New Insight of Tumor Microenvironment in Non-Small Cell Lung Cancer

New Insight of Tumor Microenvironment in Non-Small Cell Lung Cancer

As in all living cells, cancer cells are evaluating is very important in the frames of tumor development and treatment. As in all evolutionaryprocess, the environment of living cells has an important role. Tumor cells have an environment is called tumor microenvironment, affectsthe therapeutic response and clinical results. Tumor microenvironments involve various cell types, extracellular matrix substances that arein the niche of cancer cells. The microenvironment is not only important in tumorigenesis but it is effective on therapeutic efficacy. In thisreview, we carried out the interaction between non-small cell lung cancer and its microenvironment to point out the significance of the tumorenvironment.

___

  • 1. Ahmad A, Gadgeel SM. Lung Cancer and Personalized Medicine: Novel Therapies and Clinical Management. Preface. Adv Exp Med Biol. 2016;890:v-vi. [CrossRef]
  • 2. Donnem T, Andersen S, Al-Saad S, Al-Shibli K, Busund LT, Bremnes RM. Prognostic impact of angiogenic markers in non-small-cell lung cancer is related to tumor size. Clin Lung Cancer 2011;12:106-15. 10.1016/j.cllc.2011.03.00
  • 3. Wood SL, Pernemalm M, Crosbie PA, Whetton AD. The role of the tumor-microenvironment in lung cancer-metastasis and its relationship to potential therapeutic targets. Cancer Treat Rev 2014;40:558–566. [CrossRef]
  • 4. Kinoshita T, Ishii G, Hiraoka N, et al. Forkhead box P3 regulatory T cells coexisting with cancer associated fibroblasts are correlated with a poor outcome in lung adenocarcinoma. Cancer Sci 2013;104:409– 415. [CrossRef]
  • 5. Paulsson J, Micke P. Prognostic relevance of cancer-associated fibroblasts in human cancer. Sem Cancer Biol 2014;25:61–68. [CrossRef]
  • 6. Ma J, Liu L, Che G, Yu N, Dai F, You Z. The M1 form of tumorassociated macrophages in non-small cell lung cancer is positively associated with survival time. BMC Cancer 2010;10. [CrossRef]
  • 7. Dieu-Nosjean MC, Antoine M, Danel C, et al. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol 2008;26:4410–4417. [CrossRef]
  • 8. Chaisemartin L, Goc J, Damotte D, et al. Characterization of chemokines and adhesion molecules associated with T cell presence in tertiary lymphoid structures in human lung cancer. Cancer Res 2011;71:6391–6399. [CrossRef]
  • 9. Goc J, Germain C, Vo-Bourgais TKD, et al. Dendritic cells in tumorassociated tertiary lymphoid structures signal a th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ t cells. Cancer Res 2014;74:705–715. [CrossRef]
  • 10. Remark R, Alifano M, Cremer I, et al. Characteristics and clinical impacts of the immune environments in colorectal and renal cell carcinoma lung metastases: Influence of tumor origin. Clin Cancer Res 2013;19:4079–4091. [CrossRef]
  • 11. Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol 2006;6:295–307. [CrossRef]
  • 12. Wald O, Shapira OM, Izhar U. CXCR4/CXCL12 axis in non small cell lung cancer (NSCLC) pathologic roles and therapeutic potential. Theranostics 2013;3:26–33. [CrossRef]
  • 13. Kirshberg S, Izhar U, Amir G, et al. Involvement of CCR6/CCL20/ IL-17 axis in NSCLC disease progression. PLoS One 2011;6:e24856. [CrossRef]
  • 14. Ramalingam SS, Owonikoko TK, Khuri FR. Lung cancer: New biological insights and recent therapeutic advances. CA Cancer J Clin 2011;61:91–112. [CrossRef]
  • 15. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252–264. [CrossRef]
  • 16. Mu CY, Huang JA, Chen Y, Chen C, Zhang XG. High expression of PDL1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation. Med Oncol 2011;28:682–688. [CrossRef]
  • 17. Salvi S, Fontana V, Boccardo S, et al. Evaluation of CTLA-4 expression and relevance as a novel prognostic factor in patients with non-small cell lung cancer. Cancer Immunol Immunother 2012;61:1463–1472. [CrossRef]
  • 18. Ménétrier-Caux C, Gobert M, Caux C. Differences in tumor regulatory T-cell localization and activation status impact patient outcome. Cancer Res 2009;69:7895–7898. [CrossRef]
  • 19. Germain C, Gnjatic S, Tamzalit F, et al. Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer. Am J Respir Crit Care Med 2014;189:832– 44. [CrossRef]
  • 20. Al-Shibli K, Al-Saad S, Donnem T, Persson M, Bremnes RM, Busund LT. The prognostic value of intraepithelial and stromal innate immune system cells in non-small cell lung carcinoma. Histopathology 2009;55:301–312. [CrossRef]
  • 21. Hanahan D, Coussens LM. Accessories to the Crime: Functions of Cells Recruited to the Tumor Microenvironment. Cancer Cell 2012;21:309–322. [CrossRef]
  • 22. Ilie M, Hofman V, Ortholan C, et al. Predictive clinical outcome of the intratumoral CD66b-positive neutrophil-to-CD8-positive T-cell ratio in patients with resectable nonsmall cell lung cancer. Cancer 2012;118:1726– 1737. [CrossRef]
