Radiotherapy-resistant prostate cancer cells escape immune checkpoint blockade through the senescence-related ataxia telangiectasia and Rad3-related protein

Chenyi Shao , Yingyi Zhang , Hang Li , Jiajia Chen , Ting Huang , Jiaze Li , Simeng Wen , Sen Wang , Saijun Fan , Yu Zhao

Cancer Communications ›› 2025, Vol. 45 ›› Issue (3) : 218 -244.

PDF
Cancer Communications ›› 2025, Vol. 45 ›› Issue (3) : 218 -244. DOI: 10.1002/cac2.12636
ORIGINAL ARTICLE

Radiotherapy-resistant prostate cancer cells escape immune checkpoint blockade through the senescence-related ataxia telangiectasia and Rad3-related protein

Author information +
History +
PDF

Abstract

Background: The majority of patients with prostate cancer (PCa) exhibit intrinsic resistance to immune checkpoint blockade (ICB) following radiotherapy (RT). This resistance is generally attributed to the limited antigen presentation of heterogeneous cells within tumors. Here, we aimed to isolate and characterize these diverse subgroups of tumor post-RT to understand the molecular mechanisms of their resistance to ICB.

Methods: Single-cell RNA-sequencing (scRNA-seq) was used to profile senescent cancer cell clusters induced by RT in LNCaP cells. The expression and phosphorylation levels of ataxia telangiectasia and Rad3-related protein (ATR) were assessed by immunohistochemistry in clinical samples from patients with or without RT. Co-immunoprecipitation, mutagenesis, and Western blotting were used to measure the interactions between proteins. Xenograft experiments were performed to assess the tumor immune response in the mice.

Results: We identified a subset of PCa cells that exhibited resistance to RT, characterized by a reduced antigen presentation capability, which enhanced their ability to evade immune detection and resist cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) blockade. scRNA-seq revealed that the senescent state was a transient phase of PCa cells post-RT, particularly in CTLA-4 blockade treatment-resistant cells. This state was marked by increased cytosolic ATR level. Cytosolic ATR phosphorylated CD86 in its cytosolic domain and enhanced the interaction between CD86 and its E3 ligase MARCH1 through electrostatic attraction. Depletion or inhibition of Atr increased the sensitivity to immune attack and improved responses to anti-Ctla-4 antibody treatment in a mouse model.

Conclusions: Our findings indicate that the activation of cytosolic ATR, which is associated with cellular senescence, impedes the effectiveness of combined RT and ICB treatments. This discovery may provide valuable insights for improving the efficacy of combined RT and ICB therapies in PCa.

Keywords

ATR / CD86 / immune checkpoint / senescence

Cite this article

Download citation ▾
Chenyi Shao, Yingyi Zhang, Hang Li, Jiajia Chen, Ting Huang, Jiaze Li, Simeng Wen, Sen Wang, Saijun Fan, Yu Zhao. Radiotherapy-resistant prostate cancer cells escape immune checkpoint blockade through the senescence-related ataxia telangiectasia and Rad3-related protein. Cancer Communications, 2025, 45(3): 218-244 DOI:10.1002/cac2.12636

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Shitara K, Ajani JA, Moehler M, Garrido M, Gallardo C, Shen L, et al. Nivolumab plus chemotherapy or ipilimumab in gastro-oesophageal cancer. Nature. 2022; 603(7903): 942-948.

[2]

Kraehenbuehl L, Weng CH, Eghbali S, Wolchok JD, Merghoub T. Enhancing immunotherapy in cancer by targeting emerging immunomodulatory pathways. Nat Rev Clin Oncol. 2022; 19(1): 37-50.

[3]

Herrera FG, Bourhis J, Coukos G. Radiotherapy combination opportunities leveraging immunity for the next oncology practice. CA Cancer J Clin. 2017; 67(1): 65-85.

[4]

Matsumura S, Wang B, Kawashima N, Braunstein S, Badura M, Cameron TO, et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J Immunol. 2008; 181(5): 3099-3107.

[5]

Ishizuka JJ, Manguso RT, Cheruiyot CK, Bi K, Panda A, Iracheta-Vellve A, et al. Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade. Nature. 2019; 565(7737): 43-48.

[6]

Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012; 366(10): 925-931.

[7]

Formenti SC, Rudqvist NP, Golden E, Cooper B, Wennerberg E, Lhuillier C, et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat Med. 2018; 24(12): 1845-1851.

[8]

Liu B, Zhang Y, Wang D, Hu X, Zhang Z. Single-cell meta-analyses reveal responses of tumor-reactive CXCL13(+) T cells to immune-checkpoint blockade. Nat Cancer. 2022; 3(9): 1123-1136.

[9]

Deng L, Liang H, Xu M, Yang X, Burnette B, Arina A, et al. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity. 2014; 41(5): 843-852.

