AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (7.5 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

Clinical significance, molecular characterization, and immune microenvironment analysis of coagulation‐related genes in clear cell renal cell carcinoma

Weihao Chen1,Xupeng Zhao2,Yongliang Lu1,Hanfeng Wang1Xiyou Wang1Yi Wang1Chen Liang3( )Zhuomin Jia1( )Wei Ma4 ( )
Department of Urology, The Third Medical Center of PLA General Hospital, Beijing, China
School of Medicine, Nankai University, Tianjin, China
Medical Service Department, The PLA General Hospital, Beijing, China
Senior Department of Otolaryngology‐ Head & Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing, China

Weihao Chen, Xupeng Zhao, and Yongliang Lu contributed equally to this study and shared the first authorship.

Show Author Information

Graphical Abstract

Abstract

Background

Numerous studies have revealed a tight connection between tumor development and the coagulation system. However, the effects of coagulation on the prognosis and tumor microenvironment (TME) of clear cell renal cell carcinoma (ccRCC) remain poorly understood.

Methods

We employed the consensus clustering method to characterize distinct molecular subtypes associated with coagulation patterns. Subsequently, we examined variations in the overall survival (OS), genomic profiles, and TME characteristics between these subtypes. To develop a prognostic coagulation‐related risk score (CRRS) model, we utilized the least absolute shrinkage and selection operator Cox regression and stepwise multivariate Cox regression analyses. We also created a nomogram to aid in the clinical application of the risk score, evaluating the relationships between the CRRS and the immune microenvironment, responsiveness to immunotherapy, and targeted treatment. The clinical significance of PLAUR and its biological function in ccRCC were also further analyzed.

Results

There were significant differences in clinical features, prognostic stratification, genomic variation, and TME characteristics between the two coagulation‐related subtypes. We established and validated a CRRS using six coagulation‐related genes that can be employed as an effective indicator of risk stratification and prognosis estimation for ccRCC patients. Significant variations in survival outcomes were observed between the high‐ and low‐risk groups. The nomogram was proficient in predicting the 1‐, 3‐, and 5‐year OS. Additionally, the CRRS emerged as a novel tool for evaluating the clinical effectiveness of immunotherapy and targeted treatments in ccRCC. Moreover, we confirmed upregulated PLAUR expression in ccRCC samples that was significantly correlated with poor patient prognosis. PLAUR knockdown notably inhibited ccRCC cell proliferation and migration.

Conclusion

Our data suggested that CRRS may be employed as a reliable predictive biomarker that can provide therapeutic benefits for immunotherapy and targeted therapy in ccRCC.

References

1

Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM. The 2016 WHO classification of tumours of the urinary system and male genital organs‐part A: renal, penile, and testicular tumours. Eur Urol. 2016;70(1):93–105. https://doi.org/10.1016/j.eururo.2016.02.029

2

Rini BI, Campbell SC, Escudier B. Renal cell carcinoma. Lancet. 2009;373(9669):1119–32. https://doi.org/10.1016/S0140-6736(09)60229-4

3

Kotecha RR, Motzer RJ, Voss MH. Towards individualized therapy for metastatic renal cell carcinoma. Nat Rev Clin Oncol. 2019;16(10):621–33. https://doi.org/10.1038/s41571-019-0209-1

4

Kraft C, Schuettfort G, Weil Y, Tirneci V, Kasper A, Haberichter B, et al. Thrombosis of the inferior vena cava and malignant disease. Thromb Res. 2014;134(3):668–73. https://doi.org/10.1016/j.thromres.2014.07.012

5

Warsow G, Hübschmann D, Kleinheinz K, Nientiedt C, Heller M, van Coile L, et al. Genomic features of renal cell carcinoma with venous tumor thrombus. Sci Rep. 2018;8(1):7477. https://doi.org/10.1038/s41598-018-25544-z

6

Psutka SP, Leibovich BC. Management of inferior vena cava tumor thrombus in locally advanced renal cell carcinoma. Ther Adv Urol. 2015;7(4):216–29. https://doi.org/10.1177/1756287215576443

7

Xiao B, Ma LL, Zhang SD, Xiao CL, Lu J, Hong K, et al. Correlation between coagulation function, tumor stage and metastasis in patients with renal cell carcinoma: a retrospective study. Chin Med J. 2011;124(8):1205–8.

