beta from growth suppressor to growth and metastasis promoter through TMEPAI/PMEPA1 in triple negative breast cancer, Genes Cancer, 10, 134, 10.18632/genesandcancer.194
Wu, 2020, Discovery of a novel selective water-soluble SMAD3 inhibitor as an antitumor agent, Bioorg Med Chem Lett, 30, 10.1016/j.bmcl.2020.127396
Lutz-Nicoladoni, 2015, Modulation of Immune cell functions by the E3 ligase Cbl-b, Front Oncol, 5, 58, 10.3389/fonc.2015.00058
Kumar, 2021, Deletion of Cbl-b inhibits CD8+ T-cell exhaustion and promotes CAR T-cell function, J Immunother Cancer, 9, e001688, 10.1136/jitc-2020-001688
Sitaram, 2019, Beyond the cell surface: targeting intracellular negative regulators to enhance T cell anti-tumor activity, Int J Mol Sci, 20, 5821, 10.3390/ijms20235821
Chiang, 2007, Ablation of Cbl-b provides protection against transplanted and spontaneous tumors, J Clin Invest, 117, 1029, 10.1172/JCI29472
Fujiwara, 2017, Cbl-b deficiency mediates resistance to programmed death-ligand 1/programmed death-1 regulation, Front Immunol, 8, 42, 10.3389/fimmu.2017.00042
Peer, 2017, Cblb-deficient T cells are less susceptible to PD-L1-mediated inhibition, Oncotarget, 8, 41841, 10.18632/oncotarget.18360
Shah, 2019, Clonal expansion of CAR T cells harboring lentivector integration in the CBL gene following anti-CD22 CAR T-cell therapy, Blood Adv, 3, 2317, 10.1182/bloodadvances.2019000219
Loeser, 2007, Spontaneous tumor rejection by cbl-b-deficient CD8+ T cells, J Exp Med, 204, 879, 10.1084/jem.20061699
Zha, 2007, An adenoviral vector encoding dominant negative Cbl lowers the threshold for T cell activation in post-thymic T cells, Cell Immunol, 247, 95, 10.1016/j.cellimm.2007.07.006
Zhou, 2021, Targeting ubiquitin signaling for cancer immunotherapy, Signal Transduct Target Ther, 6, 16, 10.1038/s41392-020-00421-2
Jeon, 2004, Essential role of the E3 ubiquitin ligase Cbl-b in T cell anergy induction, Immunity, 21, 167, 10.1016/j.immuni.2004.07.013
Chiang, 2000, Cbl-b regulates the CD28 dependence of T-cell activation, Nature, 403, 216, 10.1038/35003235
Weber, 2021, Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling, Science, 372, 10.1126/science.aba1786
Richman, 2020, Ligand-induced degradation of a CAR permits reversible remote control of CAR T cell activity in vitro and in vivo, Mol Ther, 28, 1600, 10.1016/j.ymthe.2020.06.004
Juillerat, 2019, Modulation of chimeric antigen receptor surface expression by a small molecule switch, BMC Biotechnol, 19, 44, 10.1186/s12896-019-0537-3
Zhang, 2019, Doxycycline inducible chimeric antigen receptor T cells targeting CD147 for hepatocellular carcinoma therapy, Front Cell Dev Biol, 7, 233, 10.3389/fcell.2019.00233
Gu, 2018, Development of inducible CD19-CAR T cells with a Tet-on system for controlled activity and enhanced clinical safety, Int J Mol Sci, 19, 3455, 10.3390/ijms19113455
Drent, 2018, Feasibility of controlling CD38-CAR T cell activity with a Tet-on inducible CAR design, PLoS One, 13, 10.1371/journal.pone.0197349
Greenshpan, 2021, Synthetic promoters to induce immune-effectors into the tumor microenvironment, Commun Biol, 4, 143, 10.1038/s42003-021-01664-7
Eyquem, 2017, Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection, Nature, 543, 113, 10.1038/nature21405
Brandt, 2020, Emerging approaches for regulation and control of CAR T cells: a mini review, Front Immunol, 11, 326, 10.3389/fimmu.2020.00326
Liu, 2020, Split chimeric antigen receptor-modified T cells targeting glypican-3 suppress hepatocellular carcinoma growth with reduced cytokine release, Ther Adv Med Oncol, 12, 10.1177/1758835920910347
Leung, 2019, Sensitive and adaptable pharmacological control of CAR T cells through extracellular receptor dimerization, JCI Insight, 5, e124430, 10.1172/jci.insight.124430
Viaud, 2018, Switchable control over in vivo CAR T expansion, B cell depletion, and induction of memory, Proc Natl Acad Sci USA, 115, E10898, 10.1073/pnas.1810060115
Zajc, 2020, A conformation-specific ON-switch for controlling CAR T cells with an orally available drug, Proc Natl Acad Sci USA, 117, 14926, 10.1073/pnas.1911154117
Lee, 2019, Regulation of CAR T cell-mediated cytokine release syndrome-like toxicity using low molecular weight adapters, Nat Commun, 10, 2681, 10.1038/s41467-019-10565-7
Wu, 2015, Remote control of therapeutic T cells through a small molecule-gated chimeric receptor, Science, 350, aab4077, 10.1126/science.aab4077
Mata, 2017, Inducible activation of MyD88 and CD40 in CAR T cells results in controllable and potent antitumor activity in preclinical solid tumor models, Cancer Discov, 7, 1306, 10.1158/2159-8290.CD-17-0263
Poorebrahim, 2021, Counteracting CAR T cell dysfunction, Oncogene, 40, 421, 10.1038/s41388-020-01501-x
Catakovic, 2017, T cell exhaustion: from pathophysiological basics to tumor immunotherapy, Cell Commun Signal, 15, 1, 10.1186/s12964-016-0160-z
Du, 2018, Blockade of tumor-expressed PD-1 promotes lung cancer growth, Oncoimmunology, 7, 10.1080/2162402X.2017.1408747
McGowan, 2020, PD-1 disrupted CAR-T cells in the treatment of solid tumors: promises and challenges, Biomed Pharmacother, 121, 10.1016/j.biopha.2019.109625
Wang, 2020, Tumor cell-intrinsic PD-1 receptor is a tumor suppressor and mediates resistance to PD-1 blockade therapy, Proc Natl Acad Sci USA, 117, 6640, 10.1073/pnas.1921445117
Diaconu, 2017, Inducible caspase-9 selectively modulates the toxicities of CD19-specific chimeric antigen receptor-modified T cells, Mol Ther, 25, 580, 10.1016/j.ymthe.2017.01.011
Zhou, 2015, Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation, Blood, 125, 4103, 10.1182/blood-2015-02-628354
Lynn, 2019, c-Jun overexpression in CAR T cells induces exhaustion resistance, Nature, 576, 293, 10.1038/s41586-019-1805-z
Heitzeneder, 2022, GPC2-CAR T cells tuned for low antigen density mediate potent activity against neuroblastoma without toxicity, Cancer Cell, 40, 53, 10.1016/j.ccell.2021.12.005
Yeku, 2016, Armored CAR T-cells: utilizing cytokines and pro-inflammatory ligands to enhance CAR T-cell anti-tumour efficacy, Biochem Soc Trans, 44, 412, 10.1042/BST20150291
Yeku, 2017, Armored CAR T cells enhance antitumor efficacy and overcome the tumor microenvironment, Sci Rep, 7, 10541, 10.1038/s41598-017-10940-8
Zhao, 2021, Human hyaluronidase PH20 potentiates the antitumor activities of mesothelin-specific CAR-T cells against gastric cancer, Front Immunol, 12