New insights into immune cells in cancer immunotherapy: from epigenetic modification, metabolic modulation to cell communication
Sha Qin
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
Search for more papers by this authorBin Xie
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Search for more papers by this authorQingyi Wang
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
Search for more papers by this authorRui Yang
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
Search for more papers by this authorJingyue Sun
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
Search for more papers by this authorChaotao Hu
Regenerative Medicine, Medical School, University of Chinese Academy of Sciences, Beijing, China
Search for more papers by this authorCorresponding Author
Shuang Liu
Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. University, Changsha, Hunan, China
Correspondence
Desheng Xiao and Yongguang Tao, Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
Email: [email protected]; [email protected]
Shuang Liu, Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. University, Changsha, Hunan, China.
Email: [email protected]
Search for more papers by this authorCorresponding Author
Yongguang Tao
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medicine, Central South university, Changsha, Hunan, China
Correspondence
Desheng Xiao and Yongguang Tao, Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
Email: [email protected]; [email protected]
Shuang Liu, Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. University, Changsha, Hunan, China.
Email: [email protected]
Search for more papers by this authorCorresponding Author
Desheng Xiao
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
Correspondence
Desheng Xiao and Yongguang Tao, Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
Email: [email protected]; [email protected]
Shuang Liu, Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. University, Changsha, Hunan, China.
Email: [email protected]
Search for more papers by this authorSha Qin
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
Search for more papers by this authorBin Xie
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Search for more papers by this authorQingyi Wang
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
Search for more papers by this authorRui Yang
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
Search for more papers by this authorJingyue Sun
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
Search for more papers by this authorChaotao Hu
Regenerative Medicine, Medical School, University of Chinese Academy of Sciences, Beijing, China
Search for more papers by this authorCorresponding Author
Shuang Liu
Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. University, Changsha, Hunan, China
Correspondence
Desheng Xiao and Yongguang Tao, Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
Email: [email protected]; [email protected]
Shuang Liu, Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. University, Changsha, Hunan, China.
Email: [email protected]
Search for more papers by this authorCorresponding Author
Yongguang Tao
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medicine, Central South university, Changsha, Hunan, China
Correspondence
Desheng Xiao and Yongguang Tao, Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
Email: [email protected]; [email protected]
Shuang Liu, Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. University, Changsha, Hunan, China.
Email: [email protected]
Search for more papers by this authorCorresponding Author
Desheng Xiao
Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
Correspondence
Desheng Xiao and Yongguang Tao, Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
Email: [email protected]; [email protected]
Shuang Liu, Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. University, Changsha, Hunan, China.
Email: [email protected]
Search for more papers by this authorAbstract
Cancer is one of the leading causes of death worldwide, and more effective ways of attacking cancer are being sought. Cancer immunotherapy is a new and effective therapeutic method after surgery, radiotherapy, chemotherapy, and targeted therapy. Cancer immunotherapy aims to kill tumor cells by stimulating or rebuilding the body's immune system, with specific efficiency and high safety. However, only few tumor patients respond to immunotherapy and due to the complex and variable characters of cancer immune escape, the behavior and regulatory mechanisms of immune cells need to be deeply explored from more dimensions. Epigenetic modifications, metabolic modulation, and cell-to-cell communication are key factors in immune cell adaptation and response to the complex tumor microenvironment. They collectively determine the state and function of immune cells through modulating gene expression, changing in energy and nutrient demands. In addition, immune cells engage in complex communication networks with other immune components, which are mediated by exosomes, cytokines, and chemokines, and are pivotal in shaping the tumor progression and therapeutic response. Understanding the interactions and combined effects of such multidimensions mechanisms in immune cell modulation is important for revealing the mechanisms of immunotherapy failure and developing new therapeutic targets and strategies.
CONFLICT OF INTEREST STATEMENT
The authors declare that they have no conflict of interest.
Open Research
DATA AVAILABILITY STATEMENT
All data generated or analyzed during this study are included in this published article.
REFERENCES
- 1Advancing cancer therapy. Nat Cancer. 2021; 2(3): 245-246.
- 2Dagher OK, Schwab RD, Brookens SK. Advances in cancer immunotherapies. Cell. 2023; 186(8): 1814-1814. e1811.
- 3Esteve-Puig R, Bueno-Costa A, Esteller M. Writers, readers and erasers of RNA modifications in cancer. Cancer Lett. 2020; 474: 127-137.
- 4Nanamori H, Sawada Y. Epigenetic modification of PD-1/PD-L1-mediated cancer immunotherapy against melanoma. Int J Mol Sci. 2022; 23(3): 1119.
- 5Tao S, Liang S, Zeng T, Yin D. Epigenetic modification-related mechanisms of hepatocellular carcinoma resistance to immune checkpoint inhibition. Front Immunol. 2022; 13:1043667.
- 6Alghazali MW, Al-Hetty H, Ali ZMM, Saleh MM, Suleiman AA, Jalil AT. Non-coding RNAs, another side of immune regulation during triple-negative breast cancer. Pathol Res Pract. 2022; 239:154132.
- 7Chen L, Deng J. Role of non-coding RNA in immune microenvironment and anticancer therapy of gastric cancer. J Mol Med (Berl). 2022; 100(12): 1703-1719.
- 8Yang F, Li J, Ge Q, et al. Non-coding RNAs: emerging roles in the characterization of immune microenvironment and immunotherapy of prostate cancer. Biochem Pharmacol. 2023; 214:115669.
- 9Xia L, Oyang L, Lin J, et al. The cancer metabolic reprogramming and immune response. Mol Cancer. 2021; 20(1): 28.
- 10Yan Y, Chang L, Tian H, et al. 1-Pyrroline-5-carboxylate released by prostate Cancer cell inhibit T cell proliferation and function by targeting SHP1/cytochrome c oxidoreductase/ROS axis. J Immunother Cancer. 2018; 6(1): 148.
- 11Yang L, Chu Z, Liu M, et al. Amino acid metabolism in immune cells: essential regulators of the effector functions, and promising opportunities to enhance cancer immunotherapy. J Hematol Oncol. 2023; 16(1): 59.
- 12Cui C, Wang J, Fagerberg E, et al. Neoantigen-driven B cell and CD4 T follicular helper cell collaboration promotes anti-tumor CD8 T cell responses. Cell. 2021; 184(25): 6101-6118. e6113.
- 13Yu L, Feng R, Zhu L, et al. Promoting the activation of T cells with glycopolymer-modified dendritic cells by enhancing cell interactions. Sci Adv. 2020; 6(47):eabb6595.
- 14Balta E, Wabnitz GH, Samstag Y. Hijacked immune cells in the tumor microenvironment: molecular mechanisms of immunosuppression and cues to improve T cell-based immunotherapy of solid tumors. Int J Mol Sci. 2021; 22(11): 5736.
- 15Bai Z, Zhou Y, Ye Z, Xiong J, Lan H, Wang F. Tumor-infiltrating lymphocytes in colorectal cancer: the fundamental indication and application on immunotherapy. Front Immunol. 2021; 12:808964.
- 16Paijens ST, Vledder A, de Bruyn M, Nijman HW. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021; 18(4): 842-859.
- 17Jacquelot N, Tellier J, Nutt Sl, Belz Gt. Tertiary lymphoid structures and B lymphocytes in cancer prognosis and response to immunotherapies. Oncoimmunology. 2021; 10(1):1900508.
- 18Dong Y, Wan Z, Gao X, Yang G, Liu L. Reprogramming immune cells for enhanced cancer immunotherapy: targets and strategies. Front Immunol. 2021; 12:609762.
- 19Gómez-Aleza C, Nguyen B, Yoldi G, et al. Inhibition of RANK signaling in breast cancer induces an anti-tumor immune response orchestrated by CD8+ T cells. Nat Commun. 2020; 11(1): 6335.
- 20Finisguerra V, Dvorakova T, Formenti M, et al. Metformin improves cancer immunotherapy by directly rescuing tumor-infiltrating CD8 T lymphocytes from hypoxia-induced immunosuppression. J Immunother Cancer. 2023; 11(5):e005719.
- 21Chu Y, Dai E, Li Y, et al. Pan-cancer T cell atlas links a cellular stress response state to immunotherapy resistance. Nat Med. 2023; 29(6): 1550-1562.
- 22Peng S, Hu P, Xiao YT, et al. Single-cell analysis reveals EP4 as a target for restoring T-cell infiltration and sensitizing prostate cancer to immunotherapy. Clin Cancer Res. 2022; 28(3): 552-567.
