We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

BATF-mediated regulation of exhausted CD8+ T-cell responses and potential implications for chimeric antigen receptor-T therapy

    Chao Sun

    Liver Transplant Center, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China

    ,
    Dan Li

    Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China

    &
    Zhengxin Wang

    *Author for correspondence:

    E-mail Address: wangzhengxin@huashan.org.cn

    Liver Transplant Center, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China

    Published Online:https://doi.org/10.2217/imt-2023-0170

    Chimeric antigen receptor (CAR) T-cell therapy for malignant tumors has reached a crucial stage, with recent studies underscoring the role of T-cell exhaustion in determining the efficacy of CAR-T therapy. This trailblazing discovery has opened new avenues to augment the potency of CAR-T therapy. Basic leucine zipper ATF-like transcription factor (BATF) is indispensable in alleviating T-cell exhaustion and is pivotal in the early stages of CD8+ T-cell differentiation. In cooperation with other transcription factors, it plays a key role in the differentiation and maturation processes of exhausted T cells. A deeper comprehension of BATF‘s mechanisms in T-cell biology may yield novel insights into amplifying the efficacy of CAR-T therapy.

    Plain language summary

    Chimeric antigen receptor (CAR) T-cell therapy, a treatment that boosts the body's immune system to fight cancer, has made significant progress. Recent research has shown that T-cell exhaustion, which is when the body's immune cells become less effective, affects how well this therapy works. This finding has opened new possibilities to make CAR-T therapy more effective. There is a specific protein called BATF that plays an important role in reducing T-cell exhaustion and influencing the early development of certain immune cells. This review describes how BATF interacts with exhausted T cells, to improve CAR-T therapy. By understanding how BATF works in the immune system, new ways to enhance CAR-T therapy and its ability to fight cancer may be found.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    Reference

