Ehrlich, P. Ueber den jetzigen Stand der Karzinomforschung. Ned. Tijdschr. Genees. 53, 273–290 (1908).
Thomas, L. Mobile and Humoral Points of the Hypersenstive States (ed. Lawrence, H.) 529–532 (Hoeber-Harper, 1959).
Shankaran, V. et al. IFNγ and lymphocytes stop main tumour growth and form tumour immunogenicity. Nature 410, 1107–1111 (2001).
Zehn, D. & Bevan, M. J. T cells with low avidity for a tissue-restricted antigen routinely evade central and peripheral tolerance and trigger autoimmunity. Immunity 25, 261–270 (2006).
Brunet, J. F. et al. A brand new member of the immunoglobulin superfamily—CTLA-4. Nature 328, 267–270 (1987).
Krummel, M. F. & Allison, J. P. CD28 and CTLA-4 have opposing results on the response of T cells to stimulation. J. Exp. Med. 182, 459–465 (1995).
Wing, Ok. et al. CTLA-4 management over Foxp3+ regulatory T cell operate. Science 322, 271–275 (2008).
Zappasodi, R. et al. CTLA-4 blockade drives lack of Treg stability in glycolysis-low tumours. Nature 591, 652–658 (2021).
Ishida, Y., Agata, Y., Shibahara, Ok. & Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell loss of life. EMBO J. 11, 3887–3895 (1992).
Dong, H., Zhu, G., Tamada, Ok. & Chen, L. B7-H1, a 3rd member of the B7 household, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 5, 1365–1369 (1999).
Freeman, G. J. et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 member of the family results in unfavorable regulation of lymphocyte activation. J. Exp. Med. 192, 1027–1034 (2000).
Larkin, J. et al. Mixed nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).
Woo, S. R. et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell operate to advertise tumoral immune escape. Most cancers Res. 72, 917–927 (2012).
Anderson, A. C., Joller, N. & Kuchroo, V. Ok. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialised features in immune regulation. Immunity 44, 989–1004 (2016).
Tawbi, H. A. et al. Relatlimab and nivolumab versus nivolumab in untreated superior melanoma. N. Engl. J. Med. 386, 24–34 (2022).
Muul, L. M., Spiess, P. J., Director, E. P. & Rosenberg, S. A. Identification of particular cytolytic immune responses in opposition to autologous tumor in people bearing malignant melanoma. J. Immunol. 138, 989–995 (1987).
Mullard, A. FDA approves first tumour-infiltrating lymphocyte (TIL) remedy, bolstering hopes for cell therapies in strong cancers. Nat. Rev. Drug Discov. 23, 238 (2024).
Robbins, P. F. et al. A pilot trial utilizing lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin. Most cancers Res. 21, 1019–1027 (2015).
Imai, C. et al. Chimeric receptors with 4-1BB signaling capability provoke potent cytotoxicity in opposition to acute lymphoblastic leukemia. Leukemia 18, 676–684 (2004).
Krause, A. et al. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human main T lymphocytes. J. Exp. Med. 188, 619–626 (1998).
Baker, D. J., Arany, Z., Baur, J. A., Epstein, J. A., & June, C. H. CAR T remedy past most cancers: the evolution of a residing drug. Nature 619, 707–715 (2023).
June, C. H. & Sadelain, M. Chimeric antigen receptor remedy. N. Engl. J. Med. 379, 64–73 (2018).
Park, J. H. et al. Lengthy-term follow-up of CD19 CAR remedy in acute lymphoblastic leukemia. N. Engl. J. Med. 378, 449–459 (2018).
Maude, S. L. et al. Tisagenlecleucel in youngsters and younger adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 378, 439–448 (2018).
Rodriguez-Otero, P. et al. Ide-cel or normal regimens in relapsed and refractory a number of myeloma. N. Engl. J. Med. 388, 1002–1014 (2023).