- 23Luoma AM, Suo S, Wang Y, et al. Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell. 2022; 185(16): 2918-2935. e2929.
- 24Damei I, Trickovic T, Mami-Chouaib F, Corgnac S. Tumor-resident memory T cells as a biomarker of the response to cancer immunotherapy. Front Immunol. 2023; 14:1205984.
- 25Sim MJW, Sun PD. T cell recognition of tumor neoantigens and insights into T cell immunotherapy. Front Immunol. 2022; 13:833017.
- 26Kristensen NP, Heeke C, Tvingsholm SA, et al. Neoantigen-reactive CD8+ T cells affect clinical outcome of adoptive cell therapy with tumor-infiltrating lymphocytes in melanoma. J Clin Invest. 2022; 132(2):e150535.
- 27Meier SL, Satpathy AT, Wells DK. Bystander T cells in cancer immunology and therapy. Nat Cancer. 2022; 3(2): 143-155.
- 28Liang Y, Tan Y, Guan B, et al. Single-cell atlases link macrophages and CD8(+) T-cell subpopulations to disease progression and immunotherapy response in urothelial carcinoma. Theranostics. 2022; 12(18): 7745-7759.
- 29Trefny MP, Kirchhammer N, Auf der Maur P, et al. Deletion of SNX9 alleviates CD8 T cell exhaustion for effective cellular cancer immunotherapy. Nat Commun. 2023; 14(1): 86.
- 30Basu A, Ramamoorthi G, Albert G, et al. Differentiation and regulation of T(H) cells: a balancing act for cancer immunotherapy. Front Immunol. 2021; 12:669474.
- 31Barros MS, de Araújo ND, Magalhães-Gama F, et al. γδ T cells for leukemia immunotherapy: new and expanding trends. Front Immunol. 2021; 12:729085.
- 32Liu X, Si F, Bagley D, et al. Blockades of effector T cell senescence and exhaustion synergistically enhance antitumor immunity and immunotherapy. J Immunother Cancer. 2022; 10(10):e005020.
- 33Chen Y, Jia K, Sun Y, et al. Predicting response to immunotherapy in gastric cancer via multi-dimensional analyses of the tumour immune microenvironment. Nat Commun. 2022; 13(1): 4851.
- 34Jiang S, Ding X, Wu Q, Cheng T, Xu M, Huang J. Identifying immune cells-related phenotype to predict immunotherapy and clinical outcome in gastric cancer. Front Immunol. 2022; 13:980986.
- 35Qin Y, Lu F, Lyu K, Chang AE, Li Q. Emerging concepts regarding pro- and anti tumor properties of B cells in tumor immunity. Front Immunol. 2022; 13:881427.
- 36Guan L, Zhang Z, Gao T, et al. Depleting tumor infiltrating B cells to boost antitumor immunity with tumor immune-microenvironment reshaped hybrid nanocage. ACS Nano. 2022; 16(3): 4263-4277.
- 37Yu Z, Li Y, Li Y, et al. Bufalin stimulates antitumor immune response by driving tumor-infiltrating macrophage toward M1 phenotype in hepatocellular carcinoma. J Immunother Cancer. 2022; 10(5):e004297.
- 38Anfray C, Mainini F, Digifico E, et al. Intratumoral combination therapy with poly(I:c) and resiquimod synergistically triggers tumor-associated macrophages for effective systemic antitumoral immunity. J Immunother Cancer. 2021; 9(9):e002408.
- 39Laureano RS, Sprooten J, Vanmeerbeerk I, et al. Trial watch: dendritic cell (DC)-based immunotherapy for cancer. Oncoimmunology. 2022; 11(1):2096363.
- 40Kießler M, Plesca I, Sommer U, et al. Tumor-infiltrating plasmacytoid dendritic cells are associated with survival in human colon cancer. J Immunother Cancer. 2021; 9(3):e001813.
- 41Creasy CA, Meng YJ, Forget MA, et al. Genomic correlates of outcome in tumor-infiltrating lymphocyte therapy for metastatic melanoma. Clin Cancer Res. 2022; 28(9): 1911-1924.
- 42Huang L, Chen H, Xu Y, Chen J, Liu Z, Xu Q. Correlation of tumor-infiltrating immune cells of melanoma with overall survival by immunogenomic analysis. Cancer Med. 2020; 9(22): 8444-8456.
- 43Dai Q, Wu W, Amei A, Yan X, Lu L, Wang Z. Regulation and characterization of tumor-infiltrating immune cells in breast cancer. Int Immunopharmacol. 2021; 90:107167.
- 44Nelson MA, Ngamcherdtrakul W, Luoh SW, Yantasee W. Prognostic and therapeutic role of tumor-infiltrating lymphocyte subtypes in breast cancer. Cancer Metastasis Rev. 2021; 40(2): 519-536.
- 45Dieci MV, Miglietta F, Guarneri V. Immune infiltrates in breast cancer: recent updates and clinical implications. Cells. 2021; 10(2): 223.
- 46Chen Y, Zhao B, Wang X. Tumor infiltrating immune cells (TIICs) as a biomarker for prognosis benefits in patients with osteosarcoma. BMC Cancer. 2020; 20(1): 1022.
- 47López-Janeiro Á, Villalba-Esparza M, Brizzi ME, et al. The association between the tumor immune microenvironments and clinical outcome in low-grade, early-stage endometrial cancer patients. J Pathol. 2022; 258(4): 426-436.
- 48Che Y, Luo Z, Zhang C, Sun N, Gao S, He J. Immune signature of tumor-infiltrating immune cells predicts the prognosis and therapeutic effects in squamous cell carcinoma. Int Immunopharmacol. 2020; 87:106802.
- 49Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012; 150(1): 12-27.
- 50Dziaman T, Gackowski D, Guz J, et al. Characteristic profiles of DNA epigenetic modifications in colon cancer and its predisposing conditions-benign adenomas and inflammatory bowel disease. Clin Epigenetics. 2018; 10: 72.
- 51Liang W, Zhao Y, Huang W, et al. Non-invasive diagnosis of early-stage lung cancer using high-throughput targeted DNA methylation sequencing of circulating tumor DNA (ctDNA). Theranostics. 2019; 9(7): 2056-2070.
- 52Yadav P, Subbarayalu P, Medina D, et al. M6A RNA methylation regulates histone ubiquitination to support cancer growth and progression. Cancer Res. 2022; 82(10): 1872-1889.
- 53Shi K, Yang S, Chen C, et al. RNA methylation-mediated LINC01559 suppresses colorectal cancer progression by regulating the miR-106b-5p/PTEN axis. Int J Biol Sci. 2022; 18(7): 3048-3065.
- 54Wang Y, Mao Y, Wang C, et al. RNA methylation-related genes of m6A, m5C, and m1A predict prognosis and immunotherapy response in cervical cancer. Ann Med. 2023; 55(1):2190618.
- 55Li D, Li K, Zhang W, et al. The m6A/m5C/m1A regulated gene signature predicts the prognosis and correlates with the immune status of hepatocellular carcinoma. Front Immunol. 2022; 13:918140.
- 56Shao D, Li Y, Wu J, et al. An m6A/m5C/m1A/m7G-related long non-coding RNA signature to predict prognosis and immune features of glioma. Front Genet. 2022; 13:903117.
- 57Audia JE, Campbell RM. Histone modifications and cancer. Cold Spring Harb Perspect Biol. 2016; 8(4):a019521.
- 58Pan L, Feng F, Wu J, et al. Demethylzeylasteral targets lactate by inhibiting histone lactylation to suppress the tumorigenicity of liver cancer stem cells. Pharmacol Res. 2022; 181:106270.
- 59Chen TF, Hao HF, Zhang Y, et al. HBO1 induces histone acetylation and is important for non-small cell lung cancer cell growth. Int J Biol Sci. 2022; 18(8): 3313-3323.
- 60Deng Y, Gao J, Xu G, et al. HDAC6-dependent deacetylation of AKAP12 dictates its ubiquitination and promotes colon cancer metastasis. Cancer Lett. 2022; 549:215911.
- 61Zhang Y, Liu Z, Yang X, et al. H3K27 acetylation activated-COL6A1 promotes osteosarcoma lung metastasis by repressing STAT1 and activating pulmonary cancer-associated fibroblasts. Theranostics. 2021; 11(3): 1473-1492.
