Low level of ARID1A contributes to adaptive immune resistance and sensitizes triple-negative breast cancer to immune checkpoint inhibitors
Xin-Yu Chen
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorBin Li
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorYe Wang
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorJuan Jin
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorYu Yang
State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, P. R. China
Search for more papers by this authorLei-Huan Huang
State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, P. R. China
Search for more papers by this authorMeng-Di Yang
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorJian Zhang
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorBi-Yun Wang
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorZhi-Ming Shao
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Search for more papers by this authorTing Ni
State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, P. R. China
Search for more papers by this authorSheng-Lin Huang
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorCorresponding Author
Xi-Chun Hu
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Correspondence
Zhong-Hua Tao and Xi-Chun Hu, Department of Breast Cancer and Urologic Medical Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China.
Email: [email protected] and [email protected]
Search for more papers by this authorCorresponding Author
Zhong-Hua Tao
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Correspondence
Zhong-Hua Tao and Xi-Chun Hu, Department of Breast Cancer and Urologic Medical Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China.
Email: [email protected] and [email protected]
Search for more papers by this authorXin-Yu Chen
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorBin Li
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorYe Wang
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorJuan Jin
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorYu Yang
State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, P. R. China
Search for more papers by this authorLei-Huan Huang
State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, P. R. China
Search for more papers by this authorMeng-Di Yang
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorJian Zhang
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorBi-Yun Wang
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorZhi-Ming Shao
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Search for more papers by this authorTing Ni
State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, P. R. China
Search for more papers by this authorSheng-Lin Huang
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Search for more papers by this authorCorresponding Author
Xi-Chun Hu
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Correspondence
Zhong-Hua Tao and Xi-Chun Hu, Department of Breast Cancer and Urologic Medical Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China.
Email: [email protected] and [email protected]
Search for more papers by this authorCorresponding Author
Zhong-Hua Tao
Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, P. R. China
Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
Correspondence
Zhong-Hua Tao and Xi-Chun Hu, Department of Breast Cancer and Urologic Medical Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China.
Email: [email protected] and [email protected]
Search for more papers by this authorXin-Yu Chen and Bin Li contributed equally to this work.
Abstract
Background
Immune checkpoint inhibitors (ICIs) shed new light on triple-negative breast cancer (TNBC), but only a minority of patients demonstrate response. Therefore, adaptive immune resistance (AIR) needs to be further defined to guide the development of ICI regimens.
Methods
Databases, including The Cancer Genome Atlas, Gene Ontology Resource, University of California Santa Cruz Genome Browser, and Pubmed, were used to screen epigenetic modulators, regulators for CD8+ T cells, and transcriptional regulators of programmed cell death-ligand 1 (PD-L1). Human peripheral blood mononuclear cell (Hu-PBMC) reconstruction mice were adopted for xenograft transplantation. Tumor specimens from a TNBC cohort and the clinical trial CTR20191353 were retrospectively analyzed. RNA-sequencing, Western blotting, qPCR and immunohistochemistry were used to assess gene expression. Coculture assays were performed to evaluate the regulation of TNBC cells on T cells. Chromatin immunoprecipitation and transposase-accessible chromatin sequencing were used to determine chromatin-binding and accessibility.
Results
The epigenetic modulator AT-rich interaction domain 1A (ARID1A) gene demonstrated the highest expression association with AIR relative to other epigenetic modulators in TNBC patients. Low ARID1A expression in TNBC, causing an immunosuppressive microenvironment, promoted AIR and inhibited CD8+ T cell infiltration and activity through upregulating PD-L1. However, ARID1A did not directly regulate PD-L1 expression. We found that ARID1A directly bound the promoter of nucleophosmin 1 (NPM1) and that low ARID1A expression increased NPM1 chromatin accessibility as well as gene expression, further activating PD-L1 transcription. In Hu-PBMC mice, atezolizumab demonstrated the potential to reverse ARID1A deficiency-induced AIR in TNBC by reducing tumor malignancy and activating anti-tumor immunity. In CTR20191353, ARID1A-low patients derived more benefit from pucotenlimab compared to ARID1A-high patients.