    • 1. Wherry EJ. T cell exhaustion. Nat. Immunol. 12(6), 492–499 (2011).
    • 2. Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. 36(4), 265–276 (2015).
    • 3. June CH, O'connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science 359(6382), 1361–1365 (2018).
    • 4. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15(8), 486–499 (2015).
    • 5. Mclane LM, Abdel-Hakeem MS, Wherry EJ. CD8 T cell exhaustion during chronic viral infection and cancer. Ann. Rev. Immunol. 37, 457–495 (2019).
    • 6. Zheng L, Qin S, Si W et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science 374(6574), 1–11 (2021). •• The use of single-cell sequencing technology revealed the role of BATF in the development of tumor-infiltrating T cells.
    • 7. Paley MA, Kroy DC, Odorizzi PM et al. Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338(6111), 1220–1225 (2012).
    • 8. Wherry EJ, Ha SJ, Kaech SM et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27(4), 670–684 (2007).
    • 9. Blackburn SD, Shin H, Freeman GJ, Wherry EJ. Selective expansion of a subset of exhausted CD8 T cells by alphaPD-L1 blockade. Proc. Natl Acad. Sci. USA 105(39), 15016–15021 (2008).
    • 10. Giles JR, Ngiow SF, Manne S et al. Longitudinal single cell transcriptional and epigenetic mapping of effector, memory, and exhausted CD8 T cells reveals shared biological circuits across distinct cell fates. bioRxiv doi: 10.1101/2022.03.27.485974 (2022).
    • 11. Utzschneider DT, Alfei F, Roelli P et al. High antigen levels induce an exhausted phenotype in a chronic infection without impairing T cell expansion and survival. J. Exp. Med. 213(9), 1819–1834 (2016).
    • 12. Papavassiliou AG, Musti AM. The multifaceted output of c-Jun biological activity: focus at the junction of CD8 T cell activation and exhaustion. Cells 9(11), 1–26 (2020).
    • 13. Seo H, Gonzalez-Avalos E, Zhang W et al. BATF and IRF4 cooperate to counter exhaustion in tumor-infiltrating CAR T cells. Nat. Immunol. 22(8), 983–995 (2021). • The summary of how BATF and IRF4 jointly promote the expression of exhaustion status in CAR-T cells.
    • 14. Chen Y, Zander RA, Wu X et al. BATF regulates progenitor to cytolytic effector CD8(+) T cell transition during chronic viral infection. Nat. Immunol. 22(8), 996–1007 (2021).
    • 15. Betz BC, Jordan-Williams KL, Wang C et al. Batf coordinates multiple aspects of B and T cell function required for normal antibody responses. J. Exp. Med. 207(5), 933–942 (2010).
    • 16. Hu B, Li G, Ye Z et al. Transcription factor networks in aged naïve CD4 T cells bias lineage differentiation. Aging Cell 18(4), e12957 (2019).
    • 17. Tsao HW, Kaminski J, Kurachi M et al. Batf-mediated epigenetic control of effector CD8(+) T cell differentiation. Sci. Immunol. 7(68), eabi4919 (2022).
    • 18. Kuroda S, Yamazaki M, Abe M, Sakimura K, Takayanagi H, Iwai Y. Basic leucine zipper transcription factor, ATF-like (BATF) regulates epigenetically and energetically effector CD8 T-cell differentiation via Sirt1 expression. Proc. Natl Acad. Sci. USA 108(36), 14885–14889 (2011).
    • 19. Wang Y, Xiao X, Kong G et al. Genetically targeting the BATF family transcription factors BATF and BATF3 in the mouse abrogates effector T cell activities and enables long-term heart allograft survival. Am. J. Transplant. 22(2), 414–426 (2022).
    • 20. Li S, Zou D, Chen W et al. Ablation of BATF alleviates transplant rejection via abrogating the effector differentiation and memory responses of CD8(+) T cells. Front. Immunol. 13, 882721 (2022).
    • 21. Man K, Gabriel SS, Liao Y et al. Transcription factor IRF4 promotes CD8(+) T cell exhaustion and limits the development of memory-like T cells during chronic infection. Immunity 47(6), 1129–1141 (2017).
    • 22. Xin G, Schauder DM, Lainez B et al. A critical role of IL-21-induced BATF in sustaining CD8-T-cell-mediated chronic viral control. Cell Rep. 13(6), 1118–1124 (2015).
    • 23. Quigley M, Pereyra F, Nilsson B et al. Transcriptional analysis of HIV-specific CD8+ T cells shows that PD-1 inhibits T cell function by upregulating BATF. Nat. Med. 16(10), 1147–1151 (2010).
    • 24. Yang R, Cheng S, Luo N et al. Distinct epigenetic features of tumor-reactive CD8+ T cells in colorectal cancer patients revealed by genome-wide DNA methylation analysis. Genome Biol. 21(1), 2 (2019).
    • 25. Hanada KI, Zhao C, Gil-Hoyos R et al. A phenotypic signature that identifies neoantigen-reactive T cells in fresh human lung cancers. Cancer Cell 40(5), 479–493 (2022).
    • 26. Becker WR, Nevins SA, Chen DC et al. Single-cell analyses define a continuum of cell state and composition changes in the malignant transformation of polyps to colorectal cancer. Nat. Genet. 54(7), 985–995 (2022).
    • 27. Sade-Feldman M, Yizhak K, Bjorgaard SL et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175(4), 998–1013 (2018). •• BATF might serve as an important immune checkpoint in tumor therapy.