Zebley, C. C. et al. CD19-CAR T cells endure exhaustion DNA methylation programming in sufferers with acute lymphoblastic leukemia. Cell Rep. 37, 110079 (2021).
Cappell, Ok. M. & Kochenderfer, J. N. Lengthy-term outcomes following CAR T cell remedy: what we all know thus far. Nat. Rev. Clin. Oncol. 20, 359–371 (2023).
Albelda, S. M. CAR T cell remedy for sufferers with strong tumours: key classes to study and unlearn. Nat. Rev. Clin. Oncol. 21, 47–66 (2024).
Prinzing, B. et al. Deleting DNMT3A in CAR T cells prevents exhaustion and enhances antitumor exercise. Sci. Transl. Med. 13, eabh0272 (2021).
Bell, M. et al. Modular chimeric cytokine receptors with leucine zippers improve the antitumour exercise of CAR T cells through JAK/STAT signalling. Nat. Biomed. Eng. 8, 380–396 (2023).
Wei, J. et al. Focusing on REGNASE-1 packages long-lived effector T cells for most cancers remedy. Nature 576, 471–476 (2019).
Repair, S. M., Jazaeri, A. A. & Hwu, P. Purposes of CRISPR genome enhancing to advance the following technology of adoptive cell therapies for most cancers. Most cancers Discov. 11, 560–574 (2021).
Lin, M. J. et al. Most cancers vaccines: the following immunotherapy frontier. Nat. Most cancers 3, 911–926 (2022).
Sellars, M. C., Wu, C. J. & Fritsch, E. F. Most cancers vaccines: constructing a bridge over troubled waters. Cell 185, 2770–2788 (2022).
Zajac, A. J. et al. Viral immune evasion because of persistence of activated T cells with out effector operate. J. Exp. Med. 188, 2205–2213 (1998).
Gallimore, A. et al. Induction and exhaustion of lymphocytic choriomeningitis virus-specific cytotoxic T lymphocytes visualized utilizing soluble tetrameric main histocompatibility complicated class I–peptide complexes. J. Exp. Med. 187, 1383–1393 (1998).
Moskophidis, D., Lechner, F., Pircher, H. & Zinkernagel, R. M. Virus persistence in acutely contaminated immunocompetent mice by exhaustion of antiviral cytotoxic effector T cells. Nature 362, 758–761 (1993).
Wherry, E. J., Blattman, J. N., Murali-Krishna, Ok., van der Most, R. & Ahmed, R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and ends in distinct phases of useful impairment. J. Virol. 77, 4911–4927 (2003).
Barber, D. L. et al. Restoring operate in exhausted CD8 T cells throughout continual viral an infection. Nature 439, 682–687 (2006).
Pantaleo, G. & Koup, R. A. Correlates of immune safety in HIV-1 an infection: what we all know, what we don’t know, what we must always know. Nat. Med. 10, 806–810 (2004).
Letvin, N. L. & Walker, B. D. Immunopathogenesis and immunotherapy in AIDS virus infections. Nat. Med. 9, 861–866 (2003).
Rehermann, B. & Nascimbeni, M. Immunology of hepatitis B virus and hepatitis C virus an infection. Nat. Rev. Immunol. 5, 215–229 (2005).
Day, C. L. et al. PD-1 expression on HIV-specific T cells is related to T-cell exhaustion and illness development. Nature 443, 350–354 (2006).
Wherry, E. J. et al. Molecular signature of CD8+ T cell exhaustion throughout continual viral an infection. Immunity 27, 670–684 (2007).
Wherry, E. J. T cell exhaustion. Nat. Immunol. 12, 492–499 (2011).
Zehn, D., Thimme, R., Lugli, E., de Almeida, G. P. & Oxenius, A. ‘Stem-like’ precursors are the fount to maintain persistent CD8+ T cell responses. Nat. Immunol. 23, 836–847 (2022).
