Potential mechanisms of cancer stem-like progenitor T-cell bio-behaviours

Ling Ni

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (8) : e1817

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (8) : e1817 DOI: 10.1002/ctm2.1817
REVIEW

Potential mechanisms of cancer stem-like progenitor T-cell bio-behaviours

Author information +
History +
PDF

Abstract

•Tpex cells are located in lymph nodes and TLS.

•Several pathways control the differentiation trajectories of Tpex cells, including epigenetic factors, transcription factors, cytokines, age, sex, etc.

Keywords

cancer / CD8 + T cells / immune checkpoint inhibitors / Tex-int cells / Tex-term cells / Tpex cells

Cite this article

Download citation ▾
Ling Ni. Potential mechanisms of cancer stem-like progenitor T-cell bio-behaviours. Clinical and Translational Medicine, 2024, 14(8): e1817 DOI:10.1002/ctm2.1817

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

YangY. Cancer immunotherapy: harnessing the immune system to battle cancer. J Clin Invest. 2015;125(9):3335-3337.

[2]

TayRE, Richardson EK, TohHC. Revisiting the role of CD4(+) T cells in cancer immunotherapy-new insights into old paradigms. Cancer Gene Ther. 2021;28(1-2):5-17.

[3]

WherryEJ. T cell exhaustion. Nat Immunol. 2011;12(6):492-499.

[4]

ImSJ, Hashimoto M, GernerMY, et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature. 2016;537(7620):417-421.

[5]

HudsonWH, Gensheimer J, HashimotoM, et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1(+) stem-like CD8(+) T cells during chronic infection. Immunity. 2019;51(6):043-1058. e1044.

[6]

MillerBC, SenDR, Al AbosyR, et al. Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol. 2019;20(3):326-336.

[7]

ZanderR, Schauder D, XinG, et al. CD4(+) T cell help is required for the formation of a cytolytic CD8(+) T cell subset that protects against chronic infection and cancer. Immunity. 2019;51(6):1028-1042. e1024.

[8]

BeltraJC, ManneS, MSAbdel-Hakeem, et al. Developmental relationships of four exhausted CD8(+) T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity. 2020;52(5):825-841. e828.

[9]

ZehnD, ThimmeR, LugliE, de Almeida GP, OxeniusA. ‘Stem-like’ precursors are the fount to sustain persistent CD8(+) T cell responses. Nat Immunol. 2022;23(6):836-847.

[10]

KasmaniMY, ZanderR, ChungHK, et al. Clonal lineage tracing reveals mechanisms skewing CD8+ T cell fate decisions in chronic infection. J Exp Med. 2023;220(1):e20220679.

[11]

Sade-FeldmanM, YizhakK, BjorgaardSL, et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell. 2018;175(4):998-1013. e1020.

[12]

ConnollyKA, Kuchroo M, VenkatA, et al. A reservoir of stem-like CD8(+) T cells in the tumor-draining lymph node preserves the ongoing antitumor immune response. Sci Immunol. 2021;6(64):eabg7836.

[13]

SchenkelJM, HerbstRH, CannerD, et al. Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1(+) CD8(+) T cells in tumor-draining lymph nodes. Immunity. 2021;54(10):2338-2353. e2336.

[14]

RahimMK, OkholmTLH, JonesKB, et al. Dynamic CD8(+) T cell responses to cancer immunotherapy in human regional lymph nodes are disrupted in metastatic lymph nodes. Cell. 2023;186(6):1127-1143. e1118.

[15]

ImSJ, ObengRC, NastiTH, et al. Characteristics and anatomic location of PD-1(+)TCF1(+) stem-like CD8 T cells in chronic viral infection and cancer. Proc Natl Acad Sci USA. 2023;120(41):e2221985120.

[16]

CalaguaC, FicialM, JansenCS, et al. A subset of localized prostate cancer displays an immunogenic phenotype associated with losses of key tumor suppressor genes. Clin Cancer Res. 2021;27(17):4836-4847.

