Skip to main content

Advertisement

Log in

A risk model developed based on tumor microenvironment predicts overall survival and associates with tumor immunity of patients with lung adenocarcinoma

  • Article
  • Published:
Oncogene Submit manuscript

Abstract

Tumor microenvironment (TME) has been reported to exhibit a crucial effect in lung cancer. Therefore, this study was aimed to investigate the genes associated with TME and develop a risk score to predict the overall survival (OS) of patients with lung adenocarcinoma (LUAD) based on these genes. The immune and stromal scores were generated by the ESTIMATE algorithm for LUAD patients in The Cancer Genome Atlas (TCGA) database. Differentially expressed gene and weighted gene co-expression network analyses were used to derive immune- and stromal-related genes. The Least Absolute Shrinkage and Selection Operator (LASSO)-Cox regression was applied for further selection and the selected genes were inputted into stepwise regression to develop TME-related risk score (TMErisk) which was further validated in Gene Expression Omnibus (GEO) datasets. TMErisk-related biological phenotypes were analyzed in function enrichment, tumor immune signature, and tumor mutation signature. The patient’s response to immunotherapy was inferred by the tumor immune dysfunction and exclusion (TIDE) score and immunophenoscore (IPS). According to our results, TMErisk was developed based on SERPINE1, CX3CR1, CD200R1, GBP1, IRF1, STAP1, LOX, and OR7E47P. Furthermore, high TMErisk was identified as a poor factor for OS in TCGA and GEO datasets, as well as in subgroup analysis with different gender, smoking status, age, race, anatomic site, therapies, and tumor-node-metastasis (TNM) stages. Higher TMErisk is also associated negatively with the abundance of B cells, CD4+ T cells, CD8+ T cells, neutrophils, macrophages, and other stromal or immune cells. Several genes of the human leukocyte antigen (HLA) family and immune checkpoints were less expressed in the high-TMErisk group. Mutations of 19 genes occurred more frequently in the high-TMErisk group. These mutations may be associated with TME change and indicate patients’ response to immunotherapy. According to our analyses, a lower TMErisk score may indicate better response and OS outcome of immunotherapy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Screening for TME-related genes.
Fig. 2: Evaluation and validation for the prognostic value of the TMErisk score.
Fig. 3: Function enrichment analysis for TMErisk and correlation between TMErisk and expression of the HLA family genes/immune checkpoints.
Fig. 4: Landscape of immune and stromal cell infiltrations in the low- and high-TMErisk groups.
Fig. 5: TMErisk was related to tumor mutation status.

Similar content being viewed by others

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.

    Article  Google Scholar 

  2. Bender E. Epidemiology: the dominant malignancy. Nature. 2014;513:S2–3.

    CAS  PubMed  Google Scholar 

  3. Jamal-Hanjani M, Quezada SA, Larkin J, Swanton C. Translational implications of tumor heterogeneity. Clin Cancer Res. 2015;21:1258–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Carter BW, Godoy MC, Wu CC, Erasmus JJ, Truong MT. Current controversies in lung cancer staging. J Thorac Imaging. 2016;31:201–14.

    PubMed  Google Scholar 

  5. Martinez-Terroba E, Behrens C, de Miguel FJ, Agorreta J, Monso E, Millares L, et al. A novel protein-based prognostic signature improves risk stratification to guide clinical management in early-stage lung adenocarcinoma patients. J Pathol. 2018;245:421–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Travis WD, Brambilla E, Riely GJ. New pathologic classification of lung cancer: relevance for clinical practice and clinical trials. J Clin Oncol. 2013;31:992–1001.

    CAS  PubMed  Google Scholar 

  7. Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21:309–22.

    CAS  PubMed  Google Scholar 

  8. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70.

    CAS  PubMed  Google Scholar 

  9. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24:541–50.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Choi H, Na KJ. Integrative analysis of imaging and transcriptomic data of the immune landscape associated with tumor metabolism in lung adenocarcinoma: clinical and prognostic implications. Theranostics. 2018;8:1956–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Giraldo NA, Becht E, Vano Y, Petitprez F, Lacroix L, Validire P, et al. Tumor-Infiltrating and peripheral blood T-cell immunophenotypes predict early relapse in localized clear cell renal cell carcinoma. Clin Cancer Res. 2017;23:4416–28.

    CAS  PubMed  Google Scholar 

  12. Denton AE, Roberts EW, Fearon DT. Stromal cells in the tumor microenvironment. Adv Exp Med Biol. 2018;1060:99–114.

    CAS  PubMed  Google Scholar 

  13. Yoshihara K, Shahmoradgoli M, Martinez 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.

    PubMed  Google Scholar 

  14. Jia D, Li S, Li D, Xue H, Yang D, Liu Y. Mining TCGA database for genes of prognostic value in glioblastoma microenvironment. Aging. 2018;10:592–605.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Yan H, Qu J, Cao W, Liu Y, Zheng G, Zhang E, et al. Identification of prognostic genes in the acute myeloid leukemia immune microenvironment based on TCGA data analysis. Cancer Immunol Immunother. 2019;68:1971–8.