Kamphorst, A. O. et al. Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 355, 1423–1427 (2017).
Hui, E. et al. T cell costimulatory receptor CD28 is a main goal for PD-1-mediated inhibition. Science 355, 1428–1433 (2017).
Youngblood, B. et al. Power virus an infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8+ T cells. Immunity 35, 400–412 (2011).
Ghoneim, H. E. et al. De novo epigenetic packages inhibit PD-1 blockade-mediated T cell rejuvenation. Cell 170, 142–157 (2017).
Sen, D. R. et al. The epigenetic panorama of T cell exhaustion. Science 354, 1165–1169 (2016).
Pauken, Ok. E. et al. Epigenetic stability of exhausted T cells limits sturdiness of reinvigoration by PD-1 blockade. Science 354, 1160–1165 (2016).
Philip, M. et al. Chromatin states outline tumour-specific T cell dysfunction and reprogramming. Nature 545, 452–456 (2017).
Tempo, L. et al. The epigenetic management of stemness in CD8+ T cell destiny dedication. Science 359, 177–186 (2018).
Grey, S. M., Amezquita, R. A., Guan, T., Kleinstein, S. H. & Kaech, S. M. Polycomb repressive complicated 2-mediated chromatin repression guides effector CD8+ T cell terminal differentiation and lack of multipotency. Immunity 46, 596–608 (2017).
Schietinger, A. et al. Tumor-specific T cell dysfunction is a dynamic antigen-driven differentiation program initiated early throughout tumorigenesis. Immunity 45, 389–401 (2016).
Sade-Feldman, M. et al. Defining T cell states related to response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013 (2018).
Pelka, Ok. et al. Spatially organized multicellular immune hubs in human colorectal most cancers. Cell 184, 4734–4752 (2021).
Simoni, Y. et al. Bystander CD8+ T cells are plentiful and phenotypically distinct in human tumour infiltrates. Nature 557, 575–579 (2018).
Hoekstra, M. E. et al. Lengthy-distance modulation of bystander tumor cells by CD8+ T cell-secreted IFNγ. Nat. Most cancers 1, 291–301 (2020).
Meier, S. L., Satpathy, A. T. & Wells, D. Ok. Bystander T cells in most cancers immunology and remedy. Nat. Most cancers 3, 143–155 (2022).
Im, S. J. et al. Defining CD8+ T cells that present the proliferative burst after PD-1 remedy. Nature 537, 417–421 (2016).
Utzschneider, D. T. et al. T cell issue 1-expressing memory-like CD8+ T cells maintain the immune response to continual viral infections. Immunity 45, 415–427 (2016).
Siddiqui, I. et al. Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor management in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211 (2019).
Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor management and reply to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).
Alfei, F. et al. TOX reinforces the phenotype and longevity of exhausted T cells in continual viral an infection. Nature 571, 265–269 (2019).
Khan, O. et al. TOX transcriptionally and epigenetically packages CD8+ T cell exhaustion. Nature 571, 211–218 (2019).
Yao, C. et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in continual an infection. Nat. Immunol. 20, 890–901 (2019).
Huang, Q. et al. The primordial differentiation of tumor-specific reminiscence CD8+ T cells as bona fide responders to PD-1/PD-L1 blockade in draining lymph nodes. Cell 185, 4049–4066 (2022).
Scott, A. C. et al. TOX is a essential regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019).
Search engine optimization, H. et al. TOX and TOX2 transcription elements cooperate with NR4A transcription elements to impose CD8+ T cell exhaustion. Proc. Natl Acad. Sci. USA 116, 12410–12415 (2019).
Tsui, C. et al. MYB orchestrates T cell exhaustion and response to checkpoint inhibition. Nature 609, 354–360 (2022).
Im, S. J. et al. Traits and anatomic location of PD-1+TCF1+ stem-like CD8 T cells in continual viral an infection and most cancers. Proc. Natl Acad. Sci. USA 120, e2221985120 (2023).