- 62Wattanathamsan O, Chantaravisoot N, Wongkongkathep P, et al. Inhibition of histone deacetylase 6 destabilizes ERK phosphorylation and suppresses cancer proliferation via modulation of the tubulin acetylation-GRP78 interaction. J Biomed Sci. 2023; 30(1): 4.
- 63Vatapalli R, Sagar V, Rodriguez Y, et al. Histone methyltransferase DOT1L coordinates AR and MYC stability in prostate cancer. Nat Commun. 2020; 11(1): 4153.
- 64Lv J, Zhou Y, Zhou N, et al. Epigenetic modification of CSDE1 locus dictates immune recognition of nascent tumorigenic cells. Sci Transl Med. 2023; 15(681):eabq6024.
- 65Lukinović V, Hausmann S, Roth GS, et al. SMYD3 impedes small cell lung cancer sensitivity to alkylation damage through RNF113A methylation-phosphorylation cross-talk. Cancer Discov. 2022; 12(9): 2158-2179.
- 66Hogg SJ, Beavis PA, Dawson MA, Johnstone RW. Targeting the epigenetic regulation of antitumour immunity. Nat Rev Drug Discov. 2020; 19(11): 776-800.
- 67Villanueva L, Alvarez-Errico D, Esteller M. The contribution of epigenetics to cancer immunotherapy. Trends Immunol. 2020; 41(8): 676-691.
- 68Tough DF, Rioja I, Modis LK, Prinjha RK. Epigenetic regulation of T cell memory: recalling therapeutic implications. Trends Immunol. 2020; 41(1): 29-45.
- 69Chiappinelli KB, Strissel PL, Desrichard A, et al. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell. 2017; 169(2): 361.
- 70Zebley CC, Abdelsamed HA, Ghoneim HE, et al. Proinflammatory cytokines promote TET2-mediated DNA demethylation during CD8 T cell effector differentiation. Cell Rep. 2021; 37(2):109796.
- 71Li HB, Tong J, Zhu S, et al. m(6)A mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature. 2017; 548(7667): 338-342.
- 72Dong L, Chen C, Zhang Y, et al. The loss of RNA N(6)-adenosine methyltransferase Mettl14 in tumor-associated macrophages promotes CD8(+) T cell dysfunction and tumor growth. Cancer Cell. 2021; 39(7): 945-957. e910.
- 73Zheng X, Sarode P, Weigert A, et al. The HDAC2-SP1 axis orchestrates protumor macrophage polarization. Cancer Res. 2023; 83(14): 2345-2357.
- 74Yang W, Feng Y, Zhou J, et al. A selective HDAC8 inhibitor potentiates antitumor immunity and efficacy of immune checkpoint blockade in hepatocellular carcinoma. Sci Transl Med. 2021; 13(588):eaaz6804.
- 75Luda KM, Longo J, Kitchen-Goosen SM, et al. Ketolysis drives CD8(+) T cell effector function through effects on histone acetylation. Immunity. 2023; 56(9): 2021-2035.
- 76Cho H, Son WC, Lee YS, et al. Differential effects of histone deacetylases on the expression of NKG2D ligands and NK cell-mediated anticancer immunity in lung cancer cells. Molecules. 2021; 26(13): 3952.
- 77Li L, Hao S, Gao M, et al. HDAC3 inhibition promotes antitumor immunity by enhancing CXCL10-mediated chemotaxis and recruiting of immune cells. Cancer Immunol Res. 2023; 11(5): 657-673.
- 78Alanazi S, Rabelo Melo F, Pejler G. Tryptase regulates the epigenetic modification of core histones in mast cell leukemia cells. Front Immunol. 2021; 12:804408.
- 79Yu B, Luo F, Sun B, et al. KAT6A acetylation of SMAD3 regulates myeloid-derived suppressor cell recruitment, metastasis, and immunotherapy in triple-negative breast cancer. Adv Sci (Weinh). 2021; 8(20):e2100014.
- 80Piunti A, Shilatifard A. The roles of Polycomb repressive complexes in mammalian development and cancer. Nat Rev Mol Cell Biol. 2021; 22(5): 326-345.
- 81Li J, Li Y, Cao Y, et al. Polycomb chromobox (Cbx) 7 modulates activation-induced CD4+ T cell apoptosis. Arch Biochem Biophys. 2014; 564: 184-188.
- 82Ren L, Li Z, Zhou Y, et al. CBX4 promotes antitumor immunity by suppressing Pdcd1 expression in T cells. Mol Oncol. 2023; 17(12): 2694-2708.
- 83Jing R, Scarfo I, Najia MA, et al. EZH1 repression generates mature iPSC-derived CAR T cells with enhanced antitumor activity. Cell Stem Cell. 2022; 29(8): 1181-1196. e1186.
- 84Stairiker CJ, Pfister SX, Hendrickson E, et al. EZH2 inhibition compromises α4-1BB-mediated antitumor efficacy by reducing the survival and effector programming of CD8(+) T cells. Front Immunol. 2021; 12:770080.
- 85Ramakrishnan S, Granger V, Rak M, et al. Inhibition of EZH2 induces NK cell-mediated differentiation and death in muscle-invasive bladder cancer. Cell Death Differ. 2019; 26(10): 2100-2114.
- 86Chibaya L, Murphy KC, DeMarco KD, et al. EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance. Nat Cancer. 2023; 4(6): 872-892.
- 87DuPage M, Chopra G, Quiros J, et al. The chromatin-modifying enzyme Ezh2 is critical for the maintenance of regulatory T cell identity after activation. Immunity. 2015; 42(2): 227-238.
- 88Yin J, Leavenworth JW, Li Y, et al. Ezh2 regulates differentiation and function of natural killer cells through histone methyltransferase activity. Proc Natl Acad Sci U S A. 2015; 112(52): 15988-15993.
- 89Wang D, Quiros J, Mahuron K, et al. Targeting EZH2 reprograms intratumoral regulatory T cells to enhance cancer immunity. Cell Rep. 2018; 23(11): 3262-3274.
- 90Liu F, Wu D, Wang X. Roles of CTCF in conformation and functions of chromosome. Semin Cell Dev Biol. 2019; 90: 168-173.
- 91Holwerda SJ, de Laat W. CTCF: the protein, the binding partners, the binding sites and their chromatin loops. Philos Trans R Soc Lond B Biol Sci. 2013; 368(1620):20120369.
- 92Dehingia B, Milewska M, Janowski M, Pękowska A. CTCF shapes chromatin structure and gene expression in health and disease. EMBO Rep. 2022; 23(9):e55146.
- 93Shan Q, Zhu S, Chen X, et al. Tcf1-CTCF cooperativity shapes genomic architecture to promote CD8(+) T cell homeostasis. Nat Immunol. 2022; 23(8): 1222-1235.
- 94Quon S, Yu B, Russ BE, et al. DNA architectural protein CTCF facilitates subset-specific chromatin interactions to limit the formation of memory CD8(+) T cells. Immunity. 2023; 56(5): 959-978. e910.
- 95Yang B, Kim S, Jung WJ, et al. CTCF controls three-dimensional enhancer network underlying the inflammatory response of bone marrow-derived dendritic cells. Nat Commun. 2023; 14(1): 1277.
- 96Stik G, Vidal E, Barrero M, et al. CTCF is dispensable for immune cell transdifferentiation but facilitates an acute inflammatory response. Nat Genet. 2020; 52(7): 655-661.
- 97Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144(5): 646-674.
- 98Finley LWS. What is cancer metabolism? Cell. 2023; 186(8): 1670-1688.
- 99Faubert B, Solmonson A, DeBerardinis RJ. Metabolic reprogramming and cancer progression. Science. 2020; 368(6487):eaaw5473.
- 100Pavlova NN, Zhu J, Thompson CB. The hallmarks of cancer metabolism: still emerging. Cell Metab. 2022; 34(3): 355-377.
- 101Luo X, Zheng E, Wei L, et al. The fatty acid receptor CD36 promotes HCC progression through activating Src/PI3K/AKT axis-dependent aerobic glycolysis. Cell Death Dis. 2021; 12(4): 328.
- 102Sun Z, Zhang R, Zhang X, et al. LINE-1 promotes tumorigenicity and exacerbates tumor progression via stimulating metabolism reprogramming in non-small cell lung cancer. Mol Cancer. 2022; 21(1): 147.
- 103Jung J, Zeng H, Horng T. Metabolism as a guiding force for immunity. Nat Cell Biol. 2019; 21(1): 85-93.
