Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Molecular Diagnostics

The tumour-associated stroma correlates with poor clinical outcomes and immunoevasive contexture in patients with upper tract urothelial carcinoma: results from a multicenter real-world study (TSU-01 Study)

Abstract

Background

In this real-world study, we aimed to elucidate the predictive value of tumour-associated stroma for clinical prognostic and therapeutic response in upper tract urothelial carcinoma (UTUC) by reviewing the clinicopathologic characteristics of 1015 UTUC patients through a nationwide multicenter analysis.

Methods

The tumour–stroma ratio (TSR) was assessed based on tissue sections stained for hematoxylin and eosin (H&E), and patients were further stratified into stroma-high (>50% stroma) and stroma-low group (≤50% stroma). Kaplan–Meier curve and Cox regression hazard analysis were conducted to assess the survival outcomes of UTUC patients. Bioinformatics analysis and immunostaining analysis were applied to portray the tumour microenvironment (TME).

Results

Stroma-high UTUC was significantly associated with poorer survival outcomes and inferior chemotherapeutic responsiveness. Our established nomogram achieved a high prognostic accuracy in predicting overall survival and cancer-specific survival in both of the discovery cohort (area under the curve [AUC] 0.663 and 0.712) and the validation cohort (AUC 0.741 and 0.747). Moreover, stroma-high UTUC was correlated with immunoevasive TME accompanied by increased cancer-associated fibroblasts, tumour-associated macrophages and, conspicuously a cluster of highly exhausted CD8+ T cells.

Conclusion

Our results showed stroma-high UTUC was associated with an inferior prognosis and an immunoevasive TME with exhausted CD8+ T cells in UTUC patients. Our TSR-based nomogram could be used to refine prognosis and inform treatment decisions of patients with UTUC.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Flowchart of the study.
Fig. 2: Association of the tumour-associated stroma with cancer survival outcome.
Fig. 3: The relationship between tumour-associated stroma and benefit of adjuvant chemotherapy (ACT) in patients with Stage II–IV upper tract urothelial carcinoma (UTUC).
Fig. 4: Stroma-high tumours were characterised by abundant cancer-associated fibroblasts (CAFs).
Fig. 5: Characterisation of the immunoevasive tumour microenvironment in stroma-high tumours.
Fig. 6: Construction of a TSR-based survival nomogram for upper tract urothelial carcinoma (UTUC).

Similar content being viewed by others

Data availability

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69:7–34.

    Article  Google Scholar 

  2. Birtle A, Johnson M, Chester J, Jones R, Dolling D, Bryan RT, et al. Adjuvant chemotherapy in upper tract urothelial carcinoma (the POUT trial): a phase 3, open-label, randomised controlled trial. Lancet. 2020;395:1268–77.

    Article  CAS  Google Scholar 

  3. Lughezzani G, Burger M, Margulis V, Matin SF, Novara G, Roupret M, et al. Prognostic factors in upper urinary tract urothelial carcinomas: a comprehensive review of the current literature. Eur Urol. 2012;62:100–14.

    Article  Google Scholar 

  4. Meng ZW, Pan W, Hong HJ, Chen JZ, Chen YL. Modified staging classification for intrahepatic cholangiocarcinoma based on the sixth and seventh editions of the AJCC/UICC TNM staging systems. Medicine. 2017;96:e7891.

    Article  Google Scholar 

  5. Soukup V, Capoun O, Cohen D, Hernandez V, Babjuk M, Burger M, et al. Prognostic performance and reproducibility of the 1973 and 2004/2016 World Health Organization Grading Classification Systems in non-muscle-invasive bladder cancer: a European association of urology non-muscle invasive bladder cancer guidelines panel systematic review. Eur Urol 2017;72:801–13.

    Article  Google Scholar 

  6. Necchi A, Lo Vullo S, Mariani L, Moschini M, Hendricksen K, Rink M, et al. Adjuvant chemotherapy after radical nephroureterectomy does not improve survival in patients with upper tract urothelial carcinoma: a joint study by the European Association of Urology-Young Academic Urologists and the Upper Tract Urothelial Carcinoma Collaboration. BJU Int. 2018;121:252–9.

    Article  CAS  Google Scholar 

  7. Seisen T, Krasnow RE, Bellmunt J, Roupret M, Leow JJ, Lipsitz SR, et al. Effectiveness of adjuvant chemotherapy after radical nephroureterectomy for locally advanced and/or positive regional lymph node upper tract urothelial carcinoma. J Clin Oncol. 2017;35:852–60.

    Article  Google Scholar 

  8. Quhal F, Mori K, Sari Motlagh R, Laukhtina E, Pradere B, Roupret M, et al. Efficacy of neoadjuvant and adjuvant chemotherapy for localized and locally advanced upper tract urothelial carcinoma: a systematic review and meta-analysis. Int J Clin Oncol. 2020;25:1037–54.