[10]

Lugade AA, Moran JP, Gerber SA, Rose RC, Frelinger JG, Lord EM. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J Immunol. 2005; 174(12): 7516-7523.

[11]

Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. 2017; 8: 15618.

[12]

Burnette BC, Liang H, Lee Y, Chlewicki L, Khodarev NN, Weichselbaum RR, et al. The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res. 2011; 71(7): 2488-2496.

[13]

Herrera FG, Ronet C, Ochoa de Olza M, Barras D, Crespo I, Andreatta M, et al. Low-Dose Radiotherapy Reverses Tumor Immune Desertification and Resistance to Immunotherapy. Cancer Discov. 2022; 12(1): 108-133.

[14]

Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, et al. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res. 2005; 11(2 Pt 1): 728-734.

[15]

Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 2014; 124(2): 687-695.

[16]

Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015; 520(7547): 373-377.

[17]

Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, et al. Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 2013; 24(5): 589-602.

[18]

Dovedi SJ, Adlard AL, Lipowska-Bhalla G, McKenna C, Jones S, Cheadle EJ, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014; 74(19): 5458-5468.

[19]

de Almeida DVP, Fong L, Rettig MB, Autio KA. Immune Checkpoint Blockade for Prostate Cancer: Niche Role or Next Breakthrough? Am Soc Clin Oncol Educ Book. 2020; 40: 1-18.

[20]

Lu X, Horner JW, Paul E, Shang X, Troncoso P, Deng P, et al. Effective combinatorial immunotherapy for castration-resistant prostate cancer. Nature. 2017; 543(7647): 728-732.

[21]

Alaia C, Boccellino M, Zappavigna S, Amler E, Quagliuolo L, Rossetti S, et al. Ipilimumab for the treatment of metastatic prostate cancer. Expert Opin Biol Ther. 2018; 18(2): 205-213.

[22]

Tarhini A, Lin Y, Lin H, Rahman Z, Vallabhaneni P, Mendiratta P, et al. Neoadjuvant ipilimumab (3 mg/kg or 10 mg/kg) and high dose IFN-alpha2b in locally/regionally advanced melanoma: safety, efficacy and impact on T-cell repertoire. J Immunother Cancer. 2018; 6(1): 112.

[23]

Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, van den Eertwegh AJ, et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2014; 15(7): 700-712.

[24]

Beer TM, Kwon ED, Drake CG, Fizazi K, Logothetis C, Gravis G, et al. Randomized, Double-Blind, Phase III Trial of Ipilimumab Versus Placebo in Asymptomatic or Minimally Symptomatic Patients With Metastatic Chemotherapy-Naive Castration-Resistant Prostate Cancer. J Clin Oncol. 2017; 35(1): 40-47.

[25]

Morad G, Helmink BA, Sharma P, Wargo JA. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell. 2021; 184(21): 5309-5337.

[26]

Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016; 16(5): 275-287.

[27]

Linsley PS, Ledbetter JA. The role of the CD28 receptor during T cell responses to antigen. Annu Rev Immunol. 1993; 11: 191-212.

[28]

Singh S, Lee N, Pedroza DA, Bado IL, Hamor C, Zhang L, et al. Chemotherapy Coupled to Macrophage Inhibition Induces T-cell and B-cell Infiltration and Durable Regression in Triple-Negative Breast Cancer. Cancer Res. 2022; 82(12): 2281-2297.

[29]

Constant SL, Bottomly K. Induction of Th1 and Th2 CD4+ T cell responses: the alternative approaches. Annu Rev Immunol. 1997; 15: 297-322.

[30]

Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, et al. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 2016; 44(8): e71.

[31]

Das A, Fernandez NR, Levine A, Bianchi V, Stengs LK, Chung J, et al. Combined Immunotherapy Improves Outcome for Replication-Repair-Deficient (RRD) High-Grade Glioma Failing Anti-PD-1 Monotherapy: A Report from the International RRD Consortium. Cancer Discov. 2024; 14(2): 258-273.

[32]

Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014; 15(12): 550.

[33]

Xu S, Hu E, Cai Y, Xie Z, Luo X, Zhan L, et al. Using clusterProfiler to characterize multiomics data. Nat Protoc. 2024; 19: 3292–3320.

[34]

Weiner AB, Kakani P, Armstrong AJ, Bossi A, Cornford P, Feng F, et al. Risk Stratification of Patients with Recurrence After Primary Treatment for Prostate Cancer: A Systematic Review. Eur Urol. 2024; 86(3): 200-210.

[35]

Mukherjee A, Epperly MW, Shields D, Hou W, Fisher R, Hamade D, et al. Ionizing irradiation-induced Fgr in senescent cells mediates fibrosis. Cell Death Discov. 2021; 7(1): 349.

