We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Comprehensive analysis of microenvironment-related genes in lung adenocarcinoma

    Ye Tao

    Bioinformatics of Department, Key laboratory of Cell Biology, Ministry of Public Health, & Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang 110122, PR China

    ,
    Yunhui Li

    *Author for correspondence:

    E-mail Address: liyunhui2737@163.com

    ;

    E-mail Address: bliang@cmu.edu.cn

    Clinical Laboratory, PLA North Military Command Region General Hospital, Shenyang 110003, PR China

    Authors contributed equally

    Search for more papers by this author

    &
    Bin Liang

    *Author for correspondence:

    E-mail Address: liyunhui2737@163.com

    ;

    E-mail Address: bliang@cmu.edu.cn

    Bioinformatics of Department, Key laboratory of Cell Biology, Ministry of Public Health, & Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang 110122, PR China

    Authors contributed equally

    Search for more papers by this author

    Published Online:

    Aim: Understanding the cell types and cell compositions in tumor environment (TME) and the gene changes in lung adenocarcinoma (LUAD) may provide insights on immune profiles and treatment targets for LUAD patients. Materials & methods: The RNA-Seq data from The Cancer Genome Atlas database were used to calculate the stromal scores and immune scores and analyzed the fractions of tumor infiltrating immune cells in LUAD samples with ESTIMATE and CIBERSORT algorithm. Results: We extracted a list of TME-related differentially expressed genes and performed the functional enrichment analysis. We found these genes were mainly enriched in immune response and cancer-related signal pathways. The prognosis analysis indicated that LINC00211, MUC2, LINC00426, LY86-AS1 ZEB2-AS1 and EREG were associated with prognosis in LUAD patients. Conclusion: The current study provides novel insights into immune files and gene changes in TME in LUAD patients.