Conclusions
In AIR epigenetics, low ARID1A expression in TNBC contributed to AIR via the ARID1A/NPM1/PD-L1 axis, leading to poor outcome but sensitivity to ICI treatment.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
Open Research
DATA AVAILABILITY STATEMENT
RNA-seq, ChIP-seq and ATAC-seq data have been deposited to the GEO repository with the accession number GSE234179. Other datasets generated or analyzed during the current study are available in TCGA Program (https://www.cbioportal.org), UCSC Genome Browser (https://genome.ucsc.edu), GEO repository (https://www.ncbi.nlm.nih.gov/geo/), Molecular Signature Database (https://www.gsea-msigdb.org/gsea/msigdb/index.jsp), Gene Ontology Resource (http://geneontology.org), EnhancerDB (http://lcbb.swjtu.edu.cn/EnhancerDB/) and PubMed (https://pubmed.ncbi.nlm.nih.gov).
Supporting Information
Filename | Description |
---|---|
cac212465-sup-0001-tableS1-S6.xlsx28.4 KB | Supporting information |
cac212465-sup-0002-figureS1-S9.docx12.9 MB | Supporting information |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
REFERENCES
- 1Lei S, Zheng R, Zhang S, Wang S, Chen R, Sun K, et al. Global patterns of breast cancer incidence and mortality: A population-based cancer registry data analysis from 2000 to 2020. Cancer Commun (Lond). 2021; 41(11): 1183–94.
- 2Savas P, Salgado R, Denkert C, Sotiriou C, Darcy PK, Smyth MJ, et al. Clinical relevance of host immunity in breast cancer: from TILs to the clinic. Nat Rev Clin Oncol. 2016; 13(4): 228–41.
- 3Bareche Y, Buisseret L, Gruosso T, Girard E, Venet D, Dupont F, et al. Unraveling Triple-Negative Breast Cancer Tumor Microenvironment Heterogeneity: Towards an Optimized Treatment Approach. J Natl Cancer Inst. 2020; 112(7): 708–19.
- 4Cortes J, Cescon DW, Rugo HS, Nowecki Z, Im SA, Yusof MM, et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020; 396(10265): 1817–28.
- 5Winer EP, Lipatov O, Im SA, Goncalves A, Munoz-Couselo E, Lee KS, et al. Pembrolizumab versus investigator-choice chemotherapy for metastatic triple-negative breast cancer (KEYNOTE-119): a randomised, open-label, phase 3 trial. Lancet Oncol. 2021; 22(4): 499–511.
- 6Adams S, Diamond JR, Hamilton E, Pohlmann PR, Tolaney SM, Chang CW, et al. Atezolizumab Plus nab-Paclitaxel in the Treatment of Metastatic Triple-Negative Breast Cancer With 2-Year Survival Follow-up: A Phase 1b Clinical Trial. JAMA Oncol. 2019; 5(3): 334–42.
- 7Adams S, Schmid P, Rugo HS, Winer EP, Loirat D, Awada A, et al. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase II KEYNOTE-086 study. Ann Oncol. 2019; 30(3): 397–404.
- 8Aftimos P, Oliveira M, Irrthum A, Fumagalli D, Sotiriou C, Gal-Yam EN, et al. Genomic and Transcriptomic Analyses of Breast Cancer Primaries and Matched Metastases in AURORA, the Breast International Group (BIG) Molecular Screening Initiative. Cancer Discov. 2021; 11(11): 2796–811.
- 9Ma H, Kang Z, Foo TK, Shen Z, Xia B. Disrupted BRCA1-PALB2 interaction induces tumor immunosuppression and T-lymphocyte infiltration in HCC through cGAS-STING pathway. Hepatology. 2022; 77(1): 33–47.
- 10Ma S, Zhao Y, Lee WC, Ong LT, Lee PL, Jiang Z, et al. Hypoxia induces HIF1alpha-dependent epigenetic vulnerability in triple negative breast cancer to confer immune effector dysfunction and resistance to anti-PD-1 immunotherapy. Nat Commun. 2022 Jul 15; 13(1): 4118.
- 11Lai Q, Wang H, Li A, Xu Y, Tang L, Chen Q, et al. Decitibine improve the efficiency of anti-PD-1 therapy via activating the response to IFN/PD-L1 signal of lung cancer cells. Oncogene. 2018; 37(17): 2302–12.
- 12Shen J, Ju Z, Zhao W, Wang L, Peng Y, Ge Z, et al. ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat Med. 2018; 24(5): 556–62.