[36]

Li L, Yue HC, Han YW, Liu W, Xiong LG, Zhang JW. Relationship between the invasion of lymphocytes and cytokines in the tumor microenvironment and the interval after single brachytherapy hypofractionated radiotherapy and conventional fractionation radiotherapy in non-small cell lung Cancer. BMC Cancer. 2020; 20(1): 893.

[37]

Zhang P, Wang D, Zhao Y, Ren S, Gao K, Ye Z, et al. Intrinsic BET inhibitor resistance in SPOP-mutated prostate cancer is mediated by BET protein stabilization and AKT-mTORC1 activation. Nat Med. 2017; 23(9): 1055-1062.

[38]

Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, et al. Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018; 24(8): 1246-1256.

[39]

Zhu H, Chen J, Liu K, Gao L, Wu H, Ma L, et al. Human PBMC scRNA-seq-based aging clocks reveal ribosome to inflammation balance as a single-cell aging hallmark and super longevity. Sci Adv. 2023; 9(26): eabq7599.

[40]

Hao Y, Stuart T, Kowalski MH, Choudhary S, Hoffman P, Hartman A, et al. Dictionary learning for integrative, multimodal and scalable single-cell analysis. Nat Biotechnol. 2024; 42(2): 293-304.

[41]

Hanzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 2013; 14: 7.

[42]

Aran D. Cell-Type Enrichment Analysis of Bulk Transcriptomes Using xCell. Methods Mol Biol. 2020; 2120: 263-276.

[43]

Ntala C, Salji M, Salmond J, Officer L, Teodosio AV, Blomme A, et al. Analysis of Prostate Cancer Tumor Microenvironment Identifies Reduced Stromal CD4 Effector T-cell Infiltration in Tumors with Pelvic Nodal Metastasis. European Urology Open Science. 2021; 29: 19-29.

[44]

Wen S, Wei Y, Zen C, Xiong W, Niu Y, Zhao Y. Long non-coding RNA NEAT1 promotes bone metastasis of prostate cancer through N6-methyladenosine. Mol Cancer. 2020; 19(1): 171.

[45]

Zhao Y, Ding L, Wang D, Ye Z, He Y, Ma L, et al. EZH2 cooperates with gain-of-function p53 mutants to promote cancer growth and metastasis. EMBO J. 2019; 38(5): e99599.

[46]

Zhao Y, Li H, Zhang Y, Li L, Fang R, Li Y, et al. Oncoprotein HBXIP Modulates Abnormal Lipid Metabolism and Growth of Breast Cancer Cells by Activating the LXRs/SREBP-1c/FAS Signaling Cascade. Cancer Res. 2016; 76(16): 4696-4707.

[47]

Zhang DKY, Cheung AS, Mooney DJ. Activation and expansion of human T cells using artificial antigen-presenting cell scaffolds. Nat Protoc. 2020; 15(3): 773-798.

[48]

Chen S, Zhu G, Yang Y, Wang F, Xiao YT, Zhang N, et al. Single-cell analysis reveals transcriptomic remodellings in distinct cell types that contribute to human prostate cancer progression. Nat Cell Biol. 2021; 23(1): 87-98.

[49]

Park JW, Lee JK, Sheu KM, Wang L, Balanis NG, Nguyen K, et al. Reprogramming normal human epithelial tissues to a common, lethal neuroendocrine cancer lineage. Science. 2018; 362(6410): 91-95.

[50]

Saxena S, Zou L. Hallmarks of DNA replication stress. Mol Cell. 2022; 82(12): 2298-2314.

[51]

Toledo LI, Altmeyer M, Rask MB, Lukas C, Larsen DH, Povlsen LK, et al. ATR prohibits replication catastrophe by preventing global exhaustion of RPA. Cell. 2013; 155(5): 1088-1103.

[52]

Huang W, Hickson LJ, Eirin A, Kirkland JL, Lerman LO. Cellular senescence: the good, the bad and the unknown. Nat Rev Nephrol. 2022; 18(10): 611-627.

[53]

Cleary JM, Aguirre AJ, Shapiro GI, D'Andrea AD. Biomarker-Guided Development of DNA Repair Inhibitors. Mol Cell. 2020; 78(6): 1070-1085.

[54]

Corcoran K, Jabbour M, Bhagwandin C, Deymier MJ, Theisen DL, Lybarger L. Ubiquitin-mediated regulation of CD86 protein expression by the ubiquitin ligase membrane-associated RING-CH-1 (MARCH1). J Biol Chem. 2011; 286(43): 37168-37180.

[55]

Liu H, Wilson KR, Firth AM, Macri C, Schriek P, Blum AB, et al. Ubiquitin-like protein 3 (UBL3) is required for MARCH ubiquitination of major histocompatibility complex class II and CD86. Nat Commun. 2022; 13(1): 1934.