- 104Wu F, Fan J, He Y, et al. Single-cell profiling of tumor heterogeneity and the microenvironment in advanced non-small cell lung cancer. Nat Commun. 2021; 12(1): 2540.
- 105Xiao Z, Dai Z, Locasale JW. Metabolic landscape of the tumor microenvironment at single cell resolution. Nat Commun. 2019; 10(1): 3763.
- 106Leone RD, Powell JD. Metabolism of immune cells in cancer. Nat Rev Cancer. 2020; 20(9): 516-531.
- 107Huang B, Song BL, Xu C. Cholesterol metabolism in cancer: mechanisms and therapeutic opportunities. Nat Metab. 2020; 2(2): 132-141.
- 108Chen B, Gao A, Tu B, et al. Metabolic modulation via mTOR pathway and anti-angiogenesis remodels tumor microenvironment using PD-L1-targeting codelivery. Biomaterials. 2020; 255:120187.
- 109Zhang B, Vogelzang A, Miyajima M, et al. B cell-derived GABA elicits IL-10(+) macrophages to limit anti-tumour immunity. Nature. 2021; 599(7885): 471-476.
- 110Reinfeld BI, Madden MZ, Wolf MM, et al. Cell-programmed nutrient partitioning in the tumour microenvironment. Nature. 2021; 593(7858): 282-288.
- 111Mu X, Xiang Z, Xu Y, et al. Glucose metabolism controls human γδ T-cell-mediated tumor immunosurveillance in diabetes. Cell Mol Immunol. 2022; 19(8): 944-956.
- 112Watson MJ, Vignali PDA, Mullett SJ, et al. Metabolic support of tumour-infiltrating regulatory T cells by lactic acid. Nature. 2021; 591(7851): 645-651.
- 113Kumagai S, Koyama S, Itahashi K, et al. Lactic acid promotes PD-1 expression in regulatory T cells in highly glycolytic tumor microenvironments. Cancer Cell. 2022; 40(2): 201-218. e209.
- 114Poznanski SM, Singh K, Ritchie TM, et al. Metabolic flexibility determines human NK cell functional fate in the tumor microenvironment. Cell Metab. 2021; 33(6): 1205-1220. e1205.
- 115Feng Q, Liu Z, Yu X, et al. Lactate increases stemness of CD8 + T cells to augment anti-tumor immunity. Nat Commun. 2022; 13(1): 4981.
- 116Luu M, Riester Z, Baldrich A, et al. Microbial short-chain fatty acids modulate CD8(+) T cell responses and improve adoptive immunotherapy for cancer. Nat Commun. 2021; 12(1): 4077.
- 117Israr M, Lam F, DeVoti J, et al. PGE(2) expression by HPV6/11-induced respiratory papillomas blocks NK cell activation in patients with recurrent respiratory papillomatosis. Eur J Immunol. 2023; 53(4):e2250036.
- 118Pellegrini JM, Martin C, Morelli MP, et al. PGE2 displays immunosuppressive effects during human active tuberculosis. Sci Rep. 2021; 11(1):13559.
- 119Pi C, Jing P, Li B, et al. Reversing PD-1 resistance in B16F10 cells and recovering tumour immunity using a COX2 inhibitor. Cancers (Basel). 2022; 14(17): 4134.
- 120Thumkeo D, Punyawatthananukool S, Prasongtanakij S, et al. PGE(2)-EP2/EP4 signaling elicits immunosuppression by driving the mregDC-Treg axis in inflammatory tumor microenvironment. Cell Rep. 2022; 39(10):110914.
- 121Yamamichi K, Fukuda T, Sanui T, et al. Amelogenin induces M2 macrophage polarisation via PGE2/cAMP signalling pathway. Arch Oral Biol. 2017; 83: 241-251.
- 122Cheng X, Tan X, Wang W, et al. Long-chain acylcarnitines induce senescence of invariant natural killer T cells in hepatocellular carcinoma. Cancer Res. 2023; 83(4): 582-594.
- 123Liu X, Hartman CL, Li L, et al. Reprogramming lipid metabolism prevents effector T cell senescence and enhances tumor immunotherapy. Sci Transl Med. 2021; 13(587):eaaz6314.
- 124Edwards DN, Ngwa VM, Raybuck AL, et al. Selective glutamine metabolism inhibition in tumor cells improves antitumor T lymphocyte activity in triple-negative breast cancer. J Clin Invest. 2021; 131(4):e140100.
- 125Huang M, Xiong D, Pan J, et al. Targeting glutamine metabolism to enhance immunoprevention of EGFR-driven lung cancer. Adv Sci (Weinh). 2022; 9(26):e2105885.
- 126Leone RD, Zhao L, Englert JM, et al. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science. 2019; 366(6468): 1013-1021.
- 127Geiger R, Rieckmann JC, Wolf T, et al. l-Arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell. 2016; 167(3): 829-842. e813.
- 128Huang X, Sun T, Wang J, et al. Metformin reprograms tryptophan metabolism to stimulate CD8+ T-cell function in colorectal cancer. Cancer Res. 2023; 83(14): 2358-2371.
- 129Hung MH, Lee JS, Ma C, et al. Tumor methionine metabolism drives T-cell exhaustion in hepatocellular carcinoma. Nat Commun. 2021; 12(1): 1455.
- 130Sepich-Poore GD, Zitvogel L, Straussman R, Hasty J, Wargo JA, Knight R. The microbiome and human cancer. Science. 2021; 371(6536):eabc4552.
- 131Schaupp L, Muth S, Rogell L, et al. Microbiota-induced type I interferons instruct a poised basal state of dendritic cells. Cell. 2020; 181(5): 1080-1096. e1019.
- 132Winkler ES, Shrihari S. The intestinal microbiome restricts alphavirus infection and dissemination through a bile acid-type I IFN signaling axis. Cell. 2020; 182(4): 901-918. e918.
- 133Ma C, Han M, Heinrich B, et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science. 2018; 360(6391):eaan5931.
- 134Overacre-Delgoffe AE, Bumgarner HJ, Cillo AR, et al. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity. 2021; 54(12): 2812-2824. e2814.
- 135Pal S, Perrien DS, Yumoto T, et al. The microbiome restrains melanoma bone growth by promoting intestinal NK and Th1 cell homing to bone. J Clin Invest. 2022; 132(12):e157340.
- 136He Y, Fu L, Li Y, et al. Gut microbial metabolites facilitate anticancer therapy efficacy by modulating cytotoxic CD8(+) T cell immunity. Cell Metab. 2021; 33(5): 988-1000. e1007.
- 137Peng R, Liu S, You W, et al. Gastric microbiome alterations are associated with decreased CD8+ tissue-resident memory T cells in the tumor microenvironment of gastric cancer. Cancer Immunol Res. 2022; 10(10): 1224-1240.
- 138Xing C, Wang M, Ajibade AA, et al. Microbiota regulate innate immune signaling and protective immunity against cancer. Cell Host Microbe. 2021; 29(6): 959-974. e957.
- 139Steinert EM, Vasan K, Chandel NS. Mitochondrial metabolism regulation of T cell-mediated immunity. Annu Rev Immunol. 2021; 39: 395-416.
- 140Wang Y, Li N, Zhang X, Horng T. Mitochondrial metabolism regulates macrophage biology. J Biol Chem. 2021; 297(1):100904.
- 141Saha T, Dash C, Jayabalan R, et al. Intercellular nanotubes mediate mitochondrial trafficking between cancer and immune cells. Nat Nanotechnol. 2022; 17(1): 98-106.
- 142Gao Z, Li Y, Wang F, et al. Mitochondrial dynamics controls anti-tumour innate immunity by regulating CHIP-IRF1 axis stability. Nat Commun. 2017; 8(1): 1805.
- 143Lötscher J, Martí ILAA, Kirchhammer N, et al. Magnesium sensing via LFA-1 regulates CD8(+) T cell effector function. Cell. 2022; 185(4): 585-602. e529.
- 144Song J, Liu T, Yin Y, et al. The deubiquitinase OTUD1 enhances iron transport and potentiates host antitumor immunity. EMBO Rep. 2021; 22(2):e51162.
- 145Chen S, Cui W, Chi Z, et al. Tumor-associated macrophages are shaped by intratumoral high potassium via Kir2.1. Cell Metab. 2022; 34(11): 1843-1859. e1811.