[17]

WangPH, Washburn RS, MariuzzaDL, et al. Reciprocal transmission of activating and inhibitory signals and cell fate in regenerating T cells. Cell Rep. 2023;42(10):113155.

[18]

BaxterAE, HuangH, GilesJR, et al. The SWI/SNF chromatin remodeling complexes BAF and PBAF differentially regulate epigenetic transitions in exhausted CD8(+) T cells. Immunity. 2023;56(6):1320-1340.

[19]

KharelA, ShenJ, BrownR, et al. Loss of PBAF promotes expansion and effector differentiation of CD8(+) T cells during chronic viral infection and cancer. Cell Rep. 2023;42(6):112649.

[20]

GuoA, HuangH, ZhuZ, et al. cBAF complex components and MYC cooperate early in CD8(+) T cell fate. Nature. 2022;607(7917):135-141.

[21]

LiuY, DeboB, LiM, ShiZ, ShengW, Shi Y. LSD1 inhibition sustains T cell invigoration with a durable response to PD-1 blockade. Nat Commun. 2021;12(1):6831.

[22]

LePT, HaN, TranNK, et al. Targeting Cbx3/HP1gamma induces LEF-1 and IL-21R to promote tumor-infiltrating CD8 T-cell persistence. Front Immunol. 2021;12:738958.

[23]

KimK, ParkS, ParkSY, et al. 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. 2020;12(1):22.

[24]

ZhouP, ShiH, HuangH, et al. Single-cell CRISPR screens in vivo map T cell fate regulomes in cancer. Nature. 2023;624:154-163.

[25]

CastiglioniA, YangY, WilliamsK, et al. Combined PD-L1/TGFbeta blockade allows expansion and differentiation of stem cell-like CD8 T cells in immune excluded tumors. Nat Commun. 2023;14(1):4703.

[26]

SunQ, CaiD, LiuD, et al. BCL6 promotes a stem-like CD8(+) T cell program in cancer via antagonizing BLIMP1. Sci Immunol. 2023;8(88):eadh1306.

[27]

LiuX, HaoJ, WeiP, et al. SMAD4, activated by the TCR-triggered MEK/ERK signaling pathway, critically regulates CD8(+) T cell cytotoxic function. Sci Adv. 2022;8(30):eabo4577.

[28]

TopchyanP, XinG, ChenY, et al. Harnessing the IL-21-BATF pathway in the CD8(+) T cell anti-tumor response. Cancers (Basel). 2021(6):13.

[29]

SunQ, ZhaoX, LiR, et al. STAT3 regulates CD8+ T cell differentiation and functions in cancer and acute infection. J Exp Med. 2023;220(4).

[30]

LeeJ, LeeK, BaeH, et al. IL-15 promotes self-renewal of progenitor exhausted CD8 T cells during persistent antigenic stimulation. Front Immunol. 2023;14:1117092.

[31]

KimH, FengY, MuradR, et al. Melanoma-intrinsic NR2F6 activity regulates antitumor immunity. Sci Adv. 2023;9(27):eadf6621.

[32]

NeubertEN, DeRogatis JM, LewisSA, et al. HMGB2 regulates the differentiation and stemness of exhausted CD8(+) T cells during chronic viral infection and cancer. Nat Commun. 2023;14(1):5631.

[33]

KwonH, Schafer JM, SongNJ, et al. Androgen conspires with the CD8(+) T cell exhaustion program and contributes to sex bias in cancer. Sci Immunol. 2022;7(73):eabq2630.

[34]

LeeJ, Nicosia M, HongES, et al. Sex-biased T-cell exhaustion drives differential immune responses in glioblastoma. Cancer Discov. 2023;13(9):2090-2105.

[35]

WagleMV, Vervoort SJ, KellyMJ, et al. Antigen-driven EGR2 expression is required for exhausted CD8(+) T cell stability and maintenance. Nat Commun. 2021;12(1):2782.

[36]

LaFleurMW, NguyenTH, CoxeMA, et al. PTPN2 regulates the generation of exhausted CD8(+) T cell subpopulations and restrains tumor immunity. Nat Immunol. 2019;20(10):1335-1347.