[56]

Hilton BA, Li Z, Musich PR, Wang H, Cartwright BM, Serrano M, et al. ATR Plays a Direct Antiapoptotic Role at Mitochondria, which Is Regulated by Prolyl Isomerase Pin1. Mol Cell. 2015; 60(1): 35-46.

[57]

Shi Y, Li H, Chu D, Lin W, Wang X, Wu Y, et al. Rescuing Nucleus Pulposus Cells From Senescence via Dual-Functional Greigite Nanozyme to Alleviate Intervertebral Disc Degeneration. Adv Sci (Weinh). 2023; 10(25): e2300988.

[58]

Li Q, Zhao YH, Xu C, Liang YL, Zhao Y, He QM, et al. Chemotherapy-Induced Senescence Reprogramming Promotes Nasopharyngeal Carcinoma Metastasis by circRNA-Mediated PKR Activation. Adv Sci (Weinh). 2023; 10(8): e2205668.

[59]

Trenker R, Wu X, Nguyen JV, Wilcox S, Rubin AF, Call ME, et al. Human and viral membrane-associated E3 ubiquitin ligases MARCH1 and MIR2 recognize different features of CD86 to downregulate surface expression. J Biol Chem. 2021; 297(1): 100900.

[60]

Shi X, Yang Y, Zhang W, Wang J, Xiao D, Ren H, et al. FLASH X-ray spares intestinal crypts from pyroptosis initiated by cGAS-STING activation upon radioimmunotherapy. Proc Natl Acad Sci U S A. 2022; 119(43): e2208506119.

[61]

Paffenholz SV, Salvagno C, Ho YJ, Limjoco M, Baslan T, Tian S, et al. Senescence induction dictates response to chemo- and immunotherapy in preclinical models of ovarian cancer. Proc Natl Acad Sci U S A. 2022; 119(5): e2117754119.

[62]

Berger G, Knelson EH, Jimenez-Macias JL, Nowicki MO, Han S, Panagioti E, et al. STING activation promotes robust immune response and NK cell-mediated tumor regression in glioblastoma models. Proc Natl Acad Sci U S A. 2022; 119(28): e2111003119.

[63]

Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, et al. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature. 2016; 530(7589): 184-189.

[64]

Yang JH, Hayano M, Griffin PT, Amorim JA, Bonkowski MS, Apostolides JK, et al. Loss of epigenetic information as a cause of mammalian aging. Cell. 2023; 186(2): 305-326 e27.

[65]

Owens JL, Beketova E, Liu S, Shen Q, Pawar JS, Asberry AM, et al. Targeting Protein Arginine Methyltransferase 5 Suppresses Radiation-induced Neuroendocrine Differentiation and Sensitizes Prostate Cancer Cells to Radiation. Mol Cancer Ther. 2022; 21(3): 448-459.

[66]

Rudqvist NP, Pilones KA, Lhuillier C, Wennerberg E, Sidhom JW, Emerson RO, et al. Radiotherapy and CTLA-4 Blockade Shape the TCR Repertoire of Tumor-Infiltrating T Cells. Cancer Immunol Res. 2018; 6(2): 139-150.

[67]

Oweida AJ, Darragh L, Phan A, Binder D, Bhatia S, Mueller A, et al. STAT3 Modulation of Regulatory T Cells in Response to Radiation Therapy in Head and Neck Cancer. J Natl Cancer Inst. 2019; 111(12): 1339-1349.

[68]

Takahashi A, Loo TM, Okada R, Kamachi F, Watanabe Y, Wakita M, et al. Downregulation of cytoplasmic DNases is implicated in cytoplasmic DNA accumulation and SASP in senescent cells. Nat Commun. 2018; 9(1): 1249.

[69]

Bouchard JJ, Otero JH, Scott DC, Szulc E, Martin EW, Sabri N, et al. Cancer Mutations of the Tumor Suppressor SPOP Disrupt the Formation of Active, Phase-Separated Compartments. Mol Cell. 2018; 72(1): 19-36. e8.

[70]

Wang X, Tokheim C, Gu SS, Wang B, Tang Q, Li Y, et al. In vivo CRISPR screens identify the E3 ligase Cop1 as a modulator of macrophage infiltration and cancer immunotherapy target. Cell. 2021; 184(21): 5357-5374. e22.

[71]

Gu SS, Zhang W, Wang X, Jiang P, Traugh N, Li Z, et al. Therapeutically Increasing MHC-I Expression Potentiates Immune Checkpoint Blockade. Cancer Discov. 2021; 11(6): 1524-1541.

RIGHTS & PERMISSIONS

2024 The Author(s). Cancer Communications published by John Wiley & Sons Australia, Ltd on behalf of Sun Yat-sen University Cancer Center.

AI Summary AI Mindmap
PDF

30

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/