Comprehensive Evaluation of the m6A Regulator Prognostic Risk Score in the Prediction of Immunotherapy Response in Clear Cell Renal Cell Carcinoma

Mingke Yu, Xuefei Liu, Han Xu, Sangyu Shen, Fajiu Wang, Dajin Chen, Guorong Li, Zongping Wang, Zhixiang Zuo, An Zhao, Mingke Yu, Xuefei Liu, Han Xu, Sangyu Shen, Fajiu Wang, Dajin Chen, Guorong Li, Zongping Wang, Zhixiang Zuo, An Zhao

Abstract

Background: Clear cell renal cell carcinoma (ccRCC) is known for its high drug resistance. The tumor-immune crosstalk mediated by the epigenetic regulation of N6-methyladenosine (m6A) modification has been demonstrated in recent studies. Therefore, m6A modification-mediated immune cell infiltration characteristics may be helpful to guide immunotherapy for ccRCC.

Methods: This study comprehensively analyzed m6A modifications using the clinical parameters, single-cell RNA sequencing data, and bulk RNA sequencing data from the TCGA-ccRC cohort and 13 external validation cohorts. A series of bioinformatic approaches were applied to construct an m6A regulator prognostic risk score (MRPRS) to predict survival and immunotherapy response in ccRCC patients. Immunological characteristics, enriched pathways, and mutation were evaluated in high- and low-MRPRS groups.

Results: The expressional alteration landscape of m6A regulators was profiled in ccRCC cell clusters and tissue. The 8 regulator genes with minimal lambda were integrated to build an MRPRS, and it was positively correlated with immunotherapeutic response in extent validation cohorts. The clinicopathological features and immune infiltration characteristics could be distinguished by the high- and low-MRPRS. Moreover, the MRPRS-mediated mutation pattern has an enhanced response to immune checkpoint blockade in the ccRCC and pan-cancer cohorts.

Conclusions: The proposed MRPRS is a promising biomarker to predict clinical outcomes and therapeutic responses in ccRCC patients.

Keywords: N6-methyladenosine; clear cell renal cell carcinoma; immune infiltration characteristic; immunotherapy; mutation; prognosis.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Yu, Liu, Xu, Shen, Wang, Chen, Li, Wang, Zuo and Zhao.