- 146Lv M, Chen M, Zhang R, et al. Manganese is critical for antitumor immune responses via cGAS-STING and improves the efficacy of clinical immunotherapy. Cell Res. 2020; 30(11): 966-979.
- 147Cen D, Ge Q, Xie C, et al. ZnS@BSA nanoclusters potentiate efficacy of cancer immunotherapy. Adv Mater. 2021; 33(49):e2104037.
- 148Li Q, Chao Y, Liu B, et al. Disulfiram loaded calcium phosphate nanoparticles for enhanced cancer immunotherapy. Biomaterials. 2022; 291:121880.
- 149Zheng P, Ding B, Jiang Z, et al. Ultrasound-augmented mitochondrial calcium ion overload by calcium nanomodulator to induce immunogenic cell death. Nano Lett. 2021; 21(5): 2088-2093.
- 150Shyer JA, Flavell RA, Bailis W. Metabolic signaling in T cells. Cell Res. 2020; 30(8): 649-659.
- 151Yang F, Wang T, Du P, Fan H, Dong X, Guo H. M2 bone marrow-derived macrophage-derived exosomes shuffle microRNA-21 to accelerate immune escape of glioma by modulating PEG3. Cancer Cell Int. 2020; 20: 93.
- 152Du T, Yang CL, Ge MR, et al. M1 macrophage derived exosomes aggravate experimental autoimmune neuritis via modulating Th1 response. Front Immunol. 2020; 11: 1603.
- 153Pitt JM, André F, Amigorena S, et al. Dendritic cell-derived exosomes for cancer therapy. J Clin Invest. 2016; 126(4): 1224-1232.
- 154Viaud S, Terme M, Flament C, et al. Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: a role for NKG2D ligands and IL-15Ralpha. PLoS One. 2009; 4(3):e4942.
- 155Xiong J, Chi H, Yang G, et al. Revolutionizing anti-tumor therapy: unleashing the potential of B cell-derived exosomes. Front Immunol. 2023; 14:1188760.
- 156Klinker MW, Lizzio V, Reed TJ, Fox DA, Lundy SK. Human B cell-derived lymphoblastoid cell lines constitutively produce fas ligand and secrete MHCII(+)FasL(+) killer exosomes. Front Immunol. 2014; 5: 144.
- 157Tung SL, Boardman DA, Sen M, et al. Regulatory T cell-derived extracellular vesicles modify dendritic cell function. Sci Rep. 2018; 8(1): 6065.
- 158Zhou J, Li X, Wu X, et al. Exosomes released from tumor-associated macrophages transfer miRNAs that induce a Treg/Th17 cell imbalance in epithelial ovarian cancer. Cancer Immunol Res. 2018; 6(12): 1578-1592.
- 159Skokos D, Botros HG, Demeure C, et al. Mast cell-derived exosomes induce phenotypic and functional maturation of dendritic cells and elicit specific immune responses in vivo. J Immunol. 2003; 170(6): 3037-3045.
- 160Fabbri M. Natural killer cell-derived vesicular miRNAs: a new anticancer approach? Cancer Res. 2020; 80(1): 17-22.
- 161Pramanik A, Bhattacharyya S. Myeloid derived suppressor cells and innate immune system interaction in tumor microenvironment. Life Sci. 2022; 305:120755.
- 162Franklin RA, Liao W, Sarkar A, et al. The cellular and molecular origin of tumor-associated macrophages. Science. 2014; 344(6186): 921-925.
- 163Sinha P, Clements VK, Bunt SK, Albelda SM, Ostrand-Rosenberg S. Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J Immunol. 2007; 179(2): 977-983.
- 164Thibodeau J, Bourgeois-Daigneault MC, Huppé G, et al. Interleukin-10-induced MARCH1 mediates intracellular sequestration of MHC class II in monocytes. Eur J Immunol. 2008; 38(5): 1225-1230.
- 165Li H, Han Y, Guo Q, Zhang M, Cao X. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1. J Immunol. 2009; 182(1): 240-249.
- 166Joshi S, Sharabi A. Targeting myeloid-derived suppressor cells to enhance natural killer cell-based immunotherapy. Pharmacol Ther. 2022; 235:108114.
- 167Gabrilovich DI. Myeloid-Derived Suppressor Cells. Cancer Immunol Res. 2017; 5(1): 3-8.
- 168Zhang J, Han X, Hu X, et al. IDO1 impairs NK cell cytotoxicity by decreasing NKG2D/NKG2DLs via promoting miR-18a. Mol Immunol. 2018; 103: 144-155.
- 169Zhao H, Teng D, Yang L, et al. Myeloid-derived itaconate suppresses cytotoxic CD8(+) T cells and promotes tumour growth. Nat Metab. 2022; 4(12): 1660-1673.
- 170Grazioli P, Orlando A, Giordano N, et al. Notch-signaling deregulation induces myeloid-derived suppressor cells in T-Cell acute lymphoblastic leukemia. Front Immunol. 2022; 13:809261.
- 171Dar AA, Patil RS, Pradhan TN, Chaukar DA, D'Cruz AK, Chiplunkar SV. Myeloid-derived suppressor cells impede T cell functionality and promote Th17 differentiation in oral squamous cell carcinoma. Cancer Immunol Immunother. 2020; 69(6): 1071-1086.
- 172Haist M, Stege H, Grabbe S, Bros M. The functional crosstalk between myeloid-derived suppressor cells and regulatory T cells within the immunosuppressive tumor microenvironment. Cancers (Basel). 2021; 13(2): 210.
- 173Ostrand-Rosenberg S, Sinha P, Beury DW, Clements VK. Cross-talk between myeloid-derived suppressor cells (MDSC), macrophages, and dendritic cells enhances tumor-induced immune suppression. Semin Cancer Biol. 2012; 22(4): 275-281.
- 174Garaud S, Dieu-Nosjean MC, Willard-Gallo K. T follicular helper and B cell crosstalk in tertiary lymphoid structures and cancer immunotherapy. Nat Commun. 2022; 13(1): 2259.
- 175Poschke I, Mao Y, Adamson L, Salazar-Onfray F, Masucci G, Kiessling R. Myeloid-derived suppressor cells impair the quality of dendritic cell vaccines. Cancer Immunol Immunother. 2012; 61(6): 827-838.
- 176Ugolini A, Tyurin VA, Tyurina YY, et al. Polymorphonuclear myeloid-derived suppressor cells limit antigen cross-presentation by dendritic cells in cancer. JCI Insight. 2020; 5(15):e138581.
- 177Kouketsu A, Haruka S, Kuroda K, et al. Myeloid-derived suppressor cells and plasmacytoid dendritic cells are associated with oncogenesis of oral squamous cell carcinoma. J Oral Pathol Med. 2023; 52(1): 9-19.
- 178Mun JY, Leem SH, Lee JH, Kim HS. Dual relationship between stromal cells and immune cells in the tumor microenvironment. Front Immunol. 2022; 13:864739.
- 179Ksiazkiewicz M, Gottfried E, Kreutz M, Mack M, Hofstaedter F, Kunz-Schughart LA. Importance of CCL2-CCR2A/2B signaling for monocyte migration into spheroids of breast cancer-derived fibroblasts. Immunobiology. 2010; 215(9-10): 737-747.
- 180Gok Yavuz B, Gunaydin G, Gedik ME, et al. Cancer associated fibroblasts sculpt tumour microenvironment by recruiting monocytes and inducing immunosuppressive PD-1(+) TAMs. Sci Rep. 2019; 9(1): 3172.
- 181Cheng Y, Li H, Deng Y, et al. Cancer-associated fibroblasts induce PDL1+ neutrophils through the IL6-STAT3 pathway that foster immune suppression in hepatocellular carcinoma. Cell Death Dis. 2018; 9(4): 422.
- 182Qian L, Tang Z, Yin S, et al. Fusion of dendritic cells and cancer-associated fibroblasts for activation of anti-tumor cytotoxic T lymphocytes. J Biomed Nanotechnol. 2018; 14(10): 1826-1835.
- 183Sung E, Ko M, Won JY, et al. LAG-3xPD-L1 bispecific antibody potentiates antitumor responses of T cells through dendritic cell activation. Mol Ther. 2022; 30(8): 2800-2816.
- 184Liu L, Chen J, Bae J, et al. Rejuvenation of tumour-specific T cells through bispecific antibodies targeting PD-L1 on dendritic cells. Nat Biomed Eng. 2021; 5(11): 1261-1273.