- 13Braun DA, Ishii Y, Walsh AM, Van Allen EM, Wu CJ, Shukla SA, et al. Clinical Validation of PBRM1 Alterations as a Marker of Immune Checkpoint Inhibitor Response in Renal Cell Carcinoma. JAMA Oncol. 2019; 5(11): 1631–33.
- 14Goswami S, Chen Y, Anandhan S, Szabo PM, Basu S, Blando JM, et al. ARID1A mutation plus CXCL13 expression act as combinatorial biomarkers to predict responses to immune checkpoint therapy in mUCC. Sci Transl Med. 2020; 12(548).
- 15Wu JI, Lessard J, Crabtree GR. Understanding the words of chromatin regulation. Cell. 2009; 136(2): 200–6.
- 16Wilson BG, Roberts CW. SWI/SNF nucleosome remodellers and cancer. Nat Rev Cancer. 2011; 11(7): 481–92.
- 17Nagarajan S, Rao SV, Sutton J, Cheeseman D, Dunn S, Papachristou EK, et al. ARID1A influences HDAC1/BRD4 activity, intrinsic proliferative capacity and breast cancer treatment response. Nat Genet. 2020; 52(2): 187–97.
- 18Xu G, Chhangawala S, Cocco E, Razavi P, Cai Y, Otto JE, et al. ARID1A determines luminal identity and therapeutic response in estrogen-receptor-positive breast cancer. Nat Genet. 2020; 52(2): 198–207.
- 19Bitler BG, Fatkhutdinov N, Zhang R. Potential therapeutic targets in ARID1A-mutated cancers. Expert Opin Ther Targets. 2015; 19(11): 1419–22.
- 20Jiang T, Chen X, Su C, Ren S, Zhou C. Pan-cancer analysis of ARID1A Alterations as Biomarkers for Immunotherapy Outcomes. J Cancer. 2020; 11(4): 776–80.
- 21Sun X, Wang SC, Wei Y, Luo X, Jia Y, Li L, et al. Arid1a Has Context-Dependent Oncogenic and Tumor Suppressor Functions in Liver Cancer. Cancer Cell. 2017; 32(5): 574–89 e6.
- 22Fontana B, Gallerani G, Salamon I, Pace I, Roncarati R, Ferracin M. ARID1A in cancer: Friend or foe? Front Oncol. 2023; 13:1136248.
- 23Tao Z, Li T, Feng Z, Liu C, Shao Y, Zhu M, et al. Characterizations of Cancer Gene Mutations in Chinese Metastatic Breast Cancer Patients. Front Oncol. 2020; 10: 1023.
- 24Jiang YZ, Ma D, Suo C, Shi J, Xue M, Hu X, et al. Genomic and Transcriptomic Landscape of Triple-Negative Breast Cancers: Subtypes and Treatment Strategies. Cancer Cell. 2019; 35(3): 428-40 e5.
- 25Yates LR, Knappskog S, Wedge D, Farmery JHR, Gonzalez S, Martincorena I, et al. Genomic Evolution of Breast Cancer Metastasis and Relapse. Cancer Cell. 2017; 32(2): 169–84 e7.
- 26Li CW, Lim SO, Xia W, Lee HH, Chan LC, Kuo CW, et al. Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat Commun. 2016; 7:12632.
- 27Burr ML, Sparbier CE, Chan YC, Williamson JC, Woods K, Beavis PA, et al. CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity. Nature. 2017; 549(7670): 101–05.
- 28Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science. 2017; 355(6332): 1428–33.
- 29Zhang Q, Zhang Y, Chen Y, Qian J, Zhang X, Yu K. A Novel mTORC1/2 Inhibitor (MTI-31) Inhibits Tumor Growth, Epithelial-Mesenchymal Transition, Metastases, and Improves Antitumor Immunity in Preclinical Models of Lung Cancer. Clin Cancer Res. 2019; 25(12): 3630–42.
- 30Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012 Mar 4; 9(4): 357–9.
- 31Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 2008; 9(9): R137.
- 32Shen L, Shao NY, Liu X, Maze I, Feng J, Nestler EJ. diffReps: detecting differential chromatin modification sites from ChIP-seq data with biological replicates. PLoS One. 2013; 8(6):e65598.
- 33Buenrostro JD, Wu B, Chang HY, Greenleaf WJ. ATAC-seq: A Method for Assaying Chromatin Accessibility Genome-Wide. Curr Protoc Mol Biol. 2015; 109: 21 29 1-21 29 9.