    Article  Google Scholar 

  9. Tanaka N, Kikuchi E, Shirotake S, Kanao K, Matsumoto K, Kobayashi H, et al. The predictive value of C-reactive protein for prognosis in patients with upper tract urothelial carcinoma treated with radical nephroureterectomy: a multi-institutional study. Eur Urol. 2014;65:227–34.

    Article  CAS  Google Scholar 

  10. Tanaka N, Kikuchi E, Kanao K, Matsumoto K, Shirotake S, Miyazaki Y, et al. A multi-institutional validation of the prognostic value of the neutrophil-to-lymphocyte ratio for upper tract urothelial carcinoma treated with radical nephroureterectomy. Ann Surg Oncol. 2014;21:4041–8.

    Article  Google Scholar 

  11. Giraldo NA, Sanchez-Salas R, Peske JD, Vano Y, Becht E, Petitprez F, et al. The clinical role of the TME in solid cancer. Br J Cancer. 2019;120:45–53.

    Article  Google Scholar 

  12. Xie HY, Shao ZM, Li DQ. Tumor microenvironment: driving forces and potential therapeutic targets for breast cancer metastasis. Chin J Cancer. 2017;36:36.

    Article  Google Scholar 

  13. Park JH, McMillan DC, Edwards J, Horgan PG, Roxburgh CS. Comparison of the prognostic value of measures of the tumor inflammatory cell infiltrate and tumor-associated stroma in patients with primary operable colorectal cancer. Oncoimmunology. 2016;5:e1098801.

    Article  CAS  Google Scholar 

  14. Liu J, Liu J, Li J, Chen Y, Guan X, Wu X, et al. Tumor-stroma ratio is an independent predictor for survival in early cervical carcinoma. Gynecol Oncol. 2014;132:81–6.

    Article  Google Scholar 

  15. Lou E, Vogel RI, Hoostal S, Klein M, Linden MA, Teoh D, et al. Tumor-stroma proportion as a predictive biomarker of resistance to platinum-based chemotherapy in patients with ovarian cancer. JAMA Oncol. 2019;5:1222–4.

    Article  Google Scholar 

  16. Huijbers A, Tollenaar RA, v Pelt GW, Zeestraten EC, Dutton S, McConkey CC, et al. The proportion of tumor-stroma as a strong prognosticator for stage II and III colon cancer patients: validation in the VICTOR trial. Ann Oncol. 2013;24:179–85.

    Article  CAS  Google Scholar 

  17. Chen Y, Zhang L, Liu W, Liu X. Prognostic significance of the tumor-stroma ratio in epithelial ovarian cancer. Biomed Res Int. 2015;2015:589301.

    Article  Google Scholar 

  18. Huang J, Zhang L, Wan D, Zhou L, Zheng S, Lin S, et al. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct Target Ther. 2021;6:153.

    Article  Google Scholar 

  19. Idorn M, Olsen M, Halldorsdottir HR, Skadborg SK, Pedersen M, Hogdall C, et al. Improved migration of tumor ascites lymphocytes to ovarian cancer microenvironment by CXCR2 transduction. Oncoimmunology. 2018;7:e1412029.

    Article  Google Scholar 

  20. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–37.

    Article  CAS  Google Scholar 

  21. Xu B, Li XL, Ye F, Zhu XD, Shen YH, Huang C, et al. Development and validation of a nomogram based on perioperative factors to predict post-hepatectomy liver failure. J Clin Transl Hepatol. 2021;9:291–300.

    CAS  Google Scholar 

  22. Zhong W, Wang B, Yu H, Lin J, Xia K, Hou W, et al. Serum CCL27 predicts the response to Bacillus Calmette-Guerin immunotherapy in non-muscle-invasive bladder cancer. Oncoimmunology. 2020;9:1776060.

    Article  Google Scholar 

  23. Chen J, Zhong W, Yang M, Hou W, Wang X, Xia K, et al. Development and validation of a PD-L1/PD-1/CD8 axis-based classifier to predict cancer survival of upper tract urothelial carcinoma after radical nephroureterectomy. Cancer Immunol Immunother. 2021;70:2657–68.

  24. Cheng S, Zhong W, Xia K, Hong P, Lin R, Wang B, et al. Prognostic role of stromal tumor-infiltrating lymphocytes in locally advanced upper tract urothelial carcinoma: a retrospective multicenter study (TSU-02 study). Oncoimmunology. 2021;10:1861737.

    Article  Google Scholar 

  25. Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, et al. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 2020;48:W509–W14.

    Article  CAS  Google Scholar 

  26. Becht E, Giraldo NA, Lacroix L, Buttard B, Elarouci N, Petitprez F, et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 2016;17:218.