Chen, J. H. et al. Human lung most cancers harbors spatially organized stem-immunity hubs related to response to immunotherapy. Nat. Immunol. 25, 644–658 (2024).
Hudson, W. H. et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1+ stem-like CD8+ T cells throughout continual an infection. Immunity 51, 1043–1058 (2019).
Beltra, J. C. et al. Developmental relationships of 4 exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic panorama management mechanisms. Immunity 52, 825–841 (2020).
Rahim, M. Ok. et al. Dynamic CD8+ T cell responses to most cancers immunotherapy in human regional lymph nodes are disrupted in metastatic lymph nodes. Cell 186, 1127–1143 (2023).
Eberhardt, C. S. et al. Useful HPV-specific PD-1+ stem-like CD8 T cells in head and neck most cancers. Nature 597, 279–284 (2021).
Clean, C. U. et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 19, 665–674 (2019).
Krishna, S. et al. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy in opposition to human most cancers. Science 370, 1328–1334 (2020).
Jansen, C. S. et al. An intra-tumoral area of interest maintains and differentiates stem-like CD8 T cells. Nature 576, 465–470 (2019).
Vignali, P. D. A. et al. Hypoxia drives CD39-dependent suppressor operate in exhausted T cells to restrict antitumor immunity. Nat. Immunol. 24, 267–279 (2023).
Pfannenstiel, L. W. et al. Immune-checkpoint blockade opposes CD8+ T-cell suppression in human and murine most cancers. Most cancers Immunol. Res. 7, 510–525 (2019).
Matsushita, H. et al. Most cancers exome evaluation reveals a T-cell-dependent mechanism of most cancers immunoediting. Nature 482, 400–404 (2012).
Anagnostou, V. et al. Evolution of neoantigen panorama throughout immune checkpoint blockade in non-small cell lung most cancers. Most cancers Discov. 7, 264–276 (2017).
Chapman, N. M., Boothby, M. R. & Chi, H. Metabolic coordination of T cell quiescence and activation. Nat. Rev. Immunol. 20, 55–70 (2020).
Shi, L. Z. et al. HIF1α-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J. Exp. Med. 208, 1367–1376 (2011).
Michalek, R. D. et al. Leading edge: distinct glycolytic and lipid oxidative metabolic packages are important for effector and regulatory CD4+ T cell subsets. J. Immunol. 186, 3299–3303 (2011).
Yang, Ok. et al. T cell exit from quiescence and differentiation into TH2 cells rely upon Raptor–mTORC1-mediated metabolic reprogramming. Immunity 39, 1043–1056 (2013).
Chapman, N. M. & Chi, H. Metabolic adaptation of lymphocytes in immunity and illness. Immunity 55, 14–30 (2022).
Vardhana, S. A. et al. Impaired mitochondrial oxidative phosphorylation limits the self-renewal of T cells uncovered to persistent antigen. Nat. Immunol. 21, 1022–1033 (2020).
Yu, Y. R. et al. Disturbed mitochondrial dynamics in CD8+ TILs reinforce T cell exhaustion. Nat. Immunol. 21, 1540–1551 (2020).
Scharping, N. E. et al. Mitochondrial stress induced by steady stimulation underneath hypoxia quickly drives T cell exhaustion. Nat. Immunol. 22, 205–215 (2021).
Siska, P. J. et al. Mitochondrial dysregulation and glycolytic insufficiency functionally impair CD8 T cells infiltrating human renal cell carcinoma. JCI Perception 2, e93411 (2017).
Scharping, N. E. et al. The tumor microenvironment represses T cell mitochondrial biogenesis to drive intratumoral T cell metabolic insufficiency and dysfunction. Immunity 45, 374–388 (2016).
Chen, Y. et al. BATF regulates progenitor to cytolytic effector CD8+ T cell transition throughout continual viral an infection. Nat. Immunol. 22, 996–1007 (2021).