8

Tian YJ, Hong M, Jing SS, Liu XC, Wang HZ, Wang XP, et al. Clinical and prognostic effect of plasma fibrinogen in renal cell carcinoma: a meta‐analysis. BioMed Res Int. 2017;2017:1–8. https://doi.org/10.1155/2017/9591506

9

Galmiche A, Rak J, Roumenina LT, Saidak Z. Coagulome and the tumor microenvironment: an actionable interplay. Trends in Cancer. 2022;8(5):369–83. https://doi.org/10.1016/j.trecan.2021.12.008

10

Saidak Z, Soudet S, Lottin M, Salle V, Sevestre MA, Clatot F, et al. A pan‐cancer analysis of the human tumor coagulome and its link to the tumor immune microenvironment. Cancer Immunol Immunother. 2021;70(4):923–33. https://doi.org/10.1007/s00262-020-02739-w

11

Vuong L, Kotecha RR, Voss MH, Hakimi AA. Tumor microenvironment dynamics in clear‐cell renal cell carcinoma. Cancer Discov. 2019;9(10):1349–57. https://doi.org/10.1158/2159-8290.cd-19-0499

12

Tas F, Kilic L, Bilgin E, Keskin S, Sen F, Ciftci R, et al. Clinical and prognostic significance of coagulation assays in advanced epithelial ovarian cancer. Int J Gynecol Cancer. 2013;23(2):276–81. https://doi.org/10.1097/igc.0b013e31827b8796

13

Tikhomirova I, Petrochenko E, Malysheva Y, Ryabov M, Kislov N. Interrelation of blood coagulation and hemorheology in cancer. Clin Hemorheol Microcirc. 2017;64(4):635–44. https://doi.org/10.3233/ch-168037

14

Kawai K, Watanabe T. Colorectal cancer and hypercoagulability. Surg Today. 2014;44(5):797–803. https://doi.org/10.1007/s00595-013-0606-5

15

Ma MS, Cao R, Wang W, Wang BY, Yang YC, Huang YC, et al. The D‐dimer level predicts the prognosis in patients with lung cancer: a systematic review and meta‐analysis. J Cardiothorac Surg. 2021;16(1):243. https://doi.org/10.1186/s13019-021-01618-4

16

Tieken C, Versteeg HH. Anticoagulants versus cancer. Thromb Res. 2016;140(Suppl 1):S148–53. https://doi.org/10.1016/S0049-3848(16)30114-1

17

Haist M, Stege H, Pemler S, Heinz J, Fleischer MI, Graf C, et al. Anticoagulation with factor Xa inhibitors is associated with improved overall response and progression‐free survival in patients with metastatic malignant melanoma receiving immune checkpoint inhibitors—a retrospective, real‐world cohort study. Cancers. 2021;13(20):5103. https://doi.org/10.3390/cancers13205103

18

Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high‐throughput experiments. Bioinformatics. 2012;28(6):882–3. https://doi.org/10.1093/bioinformatics/bts034

19

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. https://doi.org/10.1101/gr.239244.118

20

Wilkerson MD, Hayes DN. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics. 2010;26(12):1572–3. https://doi.org/10.1093/bioinformatics/btq170

21

Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–7. https://doi.org/10.1038/nmeth.3337

22

Yoshihara K, Shahmoradgoli M, Martínez E, Vegesna R, Kim H, Torres‐Garcia W, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612. https://doi.org/10.1038/ncomms3612

23

Hänzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA‐seq data. BMC Bioinformatics. 2013;14:7. https://doi.org/10.1186/1471-2105-14-7

24

Charoentong P, Finotello F, Angelova M, Mayer C, Efremova M, Rieder D, et al. Pan‐cancer immunogenomic analyses reveal genotype‐immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep. 2017;18(1):248–62. https://doi.org/10.1016/j.celrep.2016.12.019

25

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. Nat Med. 2018;24(10):1550–8. https://doi.org/10.1038/s41591-018-0136-1

26

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. https://doi.org/10.1371/journal.pone.0107468

27

Chen W, Wang H, Lu Y, Huang Y, Xuan Y, Li X, et al. GTSE1 promotes tumor growth and metastasis by attenuating of KLF4 expression in clear cell renal cell carcinoma. Lab Invest. 2022;102(9):1011–22. https://doi.org/10.1038/s41374-022-00797-5

28

Falanga A, Marchetti M, Vignoli A. Coagulation and cancer: biological and clinical aspects. J Thromb Haemostasis. 2013;11(2):223–33. https://doi.org/10.1111/jth.12075

29

Falanga A, Russo L, Verzeroli C. Mechanisms of thrombosis in cancer. Thromb Res. 2013;131(Suppl 1):S59–S62. https://doi.org/10.1016/S0049-3848(13)70024-0

30

Vedovati MC, Giustozzi M, Becattini C. Venous thromboembolism and cancer: current and future role of direct‐acting oral anticoagulants. Thromb Res. 2019;177:33–41. https://doi.org/10.1016/j.thromres.2019.02.031

31

Lee AYY, Levine MN. Venous thromboembolism and cancer: risks and outcomes. Circulation. 2003;107(23 Suppl 1):I17–21. https://doi.org/10.1161/01.cir.0000078466.72504.ac

32

García‐Escobar I, Brozos‐Vázquez E, Gutierrez Abad D, Martínez‐Marín V, Pachón V, Muñoz Martín AJ, et al. Direct oral anticoagulants for the treatment and prevention of venous thromboembolism in patients with cancer: current evidence. Clin Transl Oncol. 2021;23(6):1034–46. https://doi.org/10.1007/s12094-020-02506-4