- 34Zhang P, Liu Y, Lian C, Cao X, Wang Y, Li X, et al. SH3RF3 promotes breast cancer stem-like properties via JNK activation and PTX3 upregulation. Nat Commun. 2020; 11(1): 2487.
- 35Lin S, Huang G, Cheng L, Li Z, Xiao Y, Deng Q, et al. Establishment of peripheral blood mononuclear cell-derived humanized lung cancer mouse models for studying efficacy of PD-L1/PD-1 targeted immunotherapy. MAbs. 2018; 10(8): 1301–11.
- 36Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. American Society of Clinical Oncology/College Of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol. 2010; 28(16): 2784–95.
- 37Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, et al. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. J Clin Oncol. 2018; 36(20): 2105–22.
- 38Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, et al. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J Clin. 2017; 67(2): 93–99.
- 39Giuliano AE, Edge SB, Hortobagyi GN. Eighth Edition of the AJCC Cancer Staging Manual: Breast Cancer. Ann Surg Oncol. 2018; 25(7): 1783–85.
- 40Cao J, Wang B, Zhang J, Tao Z, Wang L, Hu X. Phase 1b clinical trial of pucotenlimab (HX008), a novel anti-PD-1 monoclonal antibody, combined with gemcitabine and cisplatin in the first-line treatment of metastatic triple-negative breast cancer. Front Oncol. 2022; 12:837963.
- 41Guo H, Ding Q, Gong Y, Gilcrease MZ, Zhao M, Zhao J, et al. Comparison of three scoring methods using the FDA-approved 22C3 immunohistochemistry assay to evaluate PD-L1 expression in breast cancer and their association with clinicopathologic factors. Breast Cancer Res. 2020; 22(1): 69.
- 42Ikarashi D, Kitano S, Tsuyukubo T, Takenouchi K, Nakayama T, Onagi H, et al. Pretreatment tumour immune microenvironment predicts clinical response and prognosis of muscle-invasive bladder cancer in the neoadjuvant chemotherapy setting. Br J Cancer. 2022; 126(4): 606–14.
- 43Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005; 102(43): 15545–50.
- 44Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P, Mesirov JP. Molecular signatures database (MSigDB) 3.0. Bioinformatics. 2011; 27(12): 1739–40.
- 45Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 2017; 127(8): 2930–40.
- 46Qin Y, Ekmekcioglu S, Forget MA, Szekvolgyi L, Hwu P, Grimm EA, et al. Cervical Cancer Neoantigen Landscape and Immune Activity is Associated with Human Papillomavirus Master Regulators. Front Immunol. 2017; 8: 689.
- 47Yim SY, Kang SH, Shin JH, Jeong YS, Sohn BH, Um SH, et al. Low ARID1A Expression is Associated with Poor Prognosis in Hepatocellular Carcinoma. Cells. 2020; 9(9): 2002
- 48Mehrvarz Sarshekeh A, Alshenaifi J, Roszik J, Manyam GC, Advani SM, Katkhuda R, et al. ARID1A Mutation May Define an Immunologically Active Subgroup in Patients with Microsatellite Stable Colorectal Cancer. Clin Cancer Res. 2021; 27(6): 1663–70.
- 49Colaprico 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.
- 50Chen B, Khodadoust MS, Liu CL, Newman AM, Alizadeh AA. Profiling Tumor Infiltrating Immune Cells with CIBERSORT. Methods Mol Biol. 2018; 1711: 243–59.
- 51Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The Immune Landscape of Cancer. Immunity. 2018; 48(4): 812–30 e14.
- 52Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells. Cancer Res. 2017; 77(21): e108–e10.
- 53Picelli S, Faridani OR, Bjorklund AK, Winberg G, Sagasser S, Sandberg R. Full-length RNA-seq from single cells using Smart-seq2. Nat Protoc. 2014; 9(1): 171–81.
- 54Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001; 344(11): 783–92.
- 55Burstein HJ. The distinctive nature of HER2-positive breast cancers. N Engl J Med. 2005; 353(16): 1652–4.
- 56Cameron D, Piccart-Gebhart MJ, Gelber RD, Procter M, Goldhirsch A, de Azambuja E, et al. 11 years' follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: final analysis of the HERceptin Adjuvant (HERA) trial. Lancet. 2017; 389(10075): 1195–205.
