Journal Home > Volume 1 , Issue 2
Background

Cancer metastasis and recurrence remain major challenges in renal carcinoma patient management. There are limited biomarkers to predict the metastatic probability of renal cancer, especially in the early‐stage subgroup. Here, our study applied robust machine‐learning algorithms to identify metastatic and recurrence‐related signatures across multiple renal cancer cohorts, which reached high accuracy in both training and testing cohorts.

Methods

Clear cell renal cell carcinoma (ccRCC) patients with primary or metastatic site sequencing information from eight cohorts, including one out‐house cohort, were enrolled in this study. Three robust machine‐learning algorithms were applied to identify metastatic signatures. Then, two distinct metastatic‐related subtypes were identified and verified; matrix remodeling associated 5 (MXRA5), as a promising diagnostic and therapeutic target, was investigated in vivo and in vitro.

Results

We identified five stable metastasis‐related signatures (renin, integrin subunit beta‐like 1, MXRA5, mesenchyme homeobox 2, and anoctamin 3) from multicenter cohorts. Additionally, we verified the specificity and sensibility of these signatures in external and out‐house cohorts, which displayed a satisfactory consistency. According to these metastatic signatures, patients were grouped into two distinct and heterogeneous ccRCC subtypes named metastatic cancer subtype 1 (MTCS1) and type 2 (MTCS2). MTCS2 exhibited poorer clinical outcomes and metastatic tendencies than MTCS1. In addition, MTCS2 showed higher immune cell infiltration and immune signature expression but a lower response rate to immune blockade therapy than MTCS1. The MTCS2 subgroup was more sensitive to saracatinib, sunitinib, and several molecular targeted drugs. In addition, MTCS2 displayed a higher genome mutation burden and instability. Furthermore, we constructed a prognosis model based on subtype biomarkers, which performed well in training and validation cohorts. Finally, MXRA5, as a promising biomarker, significantly suppressed malignant ability, including the cell migration and proliferation of ccRCC cell lines in vitro and in vivo.

Conclusions

This study identified five robust metastatic signatures and proposed two metastatic probability clusters with stratified prognoses, multiomics landscapes, and treatment options. The current work not only provided new insight into the heterogeneity of renal cancer but also shed light on optimizing decision‐making in immunotherapy and chemotherapy.


menu
Abstract
Full text
Outline
About this article

Definition and verification of novel metastasis and recurrence related signatures of ccRCC: A multicohort study

Show Author's information Aimin Jiang1Qingyang Pang1Xinxin Gan1Anbang Wang2Zhenjie Wu1Bing Liu3Peng Luo4( )Le Qu5( )Linhui Wang1 ( )
Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
Department of Urology, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
Department of Urology, The Third Affiliated Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China

Abstract

Background

Cancer metastasis and recurrence remain major challenges in renal carcinoma patient management. There are limited biomarkers to predict the metastatic probability of renal cancer, especially in the early‐stage subgroup. Here, our study applied robust machine‐learning algorithms to identify metastatic and recurrence‐related signatures across multiple renal cancer cohorts, which reached high accuracy in both training and testing cohorts.

Methods

Clear cell renal cell carcinoma (ccRCC) patients with primary or metastatic site sequencing information from eight cohorts, including one out‐house cohort, were enrolled in this study. Three robust machine‐learning algorithms were applied to identify metastatic signatures. Then, two distinct metastatic‐related subtypes were identified and verified; matrix remodeling associated 5 (MXRA5), as a promising diagnostic and therapeutic target, was investigated in vivo and in vitro.

Results

We identified five stable metastasis‐related signatures (renin, integrin subunit beta‐like 1, MXRA5, mesenchyme homeobox 2, and anoctamin 3) from multicenter cohorts. Additionally, we verified the specificity and sensibility of these signatures in external and out‐house cohorts, which displayed a satisfactory consistency. According to these metastatic signatures, patients were grouped into two distinct and heterogeneous ccRCC subtypes named metastatic cancer subtype 1 (MTCS1) and type 2 (MTCS2). MTCS2 exhibited poorer clinical outcomes and metastatic tendencies than MTCS1. In addition, MTCS2 showed higher immune cell infiltration and immune signature expression but a lower response rate to immune blockade therapy than MTCS1. The MTCS2 subgroup was more sensitive to saracatinib, sunitinib, and several molecular targeted drugs. In addition, MTCS2 displayed a higher genome mutation burden and instability. Furthermore, we constructed a prognosis model based on subtype biomarkers, which performed well in training and validation cohorts. Finally, MXRA5, as a promising biomarker, significantly suppressed malignant ability, including the cell migration and proliferation of ccRCC cell lines in vitro and in vivo.