Search engine optimization, H. et al. BATF and IRF4 cooperate to counter exhaustion in tumor-infiltrating CAR T cells. Nat. Immunol. 22, 983–995 (2021).
Wenes, M. et al. The mitochondrial pyruvate provider regulates reminiscence T cell differentiation and antitumor operate. Cell Metab. 34, 731–746 (2022).
Si, X. et al. Mitochondrial isocitrate dehydrogenase impedes CAR T cell operate by restraining antioxidant metabolism and histone acetylation. Cell Metab. 36, 176–192 (2024).
Leone, R. D. et al. Glutamine blockade induces divergent metabolic packages to beat tumor immune evasion. Science 366, 1013–1021 (2019).
Arner, E. N. & Rathmell, J. C. Metabolic programming and immune suppression within the tumor microenvironment. Most cancers Cell 41, 421–433 (2023).
Ho, P. C. et al. Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses. Cell 162, 1217–1228 (2015).
Chang, C. H. et al. Metabolic competitors within the tumor microenvironment is a driver of most cancers development. Cell 162, 1229–1241 (2015).
Reinfeld, B. I. et al. Cell-programmed nutrient partitioning within the tumour microenvironment. Nature 593, 282–288 (2021).
Guo, C. et al. SLC38A2 and glutamine signalling in cDC1s dictate anti-tumour immunity. Nature 620, 200–208 (2023).
Geiger, R. et al. l-arginine modulates T cell metabolism and enhances survival and anti-tumor exercise. Cell 167, 829–842 (2016).
Bian, Y. et al. Most cancers SLC43A2 alters T cell methionine metabolism and histone methylation. Nature 585, 277–282 (2020).
Rowe, J. H. et al. Formate supplementation enhances antitumor CD8+ T-cell health and efficacy of PD-1 blockade. Most cancers Discov. 13, 2566–2583 (2023).
Lengthy, L. et al. CRISPR screens unveil sign hubs for nutrient licensing of T cell immunity. Nature 600, 308–313 (2021).
Raynor, J. L. & Chi, H. Vitamins: sign 4 in T cell immunity. J. Exp. Med. 221, e20221839 (2024).
Mager, L. F. et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science 369, 1481–1489 (2020).
Wang, T. et al. Inosine is an alternate carbon supply for CD8+-T-cell operate underneath glucose restriction. Nat. Metab. 2, 635–647 (2020).
Klysz, D. D. et al. Inosine induces stemness options in CAR-T cells and enhances efficiency. Most cancers Cell 42, 266–282 (2024).
Fan, H. et al. Trans-vaccenic acid reprograms CD8+ T cells and anti-tumour immunity. Nature 623, 1034–1043 (2023).
Ringel, A. E. et al. Weight problems shapes metabolism within the tumor microenvironment to suppress anti-tumor immunity. Cell 183, 1848–1866 (2020).
Zani, F. et al. The dietary sweetener sucralose is a unfavorable modulator of T cell-mediated responses. Nature 615, 705–711 (2023).
Model, A. et al. LDHA-associated lactic acid manufacturing blunts tumor immunosurveillance by T and NK cells. Cell Metab. 24, 657–671 (2016).
Ma, X. et al. Ldl cholesterol induces CD8+ T cell exhaustion within the tumor microenvironment. Cell Metab. 30, 143–156 (2019).
Xu, S. et al. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8+ T cells in tumors. Immunity 54, 1561–1577 (2021).
Ma, X. et al. CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector operate and impairs their antitumor potential. Cell Metab. 33, 1001–1012 (2021).
Guo, Y. et al. Metabolic reprogramming of terminally exhausted CD8+ T cells by IL-10 enhances anti-tumor immunity. Nat. Immunol. 22, 746–756 (2021).