- 57Kim TK, Vandsemb EN, Herbst RS, Chen L. Adaptive immune resistance at the tumour site: mechanisms and therapeutic opportunities. Nat Rev Drug Discov. 2022; 21(7): 529–40.
- 58Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000; 192(7): 1027–34.
- 59Kim YB, Ahn JM, Bae WJ, Sung CO, Lee D. Functional loss of ARID1A is tightly associated with high PD-L1 expression in gastric cancer. Int J Cancer. 2019; 145(4): 916–26.
- 60Kamori T, Oki E, Shimada Y, Hu Q, Hisamatsu Y, Ando K, et al. The effects of ARID1A mutations on colorectal cancer and associations with PD-L1 expression by stromal cells. Cancer Rep (Hoboken). 2022; 5(1):e1420.
- 61Li N, Liu Q, Han Y, Pei S, Cheng B, Xu J, et al. ARID1A loss induces polymorphonuclear myeloid-derived suppressor cell chemotaxis and promotes prostate cancer progression. Nat Commun. 2022; 13(1): 7281.
- 62Schick S, Rendeiro AF, Runggatscher K, Ringler A, Boidol B, Hinkel M, et al. Systematic characterization of BAF mutations provides insights into intracomplex synthetic lethalities in human cancers. Nat Genet. 2019; 51(9): 1399–410.
- 63Kelso TWR, Porter DK, Amaral ML, Shokhirev MN, Benner C, Hargreaves DC. Chromatin accessibility underlies synthetic lethality of SWI/SNF subunits in ARID1A-mutant cancers. Elife. 2017; 6.
- 64Liu X, Li Z, Wang Z, Liu F, Zhang L, Ke J, et al. Chromatin Remodeling Induced by ARID1A Loss in Lung Cancer Promotes Glycolysis and Confers JQ1 Vulnerability. Cancer Res. 2022; 82(5): 791–804.
- 65Box JK, Paquet N, Adams MN, Boucher D, Bolderson E, O'Byrne KJ, et al. Nucleophosmin: from structure and function to disease development. BMC Mol Biol. 2016; 17(1): 19.
- 66Qin G, Wang X, Ye S, Li Y, Chen M, Wang S, et al. NPM1 upregulates the transcription of PD-L1 and suppresses T cell activity in triple-negative breast cancer. Nat Commun. 2020; 11(1): 1669.
- 67De S, Babu MM. Genomic neighbourhood and the regulation of gene expression. Curr Opin Cell Biol. 2010; 22(3): 326–33.
- 68Lee JB, Park HS, Choi SJ, Heo SG, An HJ, Kim HR, et al. Plasma tumor mutation burden is associated with clinical benefit in patients with non-small cell lung cancer treated with anti-programmed death-1 monotherapy. Ther Adv Med Oncol. 2022; 14:17588359221141761.
- 69Wilke CM, Wei S, Wang L, Kryczek I, Kao J, Zou W. Dual biological effects of the cytokines interleukin-10 and interferon-gamma. Cancer Immunol Immunother. 2011; 60(11): 152figur–41.
- 70Gao Y, Yang J, Cai Y, Fu S, Zhang N, Fu X, et al. IFN-gamma-mediated inhibition of lung cancer correlates with PD-L1 expression and is regulated by PI3K-AKT signaling. Int J Cancer. 2018; 143(4): 931–43.
- 71Parsa AT, Waldron JS, Panner A, Crane CA, Parney IF, Barry JJ, et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2007; 13(1): 84–8.
- 72Casey SC, Tong L, Li Y, Do R, Walz S, Fitzgerald KN, et al. MYC regulates the antitumor immune response through CD47 and PD-L1. Science. 2016; 352(6282): 227–31.
- 73Dorand RD, Nthale J, Myers JT, Barkauskas DS, Avril S, Chirieleison SM, et al. Cdk5 disruption attenuates tumor PD-L1 expression and promotes antitumor immunity. Science. 2016; 353(6297): 399–403.
- 74Wu SY, Xu Y, Chen L, Fan L, Ma XY, Zhao S, et al. Combined angiogenesis and PD-1 inhibition for immunomodulatory TNBC: concept exploration and biomarker analysis in the FUTURE-C-Plus trial. Mol Cancer. 2022; 21(1): 84.