Conclusions

This study identified five robust metastatic signatures and proposed two metastatic probability clusters with stratified prognoses, multiomics landscapes, and treatment options. The current work not only provided new insight into the heterogeneity of renal cancer but also shed light on optimizing decision‐making in immunotherapy and chemotherapy.

Keywords: machine learning, metastasis, recurrence, clear cell renal cell carcinoma, multiple omics, single‐cell sequencing

References(73)

Capitanio U, Montorsi F. Renal cancer. Lancet. 2016;387(10021):894–906.
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.
The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013;499(7456):439. 10.1038/nature12222
Kumar R, Kapoor A. Current management of metastatic renal cell carcinoma: evolving new therapies. Curr Opin Supportive Palliative Care. 2017;11(3):2317. 10.1097/SPC.0000000000000277
Heldwein FL, Escudier B, Smyth G, Souto CA, Vallancien G. Metastatic renal cell carcinoma management. Int Braz J Urol. 2009;35(3):25670. 10.1590/S1677-55382009000300002
Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, et al. Renal cell carcinoma. Nat Rev Dis Primers. 2017;3(1):1–19.
Signoretti S, Flaifel A, Chen YB, Reuter VE. Renal cell carcinoma in the era of precision medicine: from molecular pathology to tissue‐based biomarkers. J Clin Oncol. 2018;36(36):3553–9.
Guo H, German P, Bai S, Barnes S, Guo W, Qi X, et al. The PI3K/AKT pathway and renal cell carcinoma. J Genet Genomics. 2015;42(7):343–53.
Clark DJ, Dhanasekaran SM, Petralia F, Pan J, Song X, Hu Y, et al. Integrated proteogenomic characterization of clear cell renal cell carcinoma. Cell. 2019;179(4):964–83.
Jiang A, Bao Y, Wang A, Gong W, Gan X, Wang J, et al. Establishment of a prognostic prediction and drug selection model for patients with clear cell renal cell carcinoma by multiomics data analysis. Oxid Med Cell Longevity. 2022;2022:e3617775.
Ricketts CJ, De Cubas AA, Fan H, Smith CC, Lang M, Reznik E, et al. The cancer genome Atlas comprehensive molecular characterization of renal cell carcinoma. Cell Rep. 2018;23(12):3698.
El‐Mokadem I, Fitzpatrick J, Bondad J, Rauchhaus P, Cunningham J, Pratt N, et al. Chromosome 9p deletion in clear cell renal cell carcinoma predicts recurrence and survival following surgery. Br J Cancer. 2014;111(7):1381–90.
Bao Y, Jiang A, Dong K, Gan X, Gong W, Wu Z, et al. DDX39 as a predictor of clinical prognosis and immune checkpoint therapy efficacy in patients with clear cell renal cell carcinoma. Int J Biol Sci. 2021;17(12):3158–72.
Zheng Q, Li P, Zhou X, Qiang Y, Fan J, Lin Y, et al. Deficiency of the X‐inactivation escaping gene KDM5C in clear cell renal cell carcinoma promotes tumorigenicity by reprogramming glycogen metabolism and inhibiting ferroptosis. Theranostics. 2021;11(18):8674–91.
D'alterio C, Cindolo L, Portella L, Polimeno M, Consales C, Riccio A, et al. Differential role of CD133 and CXCR4 in renal cell carcinoma. Cell Cycle. 2010;9(22):4492–500.
Tiedemann RL, Hlady RA, Hanavan PD, Lake DF, Tibes R, Lee JH, et al. Dynamic reprogramming of DNA methylation in SETD2‐deregulated renal cell carcinoma. Oncotarget. 2016;7(2):1927–46.
Chen X. Expression of microRNA‐3133 correlates with the prognosis in patients with clear cell renal cell carcinoma. Medicine. 2019;98(24):e16008.
Toriihara A, Duan H, Thompson HM, Park S, Hatami N, Baratto L, et al. 18F‐FPPRGD2 PET/CT in patients with metastatic renal cell cancer. Eur J Nucl Med Mol Imaging. 2019;46(7):1518–23.