Xu, Ok. et al. Glycolysis fuels phosphoinositide 3-kinase signaling to bolster T cell immunity. Science 371, 405–410 (2021).
Notarangelo, G. et al. Oncometabolite d-2HG alters T cell metabolism to impair CD8+ T cell operate. Science 377, 1519–1529 (2022).
Bunse, L. et al. Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate. Nat. Med. 24, 1192–1203 (2018).
Hicks, Ok. G. et al. Protein–metabolite interactomics of carbohydrate metabolism reveal regulation of lactate dehydrogenase. Science 379, 996–1003 (2023).
Kawalekar, O. U. et al. Distinct signaling of coreceptors regulates particular metabolism pathways and impacts reminiscence growth in CAR T cells. Immunity 44, 380–390 (2016).
Klein Geltink, R. I. et al. Metabolic conditioning of CD8+ effector T cells for adoptive cell remedy. Nat. Metab. 2, 703–716 (2020).
Uhl, F. M. et al. Metabolic reprogramming of donor T cells enhances graft-versus-leukemia results in mice and people. Sci. Transl. Med. 12, eabb8969 (2020).
Jaccard, A. et al. Reductive carboxylation epigenetically instructs T cell differentiation. Nature 621, 849–856 (2023).
Huang, H. et al. In vivo CRISPR screening reveals nutrient signaling processes underpinning CD8+ T cell destiny choices. Cell 184, 1245–1261 (2021).
Ye, L. et al. A genome-scale gain-of-function CRISPR display in CD8 T cells identifies proline metabolism as a way to reinforce CAR-T remedy. Cell Metab. 34, 595–614 (2022).
Canale, F. P. et al. Metabolic modulation of tumours with engineered micro organism for immunotherapy. Nature 598, 662–666 (2021).
Shi, H., Chen, S. & Chi, H. Immunometabolism of CD8+ T cell differentiation in most cancers. Developments Most cancers https://doi.org/10.1016/j.trecan.2024.03.010 (2024).
Zhou, P. et al. Single-cell CRISPR screens in vivo map T cell destiny regulomes in most cancers. Nature 624, 154–163 (2023).
Dahling, S. et al. Kind 1 typical dendritic cells keep and information the differentiation of precursors of exhausted T cells in distinct mobile niches. Immunity 55, 656–670 (2022).
Schenkel, J. M. et al. Typical sort I dendritic cells keep a reservoir of proliferative tumor-antigen particular TCF-1+ CD8+ T cells in tumor-draining lymph nodes. Immunity 54, 2338–2353 (2021).
Kanev, Ok. et al. Proliferation-competent Tcf1+ CD8 T cells in dysfunctional populations are CD4 T cell assist unbiased. Proc. Natl Acad. Sci. USA 116, 20070–20076 (2019).
Zander, R. et al. CD4+ T cell assistance is required for the formation of a cytolytic CD8+ T cell subset that protects in opposition to continual an infection and most cancers. Immunity 51, 1028–1042 (2019).
Yu, P. et al. Intratumor depletion of CD4+ cells unmasks tumor immunogenicity resulting in the rejection of late-stage tumors. J. Exp. Med. 201, 779–791 (2005).
Jing, W., Gershan, J. A. & Johnson, B. D. Depletion of CD4 T cells enhances immunotherapy for neuroblastoma after syngeneic HSCT however compromises growth of antitumor immune reminiscence. Blood 113, 4449–4457 (2009).
Kim, S. H. et al. Adoptive immunotherapy with transient anti-CD4 therapy enhances anti-tumor response by rising IL-18Rαhello CD8+ T cells. Nat. Commun. 12, 5314 (2021).
Kreiter, S. et al. Mutant MHC class II epitopes drive therapeutic immune responses to most cancers. Nature 520, 692–696 (2015).
Tay, R. E., Richardson, E. Ok. & Toh, H. C. Revisiting the function of CD4+ T cells in most cancers immunotherapy—new insights into outdated paradigms. Most cancers Gene Ther. 28, 5–17 (2021).