[37]

SiJ, ShiX, SunS, et al. hematopoietic progenitor kinase1 (HPK1) mediates T cell dysfunction and is a druggable target for T cell-based immunotherapies. Cancer Cell. 2020;38(4):551-566. e511.

[38]

LiuC, Somasundaram A, ManneS, et al. Neuropilin-1 is a T cell memory checkpoint limiting long-term antitumor immunity. Nat Immunol. 2020;21(9):1010-1021.

[39]

VardhanaSA, HweeMA, BerisaM, et al. Impaired mitochondrial oxidative phosphorylation limits the self-renewal of T cells exposed to persistent antigen. Nat Immunol. 2020;21(9):1022-1033.

[40]

LiF, LiuH, ZhangD, Ma Y, ZhuB. Metabolic plasticity and regulation of T cell exhaustion. Immunology. 2022;167(4):482-494.

[41]

BannoudN, Dalotto-Moreno T, KindgardL, et al. Hypoxia supports differentiation of terminally exhausted CD8 T cells. Front Immunol. 2021;12:660944.

[42]

ZhangC, LeiL, YangX, et al. Single-cell sequencing reveals antitumor characteristics of intratumoral immune cells in old mice. J Immunother Cancer. 2021;9(10).

[43]

DammeijerF, van Gulijk M, MulderEE, et al. The PD-1/PD-L1-checkpoint restrains t cell immunity in tumor-draining lymph nodes. Cancer Cell. 2020;38(5):685-700. e688.

[44]

LiuZ, ZhangY, MaN, et al. Progenitor-like exhausted SPRY1(+)CD8(+) T cells potentiate responsiveness to neoadjuvant PD-1 blockade in esophageal squamous cell carcinoma. Cancer Cell. 2023;41(11):1852-1870. e1859.

[45]

MagenA, HamonP, FiaschiN, et al. Intratumoral dendritic cell-CD4(+) T helper cell niches enable CD8(+) T cell differentiation following PD-1 blockade in hepatocellular carcinoma. Nat Med. 2023;29(6):1389-1399.

[46]

BufeS, Zimmermann A, RavensS, et al. PD-1/CTLA-4 blockade leads to expansion of CD8(+)PD-1(int) TILs and results in tumor remission in experimental liver cancer. Liver Cancer. 2023;12(2):129-144.

[47]

ChenCY, UehaS, IshiwataY, et al. Combining an alarmin HMGN1 peptide with PD-L1 blockade results in robust antitumor effects with a concomitant increase of stem-like/progenitor exhausted CD8(+) T cells. Cancer Immunol Res. 2021;9(10):1214-1228.

[48]

KhanSM, DesaiR, CoxonA, et al. Impact of CD4 T cells on intratumoral CD8 T-cell exhaustion and responsiveness to PD-1 blockade therapy in mouse brain tumors. J Immunother Cancer. 2022;10(12):e005293.

[49]

FalvoP, Orecchioni S, HilljeR, et al. Cyclophosphamide and vinorelbine activate stem-like CD8(+) T cells and improve anti-PD-1 efficacy in triple-negative breast cancer. Cancer Res. 2021;81(3):685-697.

[50]

KarakiS, BlancC, TranT, et al. CXCR6 deficiency impairs cancer vaccine efficacy and CD8(+) resident memory T-cell recruitment in head and neck and lung tumors. J Immunother Cancer. 2021;9(3):e001948.

[51]

Leon-LetelierRA, Castro-Medina DI, Badillo-GodinezO, et al. Induction of progenitor exhausted tissue-resident memory CD8(+) T cells upon Salmonella Typhi porins adjuvant immunization correlates with melanoma control and anti-PD-1 immunotherapy cooperation. Front Immunol. 2020;11:583382.

[52]

WuJ, MadiA, MiegA, et al. T cell factor 1 suppresses CD103+ lung tissue-resident memory T cell development. Cell Rep. 2020;31(1):107484.

RIGHTS & PERMISSIONS

2024 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

199

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/