    Article  Google Scholar 

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

    Article  Google Scholar 

  28. Subramanian A, Tamayo P, Mootha V, Mukherjee S, Ebert B, Gillette M, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. 2005;102:15545–50.

  29. Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013;39:782–95.

    Article  CAS  Google Scholar 

  30. Zheng C, Zheng L, Yoo JK, Guo H, Zhang Y, Guo X, et al. Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell. 2017;169:1342–56. e16

    Article  CAS  Google Scholar 

  31. Pagès F, Mlecnik B, Marliot F, Bindea G, Ou F-S, Bifulco C, et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet. 2018;391:2128–39.

    Article  Google Scholar 

  32. Peng C, Liu J, Yang G, Li Y. The tumor-stromal ratio as a strong prognosticator for advanced gastric cancer patients: proposal of a new TSNM staging system. J Gastroenterol. 2018;53:606–17.

    Article  Google Scholar 

  33. Birtle AJ, Chester JD, Jones RJ, Jenkins B, Johnson M, Catto JW, et al. Updated outcomes of POUT: a phase III randomized trial of peri-operative chemotherapy versus surveillance in upper tract urothelial cancer (UTUC). J Clin Oncol. 2021;39:455.

  34. van Wyk HC, Roseweir A, Alexander P, Park JH, Horgan PG, McMillan DC, et al. The relationship between tumor budding, tumor microenvironment, and survival in patients with primary operable colorectal cancer. Ann Surg Oncol. 2019;26:4397–404.

    Article  Google Scholar 

  35. Zunder SM, van Pelt GW, Gelderblom HJ, Mancao C, Putter H, Tollenaar RA, et al. Predictive potential of tumour-stroma ratio on benefit from adjuvant bevacizumab in high-risk stage II and stage III colon cancer. Br J Cancer. 2018;119:164–9.

    Article  CAS  Google Scholar 

  36. Karpathiou G, Vieville M, Gavid M, Camy F, Dumollard JM, Magne N, et al. Prognostic significance of tumor budding, tumor-stroma ratio, cell nests size, and stroma type in laryngeal and pharyngeal squamous cell carcinomas. Head Neck. 2019;41:1918–27.

    Article  Google Scholar 

  37. Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019;79:4557–66.

    Article  CAS  Google Scholar 

  38. Mhaidly R, Mechta-Grigoriou F. Role of cancer-associated fibroblast subpopulations in immune infiltration, as a new means of treatment in cancer. Immunol Rev. 2021;302:259–72.

  39. Yoo SY, Park HE, Kim JH, Wen X, Jeong S, Cho NY, et al. Whole-slide image analysis reveals quantitative landscape of tumor-immune microenvironment in colorectal cancers. Clin Cancer Res. 2020;26:870–81.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Dr. Qun He, from the Department of Pathology, Peking University First Hospital, for his assistance with IHC and IF score.

Funding

This study was supported by the National Natural Science Foundation of China (Grant Nos. 81825016, 81961128027, 81902586, 81772719, 81772728); Guangdong Provincial Natural Science Foundation (2021A1515011541); Guangdong Provincial Clinical Research Center for Urological Diseases (2020B1111170006); Key Laboratory of Malignant Tumour Gene Regulation and Target Therapy of Guangdong Higher Education Institutes (Grant No. KLB09001); Key Laboratory of Malignant Tumor Molecular Mechanism and Translational Medicine of Guangzhou Bureau of Science and Information Technology (Grant No. 013-163); The National Key Research and Development Program of China (Grant No. 2018YFA0902803); The Key Areas Research and Development Program of Guangdong (Grant No. 2018B010109006).

Author information

Authors and Affiliations

Authors

Contributions

LHX, WLZ, PH and CCL performed experiments, statistical analysis and drafted the manuscript. KX, MY, RCL and SDC analysed and interpretation of the data. BW, KX, MY and JYC provided technical and material support. LLM, XSL, LQZ and JH provided study supervision and revised the manuscript. JH and TXL designed the study.

Corresponding authors

Correspondence to Xuesong Li, Liqun Zhou, Jian Huang or Tianxin Lin.

Ethics declarations

Ethics approval and consent to participate

All patients signed an informed consent before surgery that permitted the usage of resected tumours and clinical profiles in research, under the condition of anonymity. The study was approved by the ethics committee of each institution (SYSEC-KY-KS-2022-32), and conducted in accordance with the Declaration of Helsinki. Signed written informed consent was obtained from all patients.

Consent to publish

Consent for publication was obtained from all participants.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, L., Zhong, W., Li, C. et al. The tumour-associated stroma correlates with poor clinical outcomes and immunoevasive contexture in patients with upper tract urothelial carcinoma: results from a multicenter real-world study (TSU-01 Study). Br J Cancer 128, 310–320 (2023). https://doi.org/10.1038/s41416-022-02049-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-022-02049-1

This article is cited by

Search

Quick links