    References

    • 1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J. Clin. 65(2), 87–108 (2015).
    • 2. Chen Z, Fillmore CM, Hammerman PS, Kim CF, Wong KK. Non-small-cell lung cancers: a heterogeneous set of diseases. Nat. Rev. Cancer 14(8), 535–546 (2014).
    • 3. Lin JJ, Cardarella S, Lydon CA et al. Five-year survival in EGFR-mutant metastatic lung adenocarcinoma treated with EGFR-TKIs. J. Thorac. Oncol. 11(4), 556–565 (2016).
    • 4. Gettinger S, Lynch T. A decade of advances in treatment for advanced non-small cell lung cancer. Clin. Chest Med. 32(4), 839–851 (2011).
    • 5. Best SA, Ding S, Kersbergen A et al. Distinct initiating events underpin the immune and metabolic heterogeneity of KRAS-mutant lung adenocarcinoma. Nat. Commun. 10(1), 4190 (2019).
    • 6. Lv Z, Lei T. Systematical identifications of prognostic meaningful lung adenocarcinoma subtypes and the underlying mutational and expressional characters. BMC Cancer 20(1), 56 (2020).
    • 7. El-Hussein A, Manoto SL, Ombinda-Lemboumba S, Alrowaili ZA, Mthunzi-Kufa P. A review of chemotherapy and photodynamic therapy for lung cancer treatment. Anticancer Agents Med. Chem. 18( 5), 438–407 (.2020).
    • 8. Lee SS, Cheah YK. The interplay between MicroRNAs and cellular components of tumour microenvironment (TME) on non-small-cell lung cancer (NSCLC) progression. J. Immunol. Res. 2019, 3046379 (2019).
    • 9. Pickup MW, Mouw JK, Weaver VM. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 15(12), 1243–1253 (2014).
    • 10. Guo S, Deng CX. Effect of stromal cells in tumor microenvironment on metastasis initiation. Int. J. Biol. Sci. 14(14), 2083–2093 (2018).
    • 11. Wu J, Liang C, Chen M, Su W. Association between tumor-stroma ratio and prognosis in solid tumor patients: a systematic review and meta-analysis. Oncotarget 7(42), 68954–68965 (2016).
    • 12. Zhang T, Xu J, Shen H, Dong W, Ni Y, Du J. Tumor-stroma ratio is an independent predictor for survival in NSCLC. Int. J. Clin. Exp. Pathol. 8(9), 11348–11355 (2015).
    • 13. Liu J, Liu J, Li J et al. Tumor-stroma ratio is an independent predictor for survival in early cervical carcinoma. Gynecol. Oncol. 132(1), 81–86 (2014).
    • 14. De Kruijf EM, Van Nes JG, Van De Velde CJ et al. Tumor-stroma ratio in the primary tumor is a prognostic factor in early breast cancer patients, especially in triple-negative carcinoma patients. Breast Cancer Res. Treat. 125(3), 687–696 (2011).
    • 15. Wang K, Ma W, Wang J et al. Tumor-stroma ratio is an independent predictor for survival in esophageal squamous cell carcinoma. J. Thorac. Oncol. 7(9), 1457–1461 (2012).
    • 16. Shimizu K, Okita R, Nakata M. Clinical significance of the tumor microenvironment in non-small cell lung cancer. Ann. Transl. Med. 1(2), 20 (2013).
    • 17. Wenbo L, Wang J. Uncovering the underlying mechanism of cancer tumorigenesis and development under an immune microenvironment from global quantification of the landscape. J. R. Soc. Interface 14(131), 20170105 (2017).
    • 18. Li X, Wenes M, Romero P, Huang SC, Fendt SM, Ho PC. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat. Rev. Clin. Oncol. 16(7), 425–441 (2019).
    • 19. Ojlert AK, Halvorsen AR, Nebdal D et al. The immune microenvironment in non-small cell lung cancer is predictive of prognosis after surgery. Mol. Oncol. 13(5), 1166–1179 (2019).
    • 20. Ascierto PA, Bifulco C, Buonaguro L et al. Perspectives in immunotherapy: meeting report from the “Immunotherapy Bridge 2018” (28-29 November, 2018, Naples, Italy). J. Immunother. Cancer 7(1), 332 (2019).
    • 21. Oliver AJ, Davey AS, Keam SP et al. Tissue-specific tumor microenvironments influence responses to immunotherapies. Clin. Transl. Immunol. 8(11), e1094 (2019).
    • 22. Blons H, Garinet S, Laurent-Puig P, Oudart JB. Molecular markers and prediction of response to immunotherapy in non-small cell lung cancer, an update. J. Thorac. Dis. 11(Suppl. 1), S25–S36 (2019).
    • 23. Skoulidis F, Goldberg ME, Greenawalt DM et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 8(7), 822–835 (2018).
    • 24. Dong ZY, Zhong WZ, Zhang XC et al. Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma. Clin. Cancer Res. 23(12), 3012–3024 (2017).
    • 25. Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases–elimination, equilibrium and escape. Curr. Opin. Immunol. 27, 16–25 (2014).