- 185Shapir Itai Y, Barboy O, Salomon R, et al. Bispecific dendritic-T cell engager potentiates anti-tumor immunity. Cell. 2024; 187(2): 375-389. e318.
- 186Hossain MA, Liu G, Dai B, et al. Reinvigorating exhausted CD8(+) cytotoxic T lymphocytes in the tumor microenvironment and current strategies in cancer immunotherapy. Med Res Rev. 2021; 41(1): 156-201.
- 187Leone RD, Powell JD. Fueling the revolution: targeting metabolism to enhance immunotherapy. Cancer Immunol Res. 2021; 9(3): 255-260.
- 188Li X, Wenes M, Romero P, Huang SC, Fendt SM, Ho PC. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat Rev Clin Oncol. 2019; 16(7): 425-441.
- 189Gu M, Zhou X, Sohn JH, et al. NF-κB-inducing kinase maintains T cell metabolic fitness in antitumor immunity. Nat Immunol. 2021; 22(2): 193-204.
- 190Sievers C, Craveiro M, Friedman J, et al. Phenotypic plasticity and reduced tissue retention of exhausted tumor-infiltrating T cells following neoadjuvant immunotherapy in head and neck cancer. Cancer Cell. 2023; 41(5): 887-902. e885.
- 191Wang Y, Wang F, Wang L, et al. NAD(+) supplement potentiates tumor-killing function by rescuing defective TUB-mediated NAMPT transcription in tumor-infiltrated T cells. Cell Rep. 2021; 36(6):109516.
- 192Wang X, Li B, Kim YJ, et al. Targeting monoamine oxidase A for T cell-based cancer immunotherapy. Sci Immunol. 2021; 6(59):eabh2383.
- 193Li Q, Xiang M. Metabolic reprograming of MDSCs within tumor microenvironment and targeting for cancer immunotherapy. Acta Pharmacol Sin. 2022; 43(6): 1337-1348.
- 194Gao A, Liu X, Lin W, et al. Tumor-derived ILT4 induces T cell senescence and suppresses tumor immunity. J Immunother Cancer. 2021; 9(3):e001536.
- 195Liu J, Shen H, Gu W, et al. Prediction of prognosis, immunogenicity and efficacy of immunotherapy based on glutamine metabolism in lung adenocarcinoma. Front Immunol. 2022; 13:960738.
- 196Yoshikawa T, Wu Z, Inoue S, et al. Genetic ablation of PRDM1 in antitumor T cells enhances therapeutic efficacy of adoptive immunotherapy. Blood. 2022; 139(14): 2156-2172.
- 197Zhou J, Kryczek I, Li S, et al. The ubiquitin ligase MDM2 sustains STAT5 stability to control T cell-mediated antitumor immunity. Nat Immunol. 2021; 22(4): 460-470.
- 198Zou Q, Wang X, Ren D, et al. DNA methylation-based signature of CD8+ tumor-infiltrating lymphocytes enables evaluation of immune response and prognosis in colorectal cancer. J Immunother Cancer. 2021; 9(9):e002671.
- 199Yu M, Liu X, Xu H, et al. Comprehensive evaluation of the m(6)A regulator prognostic risk score in the prediction of immunotherapy response in clear cell renal cell carcinoma. Front Immunol. 2022; 13:818120.
- 200Dong H, Xie C, Yao Z, et al. PTPRO-related CD8(+) T-cell signatures predict prognosis and immunotherapy response in patients with breast cancer. Front Immunol. 2022; 13:947841.
- 201Tanagala KKK, Morin-Baxter J, Carvajal R, et al. SP140 inhibits STAT1 signaling, induces IFN-γ in tumor-associated macrophages, and is a predictive biomarker of immunotherapy response. J Immunother Cancer. 2022; 10(12):e005088.
- 202Petitprez F, Meylan M, de Reyniès A, Sautès-Fridman C, Fridman WH. The tumor microenvironment in the response to immune checkpoint blockade therapies. Front Immunol. 2020; 11: 784.
- 203Jaiswal A, Verma A, Dannenfelser R, et al. An activation to memory differentiation trajectory of tumor-infiltrating lymphocytes informs metastatic melanoma outcomes. Cancer Cell. 2022; 40(5): 524-544. e525.
- 204Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018; 8(9): 1069-1086.
- 205Tian X, Ning Q, Yu J, Tang S. T-cell immunoglobulin and ITIM domain in cancer immunotherapy: a focus on tumor-infiltrating regulatory T cells. Mol Immunol. 2022; 147: 62-70.
- 206Tay C, Tanaka A, Sakaguchi S. Tumor-infiltrating regulatory T cells as targets of cancer immunotherapy. Cancer Cell. 2023; 41(3): 450-465.
- 207Stanczak MA, Läubli H. Siglec receptors as new immune checkpoints in cancer. Mol Aspects Med. 2023; 90:101112.
- 208Wang J, Sun J, Liu LN, et al. Siglec-15 as an immune suppressor and potential target for normalization cancer immunotherapy. Nat Med. 2019; 25(4): 656-666.
- 209Xu JX, Maher VE, Zhang L, et al. FDA approval summary: nivolumab in advanced renal cell carcinoma after anti-angiogenic therapy and exploratory predictive biomarker analysis. Oncologist. 2017; 22(3): 311-317.
- 210Kazandjian D, Suzman DL, Blumenthal G, et al. FDA approval summary: nivolumab for the treatment of metastatic non-small cell lung cancer with progression on or after platinum-based chemotherapy. Oncologist. 2016; 21(5): 634-642.
- 211Kasamon YL, de Claro RA, Wang Y, Shen YL, Farrell AT, Pazdur R. FDA approval summary: nivolumab for the treatment of relapsed or progressive classical hodgkin lymphoma. Oncologist. 2017; 22(5): 585-591.
- 212Horiba MN, Casak SJ, Mishra-Kalyani PS, et al. FDA approval summary: nivolumab for the adjuvant treatment of adults with completely resected esophageal/gastroesophageal junction cancer and residual pathologic disease. Clin Cancer Res. 2022; 28(24): 5244-5248.
- 213Shah M, Osgood CL, Amatya AK, et al. FDA approval summary: pembrolizumab for neoadjuvant and adjuvant treatment of patients with high-risk early-stage triple-negative breast cancer. Clin Cancer Res. 2022; 28(24): 5249-5253.
- 214Pai-Scherf L, Blumenthal GM, Li H, et al. FDA Approval summary: pembrolizumab for treatment of metastatic non-small cell lung cancer: first-line therapy and beyond. Oncologist. 2017; 22(11): 1392-1399.
- 215Chuk MK, Chang JT, Theoret MR, et al. FDA approval summary: accelerated approval of pembrolizumab for second-line treatment of metastatic melanoma. Clin Cancer Res. 2017; 23(19): 5666-5670.
- 216Suzman DL, Agrawal S, Ning YM, et al. FDA approval summary: atezolizumab or pembrolizumab for the treatment of patients with advanced urothelial carcinoma ineligible for cisplatin-containing chemotherapy. Oncologist. 2019; 24(4): 563-569.
- 217Mathieu LN, Larkins E, Sinha AK, et al. FDA approval summary: atezolizumab as adjuvant treatment following surgical resection and platinum-based chemotherapy for stage II to IIIA NSCLC. Clin Cancer Res. 2023; 29(16): 2973-2978.
- 218Casak SJ, Donoghue M, Fashoyin-Aje L, et al. FDA approval summary: atezolizumab plus bevacizumab for the treatment of patients with advanced unresectable or metastatic hepatocellular carcinoma. Clin Cancer Res. 2021; 27(7): 1836-1841.
- 219Powles T, Park SH, Voog E, et al. Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. N Engl J Med. 2020; 383(13): 1218-1230.
- 220D'Angelo SP, Lebbé C, Mortier L, et al. First-line avelumab in a cohort of 116 patients with metastatic Merkel cell carcinoma (JAVELIN Merkel 200): primary and biomarker analyses of a phase II study. J Immunother Cancer. 2021; 9(7):e002646.
- 221Keam SJ. Tremelimumab: first approval. Drugs. 2023; 83(1): 93-102.
- 222Cameron F, Whiteside G, Perry C. Ipilimumab: first global approval. Drugs. 2011; 71(8): 1093-1104.