    CAS  PubMed  Google Scholar 

  16. Yang S, Liu T, Nan H, Wang Y, Chen H, Zhang X, et al. Comprehensive analysis of prognostic immune-related genes in the tumor microenvironment of cutaneous melanoma. J Cell Physiol. 2020;235:1025–35.

    CAS  PubMed  Google Scholar 

  17. Xu WH, Xu Y, Wang J, Wan FN, Wang HK, Cao DL, et al. Prognostic value and immune infiltration of novel signatures in clear cell renal cell carcinoma microenvironment. Aging. 2019;11:6999–7020.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Wang H, Wu X, Chen Y. Stromal-immune score-based gene signature: a prognosis stratification tool in gastric cancer. Front Oncol. 2019;9:1212.

    PubMed  PubMed Central  Google Scholar 

  19. Kinoshita T, Muramatsu R, Fujita T, Nagumo H, Sakurai T, Noji S, et al. Prognostic value of tumor-infiltrating lymphocytes differs depending on histological type and smoking habit in completely resected non-small-cell lung cancer. Ann Oncol. 2016;27:2117–23.

    CAS  PubMed  Google Scholar 

  20. Barker HE, Cox TR, Erler JT. The rationale for targeting the LOX family in cancer. Nat Rev Cancer. 2012;12:540–52.

    CAS  PubMed  Google Scholar 

  21. Schmall A, Al-Tamari HM, Herold S, Kampschulte M, Weigert A, Wietelmann A, et al. Macrophage and cancer cell cross-talk via CCR2 and CX3CR1 is a fundamental mechanism driving lung cancer. Am J Respir Crit Care Med. 2015;191:437–47.

    CAS  PubMed  Google Scholar 

  22. Conroy MJ, Lysaght J. CX3CL1 signaling in the tumor microenvironment. Adv Exp Med Biol. 2020;1231:1–12.

    CAS  PubMed  Google Scholar 

  23. Duffy MJ. The urokinase plasminogen activator system: role in malignancy. Curr Pharm Des. 2004;10:39–49.

    CAS  PubMed  Google Scholar 

  24. Garg N, Goyal N, Strawn TL, Wu J, Mann KM, Lawrence DA, et al. Plasminogen activator inhibitor-1 and vitronectin expression level and stoichiometry regulate vascular smooth muscle cell migration through physiological collagen matrices. J Thromb Haemost. 2010;8:1847–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Erin N, Podnos A, Tanriover G, Duymus O, Cote E, Khatri I, et al. Bidirectional effect of CD200 on breast cancer development and metastasis, with ultimate outcome determined by tumor aggressiveness and a cancer-induced inflammatory response. Oncogene. 2015;34:3860–70.

    CAS  PubMed  Google Scholar 

  26. Frankel T, Lanfranca MP, Zou W. The role of tumor microenvironment in cancer immunotherapy. Adv Exp Med Biol. 2017;1036:51–64.

    CAS  PubMed  Google Scholar 

  27. Wu T, Dai Y. Tumor microenvironment and therapeutic response. Cancer Lett. 2017;387:61–8.

    CAS  Google Scholar 

  28. Zhang Y, Guoqiang L, Sun M, Lu X. Targeting and exploitation of tumor-associated neutrophils to enhance immunotherapy and drug delivery for cancer treatment. Cancer Biol Med. 2020;17:32–43.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016;17:e542–e551.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Camidge DR, Doebele RC, Kerr KM. Comparing and contrasting predictive biomarkers for immunotherapy and targeted therapy of NSCLC. Nat Rev Clin Oncol. 2019;16:341–55.

    CAS  PubMed  Google Scholar 

  31. Wojas-Krawczyk K, Kalinka E, Grenda A, Krawczyk P, Milanowski J. Beyond PD-L1 markers for lung cancer immunotherapy. Int J Mol Sci 2019; 20:1915.

  32. Zeng D, Li M, Zhou R, Zhang J, Sun H, Shi M, et al. Tumor microenvironment characterization in gastric cancer identifies prognostic and immunotherapeutically relevant gene signatures. Cancer Immunol Res. 2019;7:737–50.

    CAS  PubMed  Google Scholar 

  33. Wang S, He Z, Wang X, Li H, Liu XS. Antigen presentation and tumor immunogenicity in cancer immunotherapy response prediction. Elife 2019; 8:e49020.

  34. Ohri CM, Shikotra A, Green RH, Waller DA, Bradding P. Macrophages within NSCLC tumour islets are predominantly of a cytotoxic M1 phenotype associated with extended survival. Eur Respir J. 2009;33:118–26.

    CAS  PubMed  Google Scholar 

  35. Rakaee M, Busund LR, Jamaly S, Paulsen EE, Richardsen E, Andersen S, et al. Prognostic value of macrophage phenotypes in resectable non-small cell lung cancer assessed by multiplex immunohistochemistry. Neoplasia. 2019;21:282–93.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. 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;378:2093–104.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Hellmann MD, Callahan MK, Awad MM, Calvo E, Ascierto PA, Atmaca A. et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell. 2018;33:853–61. e854.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Assoun S, Theou-Anton N, Nguenang M, Cazes A, Danel C, Abbar B, et al. Association of TP53 mutations with response and longer survival under immune checkpoint inhibitors in advanced non-small-cell lung cancer. Lung Cancer. 2019;132:65–71.