Yi, J. S., Du, M. & Zajac, A. J. An important function for interleukin-21 within the management of a continual viral an infection. Science 324, 1572–1576 (2009).
Elsaesser, H., Sauer, Ok. & Brooks, D. G. IL-21 is required to regulate continual viral an infection. Science 324, 1569–1572 (2009).
Frohlich, A. et al. IL-21R on T cells is essential for sustained performance and management of continual viral an infection. Science 324, 1576–1580 (2009).
Hashimoto, M. et al. PD-1 mixture remedy with IL-2 modifies CD8+ T cell exhaustion program. Nature 610, 173–181 (2022).
Deng, Q. et al. Traits of anti-CD19 CAR T cell infusion merchandise related to efficacy and toxicity in sufferers with giant B cell lymphomas. Nat. Med. 26, 1878–1887 (2020).
Chen, G. M. et al. Integrative bulk and single-cell profiling of premanufacture T-cell populations reveals elements mediating long-term persistence of CAR T-cell remedy. Most cancers Discov. 11, 2186–2199 (2021).
Bell, M. & Gottschalk, S. Engineered cytokine signaling to enhance CAR T cell effector operate. Entrance. Immunol. 12, 684642 (2021).
Hurton, L. V. et al. Tethered IL-15 augments antitumor exercise and promotes a stem-cell reminiscence subset in tumor-specific T cells. Proc. Natl Acad. Sci. USA 113, E7788–E7797 (2016).
Hinrichs, C. S. et al. IL-2 and IL-21 confer opposing differentiation packages to CD8+ T cells for adoptive immunotherapy. Blood 111, 5326–5333 (2008).
Prinzing, B. et al. MyD88/CD40 signaling retains CAR T cells in a much less differentiated state. JCI Perception 5, e136093 (2020).
Lynn, R. C. et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 576, 293–300 (2019).
Doan, A. E. et al. FOXO1 is a grasp regulator of reminiscence programming in CAR T cells. Nature 629, 211–218 (2024).
Chan, J. D. et al. FOXO1 enhances CAR T cell stemness, metabolic health and efficacy. Nature 629, 201–210 (2024).
Zhu, Z. et al. FOXP1 and KLF2 reciprocally regulate checkpoints of stem-like to effector transition in CAR T cells. Nat. Immunol. 25, 117–128 (2024).
Carnevale, J. et al. RASA2 ablation in T cells boosts antigen sensitivity and long-term operate. Nature 609, 174–182 (2022).
Jain, N. et al. Lack of TET2 uncouples proliferative and effector features in CAR T cells. Blood https://doi.org/10.1182/blood-2020-142957 (2020).
Jain, N. et al. TET2 guards in opposition to unchecked BATF3-induced CAR T cell growth. Nature 615, 315–322 (2023).
Jain, N. et al. Disruption of SUV39H1-mediated H3K9 methylation sustains CAR T cell operate. Most cancers Discov. 14, 142–157 (2023).
Harrison, S. J. et al. CAR+ T-cell lymphoma put up ciltacabtagene autoleucel remedy for relapsed refractory a number of myeloma. Blood 142, 6939 (2023).
Micklethwaite, Ok. P. et al. Investigation of product-derived lymphoma following infusion of piggyBac-modified CD19 chimeric antigen receptor T cells. Blood 138, 1391–1405 (2021).
Chen, D. S. & Mellman, I. Oncology meets immunology: the cancer-immunity cycle. Immunity 39, 1–10 (2013).
Mellman, I., Chen, D. S., Powles, T. & Turley, S. J. The cancer-immunity cycle: indication, genotype, and immunotype. Immunity 56, 2188–2205 (2023).
Soerens, A. G. et al. Useful T cells are able to supernumerary cell division and longevity. Nature 614, 762–766 (2023).