- 223Blum SM, Rouhani SJ, Sullivan RJ. Effects of immune-related adverse events (irAEs) and their treatment on antitumor immune responses. Immunol Rev. 2023; 318(1): 167-178.
- 224Tong J, Kartolo A, Yeung C, Hopman W, Baetz T. Long-term toxicities of immune checkpoint inhibitor (ICI) in melanoma patients. Curr Oncol. 2022; 29(10): 7953-7963.
- 225Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010; 363(5): 411-422.
- 226Brower V. Approval of provenge seen as first step for cancer treatment vaccines. J Natl Cancer Inst. 2010; 102(15): 1108-1110.
- 227Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018; 378(5): 439-448.
- 228Bach PB, Giralt SA, Saltz LB. FDA approval of tisagenlecleucel: promise and complexities of a $475 000 cancer drug. Jama. 2017; 318(19): 1861-1862.
- 229Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017; 377(26): 2531-2544.
- 230FDA approves second CAR T-cell therapy. Cancer Discov. 2018; 8(1): 5-6.
- 231Frey NV. Approval of brexucabtagene autoleucel for adults with relapsed and refractory acute lymphocytic leukemia. Blood. 2022; 140(1): 11-15.
- 232Abramson JS, Palomba ML, Gordon LI, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020; 396(10254): 839-852.
- 233Sun D, Liu J, Zhou H, et al. Classification of tumor immune microenvironment according to programmed death-ligand 1 expression and immune infiltration predicts response to immunotherapy plus chemotherapy in advanced patients with NSCLC. J Thorac Oncol. 2023; 18(7): 869-881.
- 234Rohaan MW, Borch TH, van den Berg JH, et al. Tumor-infiltrating lymphocyte therapy or ipilimumab in advanced melanoma. N Engl J Med. 2022; 387(23): 2113-2125.
- 235Hall MS, Mullinax JE, Cox CA, et al. Combination nivolumab, CD137 agonism, and adoptive cell therapy with tumor-infiltrating lymphocytes for patients with metastatic melanoma. Clin Cancer Res. 2022; 28(24): 5317-5329.
- 236Liang YJ, Chen QY, Xu JX, et al. A phase II randomised controlled trial of adjuvant tumour-infiltrating lymphocytes for pretreatment Epstein-Barr virus DNA-selected high-risk nasopharyngeal carcinoma patients. Eur J Cancer. 2023; 191:112965.
- 237Mailankody S, Devlin SM, Landa J, et al. GPRC5D-targeted CAR T cells for myeloma. N Engl J Med. 2022; 387(13): 1196-1206.
- 238Mailankody S, Matous JV, Chhabra S, et al. Allogeneic BCMA-targeting CAR T cells in relapsed/refractory multiple myeloma: phase 1 UNIVERSAL trial interim results. Nat Med. 2023; 29(2): 422-429.
- 239Gill S, Vides V, Frey NV, et al. Anti-CD19 CAR T cells in combination with ibrutinib for the treatment of chronic lymphocytic leukemia. Blood Adv. 2022; 6(21): 5774-5785.
- 240Park JH, Nath K, Devlin SM, et al. CD19 CAR T-cell therapy and prophylactic anakinra in relapsed or refractory lymphoma: phase 2 trial interim results. Nat Med. 2023; 29(7): 1710-1717.
- 241Pan K, Farrukh H, Chittepu V, Xu H, Pan CX, Zhu Z. CAR race to cancer immunotherapy: from CAR T, CAR NK to CAR macrophage therapy. J Exp Clin Cancer Res. 2022; 41(1): 119.
- 242Chen HX, Song M, Maecker HT, et al. Network for biomarker immunoprofiling for cancer immunotherapy: cancer immune monitoring and analysis centers and cancer immunologic data commons (CIMAC-CIDC). Clin Cancer Res. 2021; 27(18): 5038-5048.
- 243Bai Y, Xie T, Wang Z, et al. Efficacy and predictive biomarkers of immunotherapy in Epstein-Barr virus-associated gastric cancer. J Immunother Cancer. 2022; 10(3):e004080.
- 244Sadeghirad H, Liu N, Monkman J, et al. Compartmentalized spatial profiling of the tumor microenvironment in head and neck squamous cell carcinoma identifies immune checkpoint molecules and tumor necrosis factor receptor superfamily members as biomarkers of response to immunotherapy. Front Immunol. 2023; 14:1135489.
- 245Zhou X, Cao J, Topatana W, et al. Evaluation of PD-L1 as a biomarker for immunotherapy for hepatocellular carcinoma: systematic review and meta-analysis. Immunotherapy. 2023; 15(5): 353-365.
- 246Marcos Rubio A, Everaert C, Van Damme E, De Preter K, Vermaelen K. Circulating immune cell dynamics as outcome predictors for immunotherapy in non-small cell lung cancer. J Immunother Cancer. 2023; 11(8):e007023.
- 247Sivapalan L, Murray JC, Canzoniero JV, et al. Liquid biopsy approaches to capture tumor evolution and clinical outcomes during cancer immunotherapy. J Immunother Cancer. 2023; 11(1):e005924.
- 248Chalfin HJ, Pramparo T, Mortazavi A, et al. Circulating tumor cell subtypes and T-cell populations as prognostic biomarkers to combination immunotherapy in patients with metastatic genitourinary cancer. Clin Cancer Res. 2021; 27(5): 1391-1398.
- 249Duan L, Liu X, Luo Z, et al. G-protein subunit gamma 4 as a potential biomarker for predicting the response of chemotherapy and immunotherapy in bladder cancer. Genes (Basel). 2022; 13(4): 693.
- 250Kim ES, Velcheti V, Mekhail T, et al. Blood-based tumor mutational burden as a biomarker for atezolizumab in non-small cell lung cancer: the phase 2 B-F1RST trial. Nat Med. 2022; 28(5): 939-945.
- 251Shen R, Postow MA, Adamow M, et al. LAG-3 expression on peripheral blood cells identifies patients with poorer outcomes after immune checkpoint blockade. Sci Transl Med. 2021; 13(608):eabf5107.
- 252Signorelli D, Ghidotti P, Proto C, et al. Circulating CD81-expressing extracellular vesicles as biomarkers of response for immune-checkpoint inhibitors in advanced NSCLC. Front Immunol. 2022; 13:987639.
- 253Clevers MR, Kastelijn EA, Peters BJM, Kelder H, Schramel F. Evaluation of serum biomarker CEA and Ca-125 as immunotherapy response predictors in metastatic non-small cell lung cancer. Anticancer Res. 2021; 41(2): 869-876.
- 254He Y, Zhang X, Zhu M, et al. Soluble PD-L1: a potential dynamic predictive biomarker for immunotherapy in patients with proficient mismatch repair colorectal cancer. J Transl Med. 2023; 21(1): 25.
- 255Tian X, Xu WH, Xu FJ, et al. Identification of prognostic biomarkers in papillary renal cell carcinoma and PTTG1 may serve as a biomarker for predicting immunotherapy response. Ann Med. 2022; 54(1): 211-226.
- 256Chang P, Chen S, Chang X, Zhu J, Tang Q, Ma L. EXTL3 could serve as a potential biomarker of prognosis and immunotherapy for prostate cancer and its potential mechanisms. Eur J Med Res. 2022; 27(1): 115.
- 257Chen L, Zhou Q, Liu J, Zhang W. CTNNB1 alternation is a potential biomarker for immunotherapy prognosis in patients with hepatocellular carcinoma. Front Immunol. 2021; 12:759565.
- 258Deng J, Ma X, Ni Y, et al. Identification of CXCL5 expression as a predictive biomarker associated with response and prognosis of immunotherapy in patients with non-small cell lung cancer. Cancer Med. 2022; 11(8): 1787-1795.
- 259Li B, Zhang G, Xu X. APC mutation correlated with poor response of immunotherapy in colon cancer. BMC Gastroenterol. 2023; 23(1): 95.
- 260Li H, Sun X, Zhao Y, et al. Pan-cancer analysis of TASL: a novel immune infiltration-related biomarker for tumor prognosis and immunotherapy response prediction. BMC Cancer. 2023; 23(1): 528.
- 261Dai Y, Qiang W, Lin K, Gui Y, Lan X, Wang D. An immune-related gene signature for predicting survival and immunotherapy efficacy in hepatocellular carcinoma. Cancer Immunol Immunother. 2021; 70(4): 967-979.