    PubMed  Google Scholar 

  39. Zhao LQ, Qu XF, Wu ZH, Li YH, Zhang XW, Guo WJ. TP53 somatic mutations are associated with poor survival in non-small cell lung cancer patients who undergo immunotherapy. Aging. 2020;12:14556–68.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Sun H, Liu SY, Zhou JY, Xu JT, Zhang HK, Yan HH, et al. Specific TP53 subtype as biomarker for immune checkpoint inhibitors in lung adenocarcinoma. Ebiomedicine 2020; 60:102990.

  41. Marinelli D, Mazzotta M, Scalera S, Terrenato I, Sperati F, D’Ambrosio L, et al. KEAP1-driven co-mutations in lung adenocarcinoma unresponsive to immunotherapy despite high tumor mutational burden. Ann Oncol. 2020;31:1746–54.

    CAS  PubMed  Google Scholar 

  42. Aran D, Sirota M, Butte AJ. Systematic pan-cancer analysis of tumour purity. Nat Commun. 2015;6:8971.

    CAS  PubMed  Google Scholar 

  43. Tibshirani R. The lasso method for variable selection in the Cox model. Stat Med. 1997;16:385–95.

    CAS  PubMed  Google Scholar 

  44. Gyorffy B, Surowiak P, Budczies J, Lanczky A. Online survival analysis software to assess the prognostic value of biomarkers using transcriptomic data in non-small-cell lung cancer. PLoS One. 2013;8:e82241.

    PubMed  PubMed Central  Google Scholar 

  45. De Simone M, Arrigoni A, Rossetti G, Gruarin P, Ranzani V, Politano C, et al. Transcriptional landscape of human tissue lymphocytes unveils uniqueness of tumor-infiltrating T regulatory cells. Immunity. 2016;45:1135–47.

    PubMed  PubMed Central  Google Scholar 

  46. Johnston RJ, Su LJ, Pinckney J, Critton D, Boyer E, Krishnakumar A, et al. VISTA is an acidic pH-selective ligand for PSGL-1. Nature. 2019;574:565–70.

    CAS  PubMed  Google Scholar 

  47. Li B, Severson E, Pignon JC, Zhao H, Li T, Novak J, et al. Comprehensive analyses of tumor immunity: implications for cancer immunotherapy. Genome Biol. 2016;17:174.

    PubMed  PubMed Central  Google Scholar 

  48. 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;12:453–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 2017;18:220.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from Wuhan University Medical Faculty Innovation Seed Fund Cultivation Project (grant no. TFZZ2018025), Chen xiao-ping foundation for the development of science and technology of Hubei province (grant no. CXPJJH12000001-2020313), and the National Natural Science Foundation of China (grant nos. 81670123 and 81670144).

Author information

Authors and Affiliations

Authors

Contributions

Research design: Bin Xu and Jie Wu; data collection: Siyi Wu and Lan Li; data analysis: Jie Wu, Huibo Zhang and Lan Li; manuscript preparation: Jie Wu, Huibo Zhang, and Lan Li; chart preparation: Jie Wu and Siyi Wu; revisions: Jie Wu, Huibo Zhang, Lan Li, Yaqi Zhao, Haohan Zhang, and Bin Xu. All authors confirm that they contributed to manuscript reviews, critical revision for important intellectual content, and read and approved the final draft for submission. All authors are also responsible for the manuscript content.

Corresponding author

Correspondence to Bin Xu.

Ethics declarations

Ethical approval and consent to participate

Not applicable.

Conflict of interest

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

Supplement files-supplement figure legends

Figure S1. The flow diagram of TMErisk development.

Figure S2. Immune and stromal scores associate with clinical features and outcomes.

Figure S3. Analysis of network topology for various soft-thresholding powers.

Figure S4. Validation for prognostic value of alternative genes selected by LASSO analysis.

Figure S5. KM curve analysis for the low- and high-TMErisk in GSE3141, GSE31210, GSE81089, GSE30219, GSE37745 and TCGA.

Figure S6. Stratified analysis for the low- and high-TMErisk groups.

Figure S7. Difference analysis for the distribution of T helper cells and macrophage phenotypes.

Figure S8. Difference analysis for expression of 19 genes in wild type and mutant type.

Figure S9. TMErisk is a potential biomarker to predict patient’s benefits from drug therapies.

Table S1. The cut-off values used in the study.

Table S2. The epidemiological and clinical characteristics before and after propensity score matching.

Table S3. Results of GSEA for molecular function in GO database.

Table S4. Results of GSEA for biology process in GO database.

Table S5. Results of GSEA in KEGG database.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, J., Li, L., Zhang, H. et al. A risk model developed based on tumor microenvironment predicts overall survival and associates with tumor immunity of patients with lung adenocarcinoma. Oncogene 40, 4413–4424 (2021). https://doi.org/10.1038/s41388-021-01853-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-021-01853-y

  • Springer Nature Limited

This article is cited by

Navigation