Mlecnik, B. et al. Functional network pipeline reveals genetic determinants associated with in situ lymphocyte proliferation and survival of cancer patients. Sci. Transl. Med. 6, 228ra237 (2014).
Pilipow, K. et al. IL15 and T-cell stemness in T-cell-based cancer immunotherapy. Cancer Res. 75, 5187â5193 (2015).
Brentjens, R. J. et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat. Med. 9, 279â286 (2003).
Hoyos, V. et al. Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia 24, 1160â1170 (2010).
Chan, E. S. et al. Immunohistochemical expression of glypican-3 in pediatric tumors: an analysis of 414 cases. Pediatr. Dev. Pathol. 16, 272â277 (2013).
Haruyama, Y. & Kataoka, H. Glypican-3 is a prognostic factor and an immunotherapeutic target in hepatocellular carcinoma. World J. Gastroenterol. 22, 275â283 (2016).
Tretiakova, M. et al. Glypican 3 overexpression in primary and metastatic Wilms tumors. Virchows Arch. 466, 67â76 (2015).
Kohashi, K. et al. Glypican 3 expression in tumors with loss of SMARCB1/INI1 protein expression. Hum. Pathol. 44, 526â533 (2013).
Zynger, D. L., Dimov, N. D., Luan, C., Teh, B. T. & Yang, X. J. Glypican 3: a novel marker in testicular germ cell tumors. Am. J. Surg. Pathol. 30, 1570â1575 (2006).
Ho, M. & Kim, H. Glypican-3: a new target for cancer immunotherapy. Eur. J. Cancer 47, 333â338 (2011).
Maude, S. L. et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 378, 439â448 (2018).
Park, J. H. et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 378, 449â459 (2018).
Yong, C. S. M. et al. CAR T-cell therapy of solid tumors. Immunol. Cell Biol. 95, 356â363 (2017).
Chen, Y. et al. Eradication of neuroblastoma by T cells redirected with an optimized GD2-specific chimeric antigen receptor and interleukin-15. Clin. Cancer Res. 25, 2915â2924 (2019).
Batra, S. A. et al. Glypican-3-specific CAR T cells coexpressing IL15 and IL21 have superior expansion and antitumor activity against hepatocellular carcinoma. Cancer Immunol. Res. 8, 309â320 (2020).
Ishiguro, T. et al. Anti-glypican 3 antibody as a potential antitumor agent for human liver cancer. Cancer Res. 68, 9832â9838 (2008).
Shi, D. et al. Chimeric antigen receptor-glypican-3 T-cell therapy for advanced hepatocellular carcinoma: results of Phase I trials. Clin. Cancer Res. 26, 3979â3989 (2020).
Sawada, Y. et al. Phase II study of the GPC3-derived peptide vaccine as an adjuvant therapy for hepatocellular carcinoma patients. Oncoimmunology 5, e1129483 (2016).
Bosse, K. R. et al. Identification of GPC2 as an oncoprotein and candidate immunotherapeutic target in high-risk neuroblastoma. Cancer Cell 32, 295â309 (2017).
Zhu, A. X. et al. First-in-man phase I study of GC33, a novel recombinant humanized antibody against glypican-3, in patients with advanced hepatocellular carcinoma. Clin. Cancer Res. 19, 920â928 (2013).
Di Stasi, A. et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 365, 1673â1683 (2011).
Eisenhauer, E. A. et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228â247 (2009).
Deng, Q. et al. Characteristics of anti-CD19 CAR T cell infusion products associated with efficacy and toxicity in patients with large B cell lymphomas. Nat. Med. 26, 1878â1887 (2020).
Gardner, R. et al. Starting T cell and cell product phenotype are associated with durable remission of leukemia following CD19 CAR-T cell immunotherapy. Blood 132, 4022â4022 (2018).
Krishna, S. et al. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science 370, 1328â1334 (2020).
Rossi, J. et al. Preinfusion polyfunctional anti-CD19 chimeric antigen receptor T cells are associated with clinical outcomes in NHL. Blood 132, 804â814 (2018).
Good, C. R. et al. An NK-like CAR T cell transition in CAR T cell dysfunction. Cell 184, 6081â6100 (2021).
Seo, H. et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8(+) T cell exhaustion. Proc. Natl Acad. Sci. USA 116, 12410â12415 (2019).
Murphy, K., Travers, P., Walport, M. & Janeway, C. Janewayâs Immunobiology (Garland Science, 2012).
Mahuron, K. M. et al. Layilin augments integrin activation to promote antitumor immunity. J. Exp. Med. 217, e20192080 (2020).
Wang, C., Lin, G. H., McPherson, A. J. & Watts, T. H. Immune regulation by 4-1BB and 4-1BBL: complexities and challenges. Immunol. Rev. 229, 192â215 (2009).
Murphy, K. M. & Weaver, C. Janewayâs Immunobiology: Tenth International Student Edition with Registration Card (W.W. Norton, 2022).
Belk, J. A. et al. Genome-wide CRISPR screens of T cell exhaustion identify chromatin remodeling factors that limit T cell persistence. Cancer Cell 40, 768â786 (2022).
Upadhye, A. et al. Intra-tumoral T cells in pediatric brain tumors display clonal expansion and effector properties. Nat. Cancer 5, 791â807 (2024).
Del Bufalo, F. et al. GD2-CART01 for relapsed or refractory high-risk neuroblastoma. N. Engl. J. Med. 388, 1284â1295 (2023).
Majzner, R. G. et al. GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas. Nature 603, 934â941 (2022).
Conlon, K. C. et al. Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. J. Clin. Oncol. 33, 74â82 (2015).
Straathof, K. C. et al. An inducible caspase 9 safety switch for T-cell therapy. Blood 105, 4247â4254 (2005).
Fehniger, T. A. et al. Fatal leukemia in interleukin 15 transgenic mice follows early expansions in natural killer and memory phenotype CD8+ T cells. J. Exp. Med. 193, 219â231 (2001).
Xu, Y. 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. Blood 123, 3750â3759 (2014).
Lynn, R. C. et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 576, 293â300 (2019).
Zhao, Z. et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell 28, 415â428 (2015).
Jung, I.-Y. et al. Type I interferon signaling via the EGR2 transcriptional regulator potentiates CAR T cell-intrinsic dysfunction. Cancer Discov. 13, 1636â1655 (2023).
Lukhele, S. et al. The transcription factor IRF2 drives interferon-mediated CD8(+) T cell exhaustion to restrict anti-tumor immunity. Immunity 55, 2369â2385 (2022).
Li, W. et al. Redirecting T cells to glypican-3 with 4-1BB zeta chimeric antigen receptors results in Th1 polarization and potent antitumor activity. Hum. Gene Ther. 28, 437â448 (2017).
Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289â1296 (2019).
Borcherding, N., Bormann, N. L. & Kraus, G. scRepertoire: an R-based toolkit for single-cell immune receptor analysis. F1000Res. 9, 47 (2020).
Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417â425 (2015).