Lightfoot N, Conlon M, Kreiger N, Bissett R, Desai M, Warde P, et al. Impact of noninvasive imaging on increased incidental detection of renal cell carcinoma. Eur Urol. 2000;37(5):521–7.
Liu P, Tian W. Identification of DNA methylation patterns and biomarkers for clear‐cell renal cell carcinoma by multi‐omics data analysis. PeerJ. 2020;8:e9654.
Nam H‐Y, Chandrashekar DS, Kundu A, Shelar S, Kho EY, Sonpavde G, et al. Integrative epigenetic and gene expression analysis of renal tumor progression to metastasis. Mol Cancer Res. 2019;17(1):84–96.
Wuttig D, Zastrow S, Füssel S, Toma MI, Meinhardt M, Kalman K, et al. CD31, EDNRB and TSPAN7 are promising prognostic markers in clear‐cell renal cell carcinoma revealed by genome‐wide expression analyses of primary tumors and metastases. Int J Cancer. 2012;131(5):E693–704.
López‐Lago MA, Thodima VJ, Guttapalli A, Chan T, Heguy A, Molina AM, et al. Genomic deregulation during metastasis of renal cell carcinoma implements a myofibroblast‐like program of gene expression. Cancer Res. 2010;70(23):9682–92.
Ho TH, Serie DJ, Parasramka M, Cheville JC, Bot BM, Tan W, et al. Differential gene expression profiling of matched primary renal cell carcinoma and metastases reveals upregulation of extracellular matrix genes. Ann Oncol. 2017;28(3):60410. 10.1093/annonc/mdw652
Braun DA, Street K, Burke KP, Cookmeyer DL, Denize T, Pedersen CB, et al. Progressive immune dysfunction with advancing disease stage in renal cell carcinoma. Cancer Cell. 2021;39(5):632–48.
Tomczak K, Czerwińska P, Wiznerowicz M. The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge. Contemp Oncol. 2015;19(1A):A68–77.
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.
Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16(5):284–7.
Liberzon A, Birger C, Thorvaldsdóttir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Systems. 2015;1(6):417–25.
Kamburov A, Wierling C, Lehrach H, Herwig R. ConsensusPathDB—a database for integrating human functional interaction networks. Nucleic Acids Res. 2009;37:D623–8.
Li 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):e108–10.
Chen B, Khodadoust MS, Liu CL, Newman AM, Alizadeh AA. Profiling tumor infiltrating immune cells with CIBERSORT. Methods Mol Biol. 2018;1711:243–59.
Finotello F, Mayer C, Plattner C, Laschober G, Rieder D, Hackl H, et al. Molecular and pharmacological modulators of the tumor immune contexture revealed by deconvolution of RNA‐seq data. Genome Med. 2019;11:34.
Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 2017;18(1):220.
Racle J, Gfeller D. EPIC: a tool to estimate the proportions of different cell types from bulk gene expression data. Methods Mol Biol. 2020;2120:23348. 10.1007/978-1-0716-0327-7_17
Hänzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA‐seq data. BMC Bioinformatics. 2013;14:7.
Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nature Med. 2018;24(10):1550–8.
Mayakonda A, Lin DC, Assenov Y, Plass C, Koeffler HP. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res. 2018;28(11):1747–56.
Mermel CH, Schumacher SE, Hill B, Meyerson ML, Beroukhim R, Getz G. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy‐number alteration in human cancers. Genome Biol. 2011;12(4):R41.
Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513(7517):2029. 10.1038/nature13480
Reinhold WC, Sunshine M, Liu H, Varma S, Kohn KW, Morris J, et al. CellMiner: a web‐based suite of genomic and pharmacologic tools to explore transcript and drug patterns in the NCI‐60 cell line set. Cancer Res. 2012;72(14):3499–511.
Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483(7391):603–7.