33

Granito A, Muratori L, Lalanne C, Quarneti C, Ferri S, Guidi M, et al. Hepatocellular carcinoma in viral and autoimmune liver diseases: role of CD4+ CD25+ Foxp3+ regulatory T cells in the immune microenvironment. World J Gastroenterol. 2021;27(22):2994–3009. https://doi.org/10.3748/wjg.v27.i22.2994

34

Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017;27(1):109–18. https://doi.org/10.1038/cr.2016.151

35

Pan YY, Yu YD, Wang XJ, Zhang T. Tumor‐associated macrophages in tumor immunity. Front Immunol. 2020;11:583084. https://doi.org/10.3389/fimmu.2020.583084

36

Rizzo A, Mollica V, Santoni M, Ricci AD, Rosellini M, Marchetti A, et al. Impact of clinicopathological features on survival in patients treated with first‐line immune checkpoint inhibitors plus tyrosine kinase inhibitors for renal cell carcinoma: a meta‐analysis of randomized clinical trials. Eur Urol Focus. 2022;8(2):514–21. https://doi.org/10.1016/j.euf.2021.03.001

37

Massari F, Mollica V, Rizzo A, Cosmai L, Rizzo M, Porta C. Safety evaluation of immune‐based combinations in patients with advanced renal cell carcinoma: a systematic review and meta‐analysis. Expert Opin Drug Saf. 2020;19(10):1329–38. https://doi.org/10.1080/14740338.2020.1811226

38

Liu M, Chen S, Zhang A, Zheng Q, Fu J. PLAUR as a potential biomarker associated with immune infiltration in bladder urothelial carcinoma. J Inflamm Res. 2021;14:4629–41. https://doi.org/10.2147/jir.s326559

39

Zeng F, Li GZ, Liu X, Zhang KN, Huang H, Jiang T, et al. Plasminogen activator urokinase receptor implies immunosuppressive features and acts as an unfavorable prognostic biomarker in glioma. Oncologist. 2021;26(8):e1460–69. https://doi.org/10.1002/onco.13750

40

Ploug M, Rønne E, Behrendt N, Jensen AL, Blasi F, Danø K. Cellular receptor for urokinase plasminogen activator. Carboxyl‐terminal processing and membrane anchoring by glycosyl‐phosphatidylinositol. J Biol Chem. 1991;266(3):1926–33. https://doi.org/10.1016/S0021-9258(18)52382-6

41

Laurenzana A, Chillà A, Luciani C, Peppicelli S, Biagioni A, Bianchini F, et al. uPA/uPAR system activation drives a glycolytic phenotype in melanoma cells. Int J Cancer. 2017;141(6):1190–200. https://doi.org/10.1002/ijc.30817

42

Hildenbrand R, Schaaf A. The urokinase‐system in tumor tissue stroma of the breast and breast cancer cell invasion. Int J Oncol. 2009;34(1):15–23. https://doi.org/10.3892/ijo_00000124

43

Boonstra MC, Verspaget HW, Ganesh S, Kubben FJGM, Vahrmeijer AL, van de Velde CJH, et al. Clinical applications of the urokinase receptor (uPAR) for cancer patients. Curr Pharm Des. 2011;17(19):1890–910. https://doi.org/10.2174/138161211796718233

44

LeBeau AM, Duriseti S, Murphy ST, Pepin F, Hann B, Gray JW, et al. Targeting uPAR with antagonistic recombinant human antibodies in aggressive breast cancer. Cancer Res. 2013;73(7):2070–81. https://doi.org/10.1158/0008-5472.can-12-3526

45

Zhang CQ, Zhang GC, Sun N, Zhang Z, Xue LY, Zhang ZH, et al. An individualized immune signature of pretreatment biopsies predicts pathological complete response to neoadjuvant chemoradiotherapy and outcomes in patients with esophageal squamous cell carcinoma. Signal Transduct Target Ther. 2020;5(1):182. https://doi.org/10.1038/s41392-020-00221-8

46

Lindsten T, Hedbrant A, Ramberg A, Wijkander J, Solterbeck A, Eriksson M, et al. Effect of macrophages on breast cancer cell proliferation, and on expression of hormone receptors, uPAR and HER‐2. Int J Oncol. 2017;51(1):104–14. https://doi.org/10.3892/ijo.2017.3996

47

Chen HM, Lu SY, Guan JQ, Zhu XQ, Sun FF, Huang JT, et al. Identification of prognostic immune‐related genes in rhabdoid tumor of kidney based on TARGET database analysis. Aging. 2021;13(4):5461–74. https://doi.org/10.18632/aging.202475

Cancer Innovation
Article number: e105
Cite this article:
Chen W, Zhao X, Lu Y, et al. Clinical significance, molecular characterization, and immune microenvironment analysis of coagulation‐related genes in clear cell renal cell carcinoma. Cancer Innovation, 2024, 3(1): e105. https://doi.org/10.1002/cai2.105

135

Views

3

Downloads

0

Crossref

0

Scopus

Altmetrics

Received: 14 July 2023
Accepted: 30 September 2023
Published: 07 January 2024
© 2024 The Authors. Tsinghua University Press.

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