Figures

Figure 1
Figure 1
(A) Expression of 23 m6A RNA methylation regulators between renal cancer and normal tissues in the TCGA-ccRCC cohort. (B, C) The UMAP plot and overview of epithelial cells by the origin and cell type of the cells. (D) Composition of various epithelial cells in different immunotherapeutic responses. (E) The heatmap of marker gene expression in 7 identified epithelial cell subsets. (F) Dot plot analysis of KEGG pathway enrichment of 7 epithelial cell subsets. (G) Violin plots showing the partial expression of m6A regulators for each epithelial cell type ("*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001; ns, not significant).
Figure 2
Figure 2
(A) LASSO coefficient profiles of the 23 m6A RNA methylation regulators in the TCGA-ccRCC cohort. (B) The prognostic analyses for 23 m6A RNA methylation regulators in the TCGA-ccRCC cohort using the univariate Cox regression model. (C) Kaplan–Meier analysis of patients between high- and low-MRPRS groups in the TCGA-ccRCC cohort. (D) Validation cohort of MRPRS from GSE29609. (E) Box plot of different MRPRSs in 3 epithelial cell subsets. (F) The MRPRS between response and non-response groups in PMID32472114. (G–I) Kaplan–Meier analysis of three validation cohorts of immunotherapy in ccRCC (PMID32472114, PMID29301960, and PMID32895571). (J) Heatmap of chemotherapy and targeted drug-related genes between high- and low-MRPRS groups. (K) The correlation of MRPRS and genes associated with angiogenesis. (L) The MRPRS between response and non-response groups in ccRCC with sunitinib (E-MTAB-3267). (M) Kaplan–Meier analysis between high- and low-MRPRS groups in ccRCC with sunitinib (E-MTAB-3267) (*p < 0.05; **p < 0.01; ***p < 0.001).
Figure 3
Figure 3
(A) Box plot of the relationship between stage T, N, M, grade, and MRPRS. (B) The heatmap of markers on multiple immune infiltrates. (C) The MCP_counter algorithm was used to estimate the abundance of various types of immune cells between high- and low-MRPRS groups. (D) The abundance and diversity of TCR clone in high- and low-MRPRS groups. (E) Crosstalk between immune cells and epithelial cells (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001).
Figure 4
Figure 4
(A) Waterfall plot of the distribution of mutations found in the high- and low-MRPRS groups of the TCGA-ccRCC cohort. (B) The top 5 genes of high- vs. low-MRPRS group mutation status. (C) Lollipop plot of somatic mutations in SETD2, TRIOBP, RYR2, ZFPM2, and ABCC6. (D, E) MCP_counter and ssGSEA algorithm were used to estimate the abundance of various types of immune cells in high- and low-MRPRS groups (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001; ns, not significant).
Figure 5
Figure 5
(A) Different distribution ratio of response and non-response in the immunotherapeutic cohort of ccRCC. (B) Kaplan–Meier analysis of patients in the mutated and non-mutated groups in the immunotherapeutic cohort of ccRCC (Van_2018, Morris_2019, PMID29337640 and PMID29301960). (C) The composition of major cancer types in the immunotherapeutic cohort contains mutations of pan-cancer. (D, E) Kaplan–Meier analysis (OS and PFS) of patients in the mutation and non-mutation groups in the immunotherapeutic cohort of pan-cancer. (F) Kaplan–Meier analysis of patients in the low TMB of mutated, low TMB of non-mutated, and high TMB groups.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2020. CA Cancer J Clin (2020) 70(1):7–30. doi: 10.3322/caac.21590
    1. Li QK, Pavlovich CP, Zhang H, Kinsinger CR, Chan DW. Challenges and Opportunities in the Proteomic Characterization of Clear Cell Renal Cell Carcinoma (ccRCC): A Critical Step Towards the Personalized Care of Renal Cancers. Semin Cancer Biol (2019) 55:8–15. doi: 10.1016/j.semcancer.2018.06.004
    1. Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, et al. . Pembrolizumab Plus Axitinib Versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med (2019) 380(12):1116–27. doi: 10.1056/NEJMoa1816714
    1. Rini BI, Powles T, Atkins MB, Escudier B, McDermott DF, Suarez C, et al. . Atezolizumab Plus Bevacizumab Versus Sunitinib in Patients With Previously Untreated Metastatic Renal Cell Carcinoma (IMmotion151): A Multicentre, Open-Label, Phase 3, Randomised Controlled Trial. Lancet (2019) 393(10189):2404–15. doi: 10.1016/S0140-6736(19)30723-8
    1. Zaccara S, Ries RJ, Jaffrey SR. Reading, Writing and Erasing mRNA Methylation. Nat Rev Mol Cell Biol (2019) 20(10):608–24. doi: 10.1038/s41580-019-0168-5