Geeleher P, Cox N, Huang RS. pRRophetic: an R package for prediction of clinical chemotherapeutic response from tumor gene expression levels. PLoS One. 2014;9(9):e107468.
Wang A, Jiang A, Gan X, Wang Z, Huang J, Dong K, et al. EGFR‐AS1 promotes bladder cancer progression by upregulating EGFR. BioMed Res Int. 2020;2020:6665974.
Jiang A, Meng J, Gong W, Zhang Z, Gan X, Wang J, et al. Elevated SNRPA1, as a promising predictor reflecting severe clinical outcome via effecting tumor immunity for ccRCC, is related to cell invasion, metastasis, and sunitinib sensitivity. Front Immunol. 2022;13:842069.
Coppin C, Kollmannsberger C, Le L, Porzsolt F, Wilt TJ. Targeted therapy for advanced renal cell cancer (RCC): a Cochrane systematic review of published randomised trials. BJU Int. 2011;108(10):1556–63.
Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, Harrison MR, et al. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J Clin Oncol. 2015;33(13):14307. 10.1200/JCO.2014.59.0703
Peña C, Lathia C, Shan M, Escudier B, Bukowski RM. Biomarkers predicting outcome in patients with advanced renal cell carcinoma: results from sorafenib phase III treatment approaches in renal cancer global evaluation trial. Clinical Cancer Res. 2010;16(19):4853–63.
Qu L, Wang ZL, Chen Q, Li YM, He HW, Hsieh JJ, et al. Prognostic value of a long non‐coding RNA signature in localized clear cell renal cell carcinoma. Eur Urol. 2018;74(6):756–63.
Wei J‐H, Feng ZH, Cao Y, Zhao HW, Chen ZH, Liao B, et al. Predictive value of single‐nucleotide polymorphism signature for recurrence in localised renal cell carcinoma: a retrospective analysis and multicentre validation study. Lancet Oncol. 2019;20(4):591–600.
Mehranfard D, Perez G, Rodriguez A, Ladna JM, Neagra CT, Goldstein B, et al. Alterations in gene expression of renin‐angiotensin system components and related proteins in colorectal cancer. J Renin‐Angiotensin‐Aldosterone Syst. 2021;2021:9987115.
Xie G, Cheng T, Lin J, Zhang L, Zheng J, Liu Y, et al. Local angiotensin II contributes to tumor resistance to checkpoint immunotherapy. J Immunother Cancer. 2018;6(1):88.
Li X‐Q, Du X, Li DM, Kong PZ, Sun Y, Liu PF, et al. ITGBL1 is a Runx2 transcriptional target and promotes breast cancer bone metastasis by activating the TGFβ signaling pathway. Cancer Res. 2015;75(16):3302–13.
Li W, Li S, Yang J, Cui C, Yu M, Zhang Y. ITGBL1 promotes EMT, invasion and migration by activating NF‐κB signaling pathway in prostate cancer. OncoTargets Ther. 2019;12:3753–63.
Matsuyama T, Ishikawa T, Takahashi N, Yamada Y, Yasuno M, Kawano T, et al. Transcriptomic expression profiling identifies ITGBL1, an epithelial to mesenchymal transition (EMT)‐associated gene, is a promising recurrence prediction biomarker in colorectal cancer. Mol Cancer. 2019;18(1):19.
Cortez AJ, Kujawa KA, Wilk AM, Sojka DR, Syrkis JP, Olbryt M, et al. Evaluation of the role of ITGBL1 in ovarian cancer. Cancers. 2020;12(9):E2676.
Huang W, Yu D, Wang M, Han Y, Lin J, Wei D, et al. ITGBL1 promotes cell migration and invasion through stimulating the TGF‐β signalling pathway in hepatocellular carcinoma. Cell Proliferation. 2020;53(7):e12836.
Ji Q, Zhou L, Sui H, Yang L, Wu X, Song Q, et al. Primary tumors release ITGBL1‐rich extracellular vesicles to promote distal metastatic tumor growth through fibroblast‐niche formation. Nat Commun. 2020;11(1):1211.
Cheli Y, Tulic MK, El Hachem N, Nottet N, Jacquel A, Gesson M, et al. ITGBL1 is a new immunomodulator that favors development of melanoma tumors by inhibiting natural killer cells cytotoxicity. Mol Cancer. 2021;20(1):12.