    • 28. Ma J, Zheng B, Goswami S et al. PD1(Hi) CD8(+) T cells correlate with exhausted signature and poor clinical outcome in hepatocellular carcinoma. J. Immunother. Cancer 7(1), 331 (2019).
    • 29. Yao C, Sun HW, Lacey NE et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8(+) T cell persistence in chronic infection. Nat. Immunol. 20(7), 890–901 (2019). • The application of single-cell technology revealed the significant role of BATF in the induction of T-cell exhaustion during chronic infection.
    • 30. Glasmacher E, Agrawal S, Chang AB et al. A genomic regulatory element that directs assembly and function of immune-specific AP-1-IRF complexes. Science 338(6109), 975–980 (2012).
    • 31. Li P, Spolski R, Liao W et al. BATF-JUN is critical for IRF4-mediated transcription in T cells. Nature 490(7421), 543–546 (2012).
    • 32. Jiang P, Gu S, Pan D et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat. Med. 24(10), 1550–1558 (2018).
    • 33. Kamphorst AO, Wieland A, Nasti T et al. Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 355(6332), 1423–1427 (2017).
    • 34. Quigley M, Pereyra F, Nilsson B et al. Transcriptional analysis of HIV-specific CD8 + T cells shows that PD-1 inhibits T cell function by upregulating BATF. Nat. Med. 16(10), 1147–1151 (2010).
    • 35. Liu LC, Chen JH, Bae J et al. Rejuvenation of tumour-specific T cells through bispecific antibodies targeting PD-L1 on dendritic cells. Nat. Biomed. Eng. 5(11), 1261–1273 (2021).
    • 36. Spolski R, Leonard WJ. Interleukin-21: basic biology and implications for cancer and autoimmunity. Ann. Rev. Immunol. 26, 57–79 (2008).
    • 37. Topchyan P, Xin G, Chen Y et al. Harnessing the IL-21-BATF pathway in the CD8(+) T cell anti-tumor response. Cancers (Basel) 13(6), 1–15 (2021). •• The first time the signaling pathways associated with IL-21 and BATF were uncovered.
    • 38. Lagumdzic E, Pernold C, Viano M et al. Transcriptome profiling of porcine naïve, intermediate and terminally differentiated CD8(+) T cells. Front. Immunol. 13, 849922 (2022).
    • 39. Cheung KL, Zhang F, Jaganathan A et al. Distinct roles of Brd2 and Brd4 in potentiating the transcriptional program for Th17 cell differentiation. Mol. Cell 65(6), 1068–1080 (2017).
    • 40. Belkina AC, Denis GV. BET domain co-regulators in obesity, inflammation and cancer. Nat. Rev. Cancer 12(7), 465–477 (2012).
    • 41. Maldonado L, Teague JE, Morrow MP et al. Intramuscular therapeutic vaccination targeting HPV16 induces T cell responses that localize in mucosal lesions. Sci. Transl. Med. 6(221), 221ra213 (2014).
    • 42. Wei J, Zheng W, Chapman NM, Geiger TL, Chi H. T cell metabolism in homeostasis and cancer immunity. Curr. Opin. Biotechnol. 68, 240–250 (2021).
    • 43. Brentjens RJ, Rivière I, Park JH et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 118(18), 4817–4828 (2011).
    • 44. Zhang C, Wang Z, Yang Z et al. Phase I escalating-dose trial of CAR-T therapy targeting CEA(+) metastatic colorectal cancers. Mol. Ther. 25(5), 1248–1258 (2017).
    • 45. Wang H, Kaur G, Sankin AI, Chen F, Guan F, Zang X. Immune checkpoint blockade and CAR-T cell therapy in hematologic malignancies. J. Hematol. Oncol. 12(1), 59 (2019).
    • 46. Jiang P, Zhang Z, Hu Y et al. Single-cell ATAC-seq maps the comprehensive and dynamic chromatin accessibility landscape of CAR-T cell dysfunction. Leukemia 36(11), 2656–2668 (2022).
    • 47. Zhang X, Zhang C, Qiao M et al. Depletion of BATF in CAR-T cells enhances antitumor activity by inducing resistance against exhaustion and formation of central memory cells. Cancer Cell 40(11), 1407–1422 (2022). • Experimental studies demonstrated the significant utility of BATF in CAR-T-cell therapy.
    • 48. Guedan S, Posey AD Jr, Shaw C et al. Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight 3(1), 1–17 (2018).
    • 49. BATF and IRF4 prevent CAR T-cell exhaustion. Cancer Discov. 11(9), Of9 (2021). •• The first time the reversal effect of BATF and IRF4 on the exhaustion status of CAR-T cells was revealed.
    • 50. Yang X, Wang B, Cao X. Transcriptional suppression of CD8(+) T cell exhaustion for improving T cell immunotherapy. Cancer Commun. (Lond.) 41(11), 1228–1231 (2021).
    • 51. Scott AC, Dündar F, Zumbo P et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571(7764), 270–274 (2019).
    • 52. Kurachi M, Barnitz RA, Yosef N et al. The transcription factor BATF operates as an essential differentiation checkpoint in early effector CD8+ T cells. Nat. Immunol. 15(4), 373–383 (2014). •• The critical role of BATF in the early differentiation of CD8+ T cells was finally elucidated.
    • 53. Khan O, Giles JR, Mcdonald S et al. TOX transcriptionally and epigenetically programs CD8(+) T cell exhaustion. Nature 571(7764), 211–218 (2019).