    1. Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, et al. . Recognition of RNA N(6)-Methyladenosine by IGF2BP Proteins Enhances mRNA Stability and Translation. Nat Cell Biol (2018) 20(3):285–95. doi: 10.1038/s41556-018-0045-z
    1. Zhao X, Yang Y, Sun BF, Shi Y, Yang X, Xiao W, et al. . FTO-Dependent Demethylation of N6-Methyladenosine Regulates mRNA Splicing and is Required for Adipogenesis. Cell Res (2014) 24(12):1403–19. doi: 10.1038/cr.2014.151
    1. Lin S, Choe J, Du P, Triboulet R, Gregory RI. The M(6)A Methyltransferase METTL3 Promotes Translation in Human Cancer Cells. Mol Cell (2016) 62(3):335–45. doi: 10.1016/j.molcel.2016.03.021
    1. Liu J, Eckert MA, Harada BT, Liu SM, Lu Z, Yu K, et al. . M(6)A mRNA Methylation Regulates AKT Activity to Promote the Proliferation and Tumorigenicity of Endometrial Cancer. Nat Cell Biol (2018) 20(9):1074–83. doi: 10.1038/s41556-018-0174-4
    1. Wang H, Hu X, Huang M, Liu J, Gu Y, Ma L, et al. . Mettl3-Mediated mRNA M(6)A Methylation Promotes Dendritic Cell Activation. Nat Commun (2019) 10(1):1898. doi: 10.1038/s41467-019-09903-6
    1. Qiu X, Yang S, Wang S, Wu J, Zheng B, Wang K, et al. . M6A Demethylase ALKBH5 Regulates PD-L1 Expression and Tumor Immunoenvironment in Intrahepatic Cholangiocarcinoma. Cancer Res (2021) 81(18):4778–93. doi: 10.1158/0008-5472.CAN-21-0468
    1. von Roemeling CA, Radisky DC, Marlow LA, Cooper SJ, Grebe SK, Anastasiadis PZ, et al. . Neuronal Pentraxin 2 Supports Clear Cell Renal Cell Carcinoma by Activating the AMPA-Selective Glutamate Receptor-4. Cancer Res (2014) 74(17):4796–810. doi: 10.1158/0008-5472.CAN-14-0210
    1. Wozniak MB, Le Calvez-Kelm F, Abedi-Ardekani B, Byrnes G, Durand G, Carreira C, et al. . Integrative Genome-Wide Gene Expression Profiling of Clear Cell Renal Cell Carcinoma in Czech Republic and in the United States. PLoS One (2013) 8(3):e57886. doi: 10.1371/journal.pone.0057886
    1. Edeline J, Mottier S, Vigneau C, Jouan F, Perrin C, Zerrouki S, et al. . Description of 2 Angiogenic Phenotypes in Clear Cell Renal Cell Carcinoma. Hum Pathol (2012) 43(11):1982–90. doi: 10.1016/j.humpath.2012.01.023
    1. Beuselinck B, Job S, Becht E, Karadimou A, Verkarre V, Couchy G, et al. . Molecular Subtypes of Clear Cell Renal Cell Carcinoma are Associated With Sunitinib Response in the Metastatic Setting. Clin Cancer Res (2015) 21(6):1329–39. doi: 10.1158/1078-0432.CCR-14-1128
    1. Braun DA, Hou Y, Bakouny Z, Ficial M, Sant' Angelo M, Forman J, et al. . Interplay of Somatic Alterations and Immune Infiltration Modulates Response to PD-1 Blockade in Advanced Clear Cell Renal Cell Carcinoma. Nat Med (2020) 26(6):909–18. doi: 10.1038/s41591-020-0839-y
    1. Miao D, Margolis CA, Gao W, Voss MH, Li W, Martini DJ, et al. . Genomic Correlates of Response to Immune Checkpoint Therapies in Clear Cell Renal Cell Carcinoma. Science (2018) 359(6377):801–6. doi: 10.1126/science.aan5951
    1. Motzer RJ, Robbins PB, Powles T, Albiges L, Haanen JB, Larkin J, et al. . Avelumab Plus Axitinib Versus Sunitinib in Advanced Renal Cell Carcinoma: Biomarker Analysis of the Phase 3 JAVELIN Renal 101 Trial. Nat Med (2020) 26(11):1733–41. doi: 10.1038/s41591-020-1044-8
    1. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. . TGFbeta Attenuates Tumour Response to PD-L1 Blockade by Contributing to Exclusion of T Cells. Nature (2018) 554(7693):544–8. doi: 10.1038/nature25501
    1. Gide TN, Quek C, Menzies AM, Tasker AT, Shang P, Holst J, et al. . Distinct Immune Cell Populations Define Response to Anti-PD-1 Monotherapy and Anti-PD-1/Anti-CTLA-4 Combined Therapy. Cancer Cell (2019) 35(2):238–55.e6. doi: 10.1016/j.ccell.2019.01.003
    1. Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, et al. . Genomic Correlates of Response to CTLA-4 Blockade in Metastatic Melanoma. Science (2015) 350(6257):207–11. doi: 10.1126/science.aad0095
    1. Miao D, Margolis CA, Vokes NI, Liu D, Taylor-Weiner A, Wankowicz SM, et al. . Genomic Correlates of Response to Immune Checkpoint Blockade in Microsatellite-Stable Solid Tumors. Nat Genet (2018) 50(9):1271–81. doi: 10.1038/s41588-018-0200-2
    1. Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. . Tumor Mutational Load Predicts Survival After Immunotherapy Across Multiple Cancer Types. Nat Genet (2019) 51(2):202–6. doi: 10.1038/s41588-018-0312-8
    1. Rizvi H, Sanchez-Vega F, La K, Chatila W, Jonsson P, Halpenny D, et al. . Molecular Determinants of Response to Anti-Programmed Cell Death (PD)-1 and Anti-Programmed Death-Ligand 1 (PD-L1) Blockade in Patients With Non-Small-Cell Lung Cancer Profiled With Targeted Next-Generation Sequencing. J Clin Oncol (2018) 36(7):633–41. doi: 10.1200/JCO.2017.75.3384