- 262Pan D, Hu AY, Antonia SJ, Li CY. A gene mutation signature predicting immunotherapy benefits in patients with NSCLC. J Thorac Oncol. 2021; 16(3): 419-427.
- 263Zhang Y, Vu T, Palmer DC, et al. A T cell resilience model associated with response to immunotherapy in multiple tumor types. Nat Med. 2022; 28(7): 1421-1431.
- 264Zhang Y, Wang Q, Yang WK, et al. Development of an immune-related prognostic biomarker for triple-negative breast cancer. Ann Med. 2022; 54(1): 1212-1220.
- 265Ghiringhelli F, Bibeau F, Greillier L, et al. Immunoscore immune checkpoint using spatial quantitative analysis of CD8 and PD-L1 markers is predictive of the efficacy of anti- PD1/PD-L1 immunotherapy in non-small cell lung cancer. EBioMedicine. 2023; 92:104633.
- 266Tsakiroglou AM, Fergie M, Oguejiofor K, et al. Spatial proximity between T and PD-L1 expressing cells as a prognostic biomarker for oropharyngeal squamous cell carcinoma. Br J Cancer. 2020; 122(4): 539-544.
- 267Zhang Z, Wang ZX, Chen YX, et al. Integrated analysis of single-cell and bulk RNA sequencing data reveals a pan-cancer stemness signature predicting immunotherapy response. Genome Med. 2022; 14(1): 45.
- 268Zhang HC, Deng SH, Pi YN, et al. Identification and validation in a novel quantification system of ferroptosis patterns for the prediction of prognosis and immunotherapy response in left- and right-sided colon cancer. Front Immunol. 2022; 13:855849.
- 269Tang B, Yan R, Zhu J, et al. Integrative analysis of the molecular mechanisms, immunological features and immunotherapy response of ferroptosis regulators across 33 cancer types. Int J Biol Sci. 2022; 18(1): 180-198.
- 270Zhou M, Zhang Z, Bao S, et al. Computational recognition of lncRNA signature of tumor-infiltrating B lymphocytes with potential implications in prognosis and immunotherapy of bladder cancer. Brief Bioinform. 2021; 22(3):bbaa047.
- 271Zhang X, Liang H, Tang Q, Chen H, Guo F. Pyroptosis-related gene to construct prognostic signature and explore immune microenvironment and immunotherapy biomarkers in bladder cancer. Front Genet. 2022; 13:801665.
- 272Zeng Z, Liang Y, Shi J, et al. Identification and application of a novel immune-related lncRNA signature on the prognosis and immunotherapy for lung adenocarcinoma. Diagnostics (Basel). 2022; 12(11): 2891.
- 273Chen M, Lu H, Copley SJ, et al. A novel radiogenomics biomarker for predicting treatment response and pneumotoxicity from programmed cell death protein or ligand-1 inhibition immunotherapy in NSCLC. J Thorac Oncol. 2023; 18(6): 718-730.
- 274Bosenberg M, Liu ET, Yu CI, Palucka K. Mouse models for immuno-oncology. Trends Cancer. 2023; 9(7): 578-590.
- 275Sato Y, Fu Y, Liu H, Lee MY, Shaw MH. Tumor-immune profiling of CT-26 and Colon 26 syngeneic mouse models reveals mechanism of anti-PD-1 response. BMC Cancer. 2021; 21(1): 1222.
- 276Denis M, Grasselly C, Choffour PA, et al. In vivo syngeneic tumor models with acquired resistance to anti-PD-1/PD-L1 therapies. Cancer Immunol Res. 2022; 10(8): 1013-1027.
- 277Katuwal NB, Park N, Pandey K, et al. Preclinical platform using a triple-negative breast cancer syngeneic murine model to evaluate immune checkpoint inhibitors. Anticancer Res. 2023; 43(1): 85-95.
- 278Ghosh S, He X, Huang WC, Lovell JF. Immune checkpoint blockade enhances chemophototherapy in a syngeneic pancreatic tumor model. APL Bioeng. 2022; 6(3):036105.
- 279Dabagian H, Taghvaee T, Martorano P, et al. PARP targeted alpha-particle therapy enhances response to PD-1 immune-checkpoint blockade in a syngeneic mouse model of glioblastoma. ACS Pharmacol Transl Sci. 2021; 4(1): 344-351.
- 280Kim H, Kim M, Im SK, Fang S. Mouse Cre-LoxP system: general principles to determine tissue-specific roles of target genes. Lab Anim Res. 2018; 34(4): 147-159.
- 281Takada I, Hidano S, Takahashi S, et al. Transcriptional coregulator Ess2 controls survival of post-thymic CD4(+) T cells through the Myc and IL-7 signaling pathways. J Biol Chem. 2022; 298(9):102342.
- 282Zhong H, Lu W, Tang Y, et al. SOX9 drives KRAS-induced lung adenocarcinoma progression and suppresses anti-tumor immunity. Oncogene. 2023; 42(27): 2183-2194.
- 283Galvani E, Mundra PA, Valpione S, et al. Stroma remodeling and reduced cell division define durable response to PD-1 blockade in melanoma. Nat Commun. 2020; 11(1): 853.
- 284Chuprin J, Buettner H, Seedhom MO, et al. Humanized mouse models for immuno-oncology research. Nat Rev Clin Oncol. 2023; 20(3): 192-206.
- 285Pearson T, Greiner DL, Shultz LD. Creation of “humanized” mice to study human immunity. Curr Protoc Immunol. 2008; Chapter 15: 15.21.11-15.21.21.
- 286Cheng H, Zheng Z, Cheng T. New paradigms on hematopoietic stem cell differentiation. Protein Cell. 2020; 11(1): 34-44.
- 287McCune JM, Namikawa R, Kaneshima H, Shultz LD, Lieberman M, Weissman IL. The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function. Science. 1988; 241(4873): 1632-1639.
- 288Phung SK, Miller JS, Felices M. Bi-specific and tri-specific NK cell engagers: the new avenue of targeted NK cell immunotherapy. Mol Diagn Ther. 2021; 25(5): 577-592.
- 289Mhaidly R, Verhoeyen E. Humanized mice are precious tools for preclinical evaluation of CAR T and CAR NK cell therapies. Cancers (Basel). 2020; 12(7): 1915.
- 290Leclercq G, Haegel H, Toso A, et al. JAK and mTOR inhibitors prevent cytokine release while retaining T cell bispecific antibody in vivo efficacy. J Immunother Cancer. 2022; 10(1):e003766.
- 291Wang M, Yao LC, Cheng M, et al. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy. Faseb j. 2018; 32(3): 1537-1549.
- 292Oswald E, Bug D, Grote A, et al. Immune cell infiltration pattern in non-small cell lung cancer PDX models is a model immanent feature and correlates with a distinct molecular and phenotypic make-up. J Immunother Cancer. 2022; 10(4):e004412.
- 293Qin H, Yang L, Chukinas JA, et al. Systematic preclinical evaluation of CD33-directed chimeric antigen receptor T cell immunotherapy for acute myeloid leukemia defines optimized construct design. J Immunother Cancer. 2021; 9(9):e003149.
- 294Gambacorta V, Beretta S, Ciccimarra M, et al. Integrated multiomic profiling identifies the epigenetic regulator PRC2 as a therapeutic target to counteract leukemia immune escape and relapse. Cancer Discov. 2022; 12(6): 1449-1461.
- 295Seymour L, Bogaerts J, Perrone A, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017; 18(3): e143-e152.
- 296Vaseq R, Sharma A, Li Y, Schmidt-Wolf IGH. Revising the landscape of cytokine-induced killer cell therapy in lung cancer: focus on immune checkpoint inhibitors. International Journal of Molecular Sciences. 2023; 24(6): 5626.
- 297Mohsenzadegan M, Peng RW, Roudi R. Dendritic cell/cytokine-induced killer cell-based immunotherapy in lung cancer: what we know and future landscape. J Cell Physiol. 2020; 235(1): 74-86.
- 298Zhang L, Yang X, Sun Z, et al. Dendritic cell vaccine and cytokine-induced killer cell therapy for the treatment of advanced non-small cell lung cancer. Oncol Lett. 2016; 11(4): 2605-2610.
- 299Zhou L, Xiong Y, Wang Y, et al. A phase IB trial of autologous cytokine-induced killer cells in combination with sintilimab, monoclonal antibody against programmed cell death-1, plus chemotherapy in patients with advanced non-small-cell lung cancer. Clin Lung Cancer. 2022; 23(8): 709-719.