Poveda J, Sanz AB, Fernandez‐Fernandez B, Carrasco S, Ruiz‐Ortega M, Cannata‐Ortiz P, et al. MXRA5 is a TGF‐β1‐regulated human protein with anti‐inflammatory and anti‐fibrotic properties. J Cell Mol Med. 2017;21(1):154–64.
He Y, Chen X, Liu H, Xiao H, Kwapong WR, Mei J. Matrix‐remodeling associated 5 as a novel tissue biomarker predicts poor prognosis in non‐small cell lung cancers. Cancer Biomarkers. 2015;15(5):645–51.
Xiong D, Li G, Li K, Xu Q, Pan Z, Ding F, et al. Exome sequencing identifies MXRA5 as a novel cancer gene frequently mutated in non‐small cell lung carcinoma from Chinese patients. Carcinogenesis. 2012;33(9):1797–805.
Sun J‐Z, Zhang JH, Li JB, Yuan F, Tong LQ, Wang XY, et al. MXRA5 is a novel Immune‐Related biomarker that predicts poor prognosis in glioma. Dis Markers. 2021;2021:6680883.
Peralta‐Arrieta I, Trejo‐Villegas OA, Armas‐López L, Ceja‐Rangel HA, Ordóñez‐Luna MDC, Pineda‐Villegas P, et al. Failure to EGFR‐TKI‐based therapy and tumoural progression are promoted by MEOX2/GLI1‐mediated epigenetic regulation of EGFR in the human lung cancer. Eur J Cancer. 2022;160:189205. 10.1016/j.ejca.2021.10.032
Tachon G, Masliantsev K, Rivet P, Desette A, Milin S, Gueret E, et al. MEOX2 transcription factor is involved in survival and adhesion of glioma stem‐like cells. Cancers. 2021;13(23):5943.
Tachon G, Masliantsev K, Rivet P, Petropoulos C, Godet J, Milin S, et al. Prognostic significance of MEOX2 in gliomas. Mod Pathol. 2019;32(6):77486. 10.1038/s41379-018-0192-6
Yun JW, Yang L, Park HY, Lee CW, Cha H, Shin HT, et al. Dysregulation of cancer genes by recurrent intergenic fusions. Genome Biol. 2020;21(1):166.
Vogel CJ, Smit MA, Maddalo G, Possik PA, Sparidans RW, van der Burg SH, et al. Cooperative induction of apoptosis in NRAS mutant melanoma by inhibition of MEK and ROCK. Pigm Cell Melanoma Res. 2015;28(3):307–17.
Rontogianni S, Iskit S, van Doorn S, Peeper DS, Altelaar M. Combined EGFR and ROCK inhibition in triple‐negative breast cancer leads to cell death via impaired autophagic flux. Mol Cell Proteomics. 2020;19(2):26177. 10.1074/mcp.RA119.001800
Weiße J, Rosemann J, Müller L, Kappler M, Eckert AW, Glaß M, et al. Identification of lymphocyte cell‐specific protein‐tyrosine kinase (LCK) as a driver for invasion and migration of oral cancer by tumor heterogeneity exploitation. Mol Cancer. 2021;20(1):88.
Duan Z, Zhang J, Ye S, Shen J, Choy E, Cote G, et al. A‐770041 reverses paclitaxel and doxorubicin resistance in osteosarcoma cells. BMC Cancer. 2014;14:681.
Jin L, Chun J, Pan C, Alesi GN, Li D, Magliocca KR, et al. Phosphorylation‐mediated activation of LDHA promotes cancer cell invasion and tumour metastasis. Oncogene. 2017;36(27):3797–806.
Chakraborty G, Patail NK, Hirani R, Nandakumar S, Mazzu YZ, Yoshikawa Y, et al. Attenuation of SRC kinase activity augments PARP inhibitor‐mediated synthetic lethality in BRCA2‐altered prostate tumors. Clin Cancer Res. 2021;27(6):17921806. 10.1158/1078-0432.CCR-20-2483
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 30 May 2022
Revised: 27 June 2022
Accepted: 14 July 2022
Published: 30 August 2022
Issue date: August 2022

Copyright

© 2022 The Authors.

Acknowledgements

The authors would like to thank Dr. Jianming Zeng (University of Macau), and all the members of his bioinformatics team, Biotrainee, for generously sharing their experience and codes. The use of the Biorstudio high‐performance computing cluster (https://biorstudio.cloud) at Biotrainee and the Shanghai HS Biotech Co., Ltd., for conducting the research reported in this paper.

Rights and permissions

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

Return