    1. Krishna C, DiNatale RG, Kuo F, Srivastava RM, Vuong L, Chowell D, et al. . Single-Cell Sequencing Links Multiregional Immune Landscapes and Tissue-Resident T Cells in ccRCC to Tumor Topology and Therapy Efficacy. Cancer Cell (2021) 39(5):662–77.e6. doi: 10.1016/j.ccell.2021.03.007
    1. Efremova M, Vento-Tormo M, Teichmann SA, Vento-Tormo R. CellPhoneDB: Inferring Cell-Cell Communication From Combined Expression of Multi-Subunit Ligand-Receptor Complexes. Nat Protoc (2020) 15(4):1484–506. doi: 10.1038/s41596-020-0292-x
    1. Li H, Hu J, Yu A, Othmane B, Guo T, Liu J, et al. . RNA Modification of N6-Methyladenosine Predicts Immune Phenotypes and Therapeutic Opportunities in Kidney Renal Clear Cell Carcinoma. Front Oncol (2021) 11:642159. doi: 10.3389/fonc.2021.642159
    1. Chen L, Hu B, Song X, Wang L, Ju M, Li Z, et al. . M(6)A RNA Methylation Regulators Impact Prognosis and Tumor Microenvironment in Renal Papillary Cell Carcinoma. Front Oncol (2021) 11:598017. doi: 10.3389/fonc.2021.598017
    1. Huang H, Weng H, Chen J. M(6)A Modification in Coding and Non-Coding RNAs: Roles and Therapeutic Implications in Cancer. Cancer Cell (2020) 37(3):270–88. doi: 10.1016/j.ccell.2020.02.004
    1. Chen XY, Zhang J, Zhu JS. The Role of M(6)A RNA Methylation in Human Cancer. Mol Cancer (2019) 18(1):103. doi: 10.1186/s12943-019-1033-z
    1. Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, et al. . STRING V10: Protein-Protein Interaction Networks, Integrated Over the Tree of Life. Nucleic Acids Res (2015) 43:D447–52. doi: 10.1093/nar/gku1003
    1. Diaz-Montero CM, Rini BI, Finke JH. The Immunology of Renal Cell Carcinoma. Nat Rev Nephrol (2020) 16(12):721–35. doi: 10.1038/s41581-020-0316-3
    1. Thommen DS, Schumacher TN. T Cell Dysfunction in Cancer. Cancer Cell (2018) 33(4):547–62. doi: 10.1016/j.ccell.2018.03.012
    1. He QF, Xu Y, Li J, Huang ZM, Li XH, Wang X. CD8+ T-Cell Exhaustion in Cancer: Mechanisms and New Area for Cancer Immunotherapy. Brief Funct Genomics (2019) 18(2):99–106. doi: 10.1093/bfgp/ely006
    1. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. . Nivolumab Versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med (2015) 373(19):1803–13. doi: 10.1056/NEJMoa1510665
    1. Shulman Z, Stern-Ginossar N. The RNA Modification N(6)-Methyladenosine as a Novel Regulator of the Immune System. Nat Immunol (2020) 21(5):501–12. doi: 10.1038/s41590-020-0650-4
    1. Helmink BA, Reddy SM, Gao J, Zhang S, Basar R, Thakur R, et al. . B Cells and Tertiary Lymphoid Structures Promote Immunotherapy Response. Nature (2020) 577(7791):549–55. doi: 10.1038/s41586-019-1922-8
    1. Cabrita R, Lauss M, Sanna A, Donia M, Skaarup Larsen M, Mitra S, et al. . Tertiary Lymphoid Structures Improve Immunotherapy and Survival in Melanoma. Nature (2020) 577(7791):561–5. doi: 10.1038/s41586-019-1914-8
    1. Sanmamed MF, Chen L. A Paradigm Shift in Cancer Immunotherapy: From Enhancement to Normalization. Cell (2018) 175(2):313–26. doi: 10.1016/j.cell.2018.09.035
    1. Ji RR, Chasalow SD, Wang L, Hamid O, Schmidt H, Cogswell J, et al. . An Immune-Active Tumor Microenvironment Favors Clinical Response to Ipilimumab. Cancer Immunol Immunother (2012) 61(7):1019–31. doi: 10.1007/s00262-011-1172-6
    1. Gajewski TF, Corrales L, Williams J, Horton B, Sivan A, Spranger S. Cancer Immunotherapy Targets Based on Understanding the T Cell-Inflamed Versus Non-T Cell-Inflamed Tumor Microenvironment. Adv Exp Med Biol (2017) 1036:19–31. doi: 10.1007/978-3-319-67577-0_2
    1. Conway JR, Kofman E, Mo SS, Elmarakeby H, Van Allen E. Genomics of Response to Immune Checkpoint Therapies for Cancer: Implications for Precision Medicine. Genome Med (2018) 10(1):93. doi: 10.1186/s13073-018-0605-7
    1. Hellmann MD, Ciuleanu TE, Pluzanski A, Lee JS, Otterson GA, Audigier-Valette C, et al. . Nivolumab Plus Ipilimumab in Lung Cancer With a High Tumor Mutational Burden. N Engl J Med (2018) 378(22):2093–104. doi: 10.1056/NEJMoa1801946
    1. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. . Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N Engl J Med (2014) 371(23):2189–99. doi: 10.1056/NEJMoa1406498
    1. McGrail DJ, Pilie PG, Rashid NU, Voorwerk L, Slagter M, Kok M, et al. . High Tumor Mutation Burden Fails to Predict Immune Checkpoint Blockade Response Across All Cancer Types. Ann Oncol (2021) 32(5):661–72. doi: 10.1016/j.annonc.2021.02.006

Source: PubMed

3
Subscribe