    • 26. Tuminello S, Veluswamy R, Lieberman-Cribbin W et al. Prognostic value of immune cells in the tumor microenvironment of early-stage lung cancer: a meta-analysis. Oncotarget 10(67), 7142–7155 (2019).
    • 27. Gentles AJ, Newman AM, Liu CL et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21(8), 938–945 (2015).
    • 28. Sumitomo R, Hirai T, Fujita M, Murakami H, Otake Y, Huang CL. M2 tumor-associated macrophages promote tumor progression in non-small-cell lung cancer. Exp. Ther. Med. 18(6), 4490–4498 (2019).
    • 29. Sun Y, Xu J. TCF-4 regulated lncRNA-XIST promotes M2 polarization of macrophages and is associated with lung cancer. Onco Targets Ther. 12, 8055–8062 (2019).
    • 30. Jackute J, Zemaitis M, Pranys D et al. Distribution of M1 and M2 macrophages in tumor islets and stroma in relation to prognosis of non-small cell lung cancer. BMC Immunol. 19(1), 3 (2018).
    • 31. Zhang R, Li F, Li H, Yu J, Ren X. The clinical significance of memory T cells and its subsets in gastric cancer. Clin. Transl. Oncol. 16(3), 257–265 (2014).
    • 32. Dadabayev AR, Sandel MH, Menon AG et al. Dendritic cells in colorectal cancer correlate with other tumor-infiltrating immune cells. Cancer immunol. Immunother. 53(11), 978–986 (2004).
    • 33. Vahidi Y, Faghih Z, Talei AR, Doroudchi M, Ghaderi A. Memory CD4(+) T cell subsets in tumor draining lymph nodes of breast cancer patients: a focus on T stem cell memory cells. Cell. Oncol. 41(1), 1–11 (2018).
    • 34. Huang HY, Rivas-Caicedo A, Renevey F et al. Identification of a new subset of lymph node stromal cells involved in regulating plasma cell homeostasis. Proc. Natl Acad. Sci. USA 115(29), E6826–E6835 (2018).
    • 35. Lohr M, Edlund K, Botling J et al. The prognostic relevance of tumour-infiltrating plasma cells and immunoglobulin kappa C indicates an important role of the humoral immune response in non-small cell lung cancer. Cancer Lett. 333(2), 222–228 (2013).
    • 36. Xu F, Chen JX, Yang XB et al. Analysis of lung adenocarcinoma subtypes based on immune signatures identifies clinical implications for cancer therapy. Mol. Ther. Oncolytics 17, 241–249 (2020).
    • 37. Mony JT, Schuchert MJ. Prognostic implications of heterogeneity in intra-tumoral immune composition for recurrence in early stage lung cancer. Front. Immunol. 9, 2298 (2018).
    • 38. Li M, Li A, He R et al. Gene polymorphism of cytochrome P450 significantly affects lung cancer susceptibility. Cancer Med. 8(10), 4892–4905 (2019).
    • 39. Hoxhaj G, Manning BD. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat. Rev. Cancer 20(2), 74–88 (.2020).
    • 40. Yuan S, Dong Y, Peng L et al. Tumor-associated macrophages affect the biological behavior of lung adenocarcinoma A549 cells through the PI3K/AKT signaling pathway. Oncol. Lett. 18(2), 1840–1846 (2019).
    • 41. Groner B, Von Manstein V. Jak Stat signaling and cancer: opportunities, benefits and side effects of targeted inhibition. Mol. Cell. Endocrinol. 451, 1–14 (2017).
    • 42. Tang S, Yuan X, Song J, Chen Y, Tan X, Li Q. Association analyses of the JAK/STAT signaling pathway with the progression and prognosis of colon cancer. Oncol. Lett. 17(1), 159–164 (2019).
    • 43. Mohrherr J, Haber M, Breitenecker K et al. JAK-STAT inhibition impairs K-RAS-driven lung adenocarcinoma progression. Int. J. Cancer 145(12), 3376–3388 (2019).
    • 44. Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. A ceRNA hypothesis: the Rosetta Stone of a hidden RNA language? Cell 146(3), 353–358 (2011).
    • 45. Huang R, Meng T, Chen R et al. The construction and analysis of tumor-infiltrating immune cell and ceRNA networks in recurrent soft tissue sarcoma. Aging (Albany NY). 11(22), 10116–10143 (2019).
    • 46. Liu Z, Mi M, Li X, Zheng X, Wu G, Zhang L. lncRNA OSTN-AS1 may represent a novel immune-related prognostic marker for triple-negative breast cancer based on Integrated analysis of a ceRNA network. Front. Genet. 10, 850 (2019).
    • 47. Sun F, Liang W, Tang K, Hong M, Qian J. Profiling the lncRNA-miRNA-mRNA ceRNA network to reveal potential crosstalk between inflammatory bowel disease and colorectal cancer. PeerJ 7, e7451 (2019).
    • 48. Du W, Sun J, Gu J, Zhang S, Zhang T. Bioinformatics analysis of LINC00426 expression in lung cancer and its correlation with patients' prognosis. Thorac. Cancer 11(1), 150–155 (.2020).
    • 49. Guo Y, Hu Y, Hu M, He J, Li B. Long non-coding RNA ZEB2-AS1 promotes proliferation and inhibits apoptosis in human lung cancer cells. Oncol. Lett. 15(4), 5220–5226 (2018).