  • 23. Graves E, Maity A, Le Q. The tumor microenvironment in Non-SmalCell Lung Cancer. Semin Radiat Oncol 2010;20:156–163. [CrossRef]
  • 24. Sica A, Allavena P, Mantovani A. Cancer related inflammation: The macrophage connection. Cancer Lett 2008;267:204–215. [CrossRef]
  • 25. Kumar S, Chan CJ, Coussens LM. Inflammation and Cancer. In: Immunity to Pathogens and Tumors . Elsevier Inc.; 2016. pp.406–415. [CrossRef]
  • 26. Ben-Baruch A. Inflammation-associated immune suppression in cancer: The roles played by cytokines, chemokines and additional mediators. Semin Cancer Biol 2006;16:38–52. [CrossRef]
  • 27. Aggarwal BB, Vijayalekshmi RV, Sung B. Targeting inflammatory pathways for prevention and therapy of cancer: Short-term friend, long-term foe. Clin Cancer Res 2009;15:425–430. [CrossRef]
  • 28. Lin GN, Peng JW, Liu PP, Liu DY, Xiao JJ, Chen XQ. Elevated neutrophilto-lymphocyte ratio predicts poor outcome in patients with advanced non-small-cell lung cancer receiving first-line gefitinib or erlotinib treatment. Asia Pac J Clin Oncol 2017;13:e189–e194. [CrossRef]
  • 29. Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer 2004;4:71–78. [CrossRef]
  • 30. Woods NT, Monteiro AN, Thompson ZJ, et al. Interleukin polymorphisms associated with overall survival, disease-free survival, and recurrence in non-small cell lung cancer patients. Mol Carcinog 2015;54:E172–E184. [CrossRef]
  • 31. Wang XF, Zhu YT, Wang JJ, et al. The prognostic value of interleukin-17 in lung cancer: A systematic review with meta-analysis based on Chinese patients. PLoS One 2017;12:e0185168. [CrossRef]
  • 32. Gomes M, Coelho A, Araújo A, et al. IL-6 polymorphism in non-small cell lung cancer: a prognostic value? Tumor Biol 2015;36:3679–3684. [CrossRef]
  • 33. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the Treatment of Non-Small-Cell Lung Cancer. N Engl J Med 2015;372:2018–2028. [CrossRef]
  • 34. Semenza GL. Hypoxia-inducible factors: Mediators of cancer progression and targets for cancer therapy. Trends Pharmacol Sci 2012;33:207–214. [CrossRef]
  • 35. Karetsi E, Ioannou M, Kerenidi T, et al. Differential expression of hypoxia-inducible factor 1α in non-small cell lung cancer and small cell lung cancer. Clinics (Sao Paulo) 2012;67:1373–1378. [CrossRef]
  • 36. Adini A, Adini I, Chi Z, Derda R, et al. A novel strategy to enhance angiogenesis in vivo using the small VEGF-binding peptide PR1P. Angiogenesis 2017;20:399–408. [CrossRef]
  • 37. Swinson DEB, Jones JL, Cox G, Richardson D, Harris AL, O’Byrne KJ. Hypoxia-inducible factor-1α in non small cell lung cancer: Relation to growth factor, protease and apoptosis pathways. Int J Cancer 2004;111:43–50. [CrossRef]
  • 38. Frezzetti D, Gallo M, Maiello MR, et al. VEGF as a potential target in lung cancer. Expert Opin Ther Targets 2017;21:959–966. [CrossRef]
  • 39. Enatsu S, Iwasaki A, Shirakusa T, et al. Expression of hypoxiainducible factor-1 alpha and its prognostic significance in smallsized adenocarcinomas of the lung. Eur J Cardio-thoracic Surg 2006;29:891–895. [CrossRef]
  • 40. Herbst RS, Onn A, Sandler A. Angiogenesis and lung cancer: Prognostic and therapeutic implications. J Clin Oncol 2005;23:3243– 3256. [CrossRef]
  • 41. Chen Z, Fillmore CM, Hammerman PS, Kim CF, Wong KK. Non-smallcell lung cancers: A heterogeneous set of diseases. Nat Rev Cancer 2014;14:535–546. [CrossRef]
  • 42. Kargl J, Busch SE, Yang GHY, et al. Neutrophils dominate the immune cell composition in non-small cell lung cancer. Nat Commun 2017;8:14381. [CrossRef]
  • 43. Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer 2016;16:582–598. [CrossRef]
  • 44. Banat GA, Tretyn A, Pullamsetti SS, et al. Immune and Inflammatory Cell Composition of Human Lung Cancer Stroma. PLoS ONE 2015;10:e0139073. [CrossRef]
  • 45. Salmon H, Franciszkiewicz K, Damotte D, et al. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J Clin Invest 2012;122:899–910. [CrossRef]
  • 46. Villano JL, Durbin EB, Normandeau C, Thakkar JP, Moirangthem V, Davis FG. Incidence of brain metastasis at initial presentation of lung cancer. Neuro Oncol 2015;17:122–28. [CrossRef]
  • 47. Wang L, Cossette SM, Rarick KR, et al. Astrocytes directly influence tumor cell invasion and metastasis in vivo. PLoS One 2013;8:e80933. [CrossRef]
  • 48. Valiente M, Obenauf AC, Jin X, et al. Serpins promote cancer cell survival and vascular Co-option in brain metastasis. Cell 2014;156:1002–1016. [CrossRef]
  • 49. Müller P, Rothschild SI, Arnold W, et al. Metastatic spread in patients with non-small cell lung cancer is associated with a reduced density of tumor–infiltrating T cells. Cancer Immunol Immunother 2016;65:1–11. [CrossRef]
  • 50. Reticker-Flynn NE, Bhatia SN. Aberrant glycosylation promotes lung cancer metastasis through adhesion to galectins in the metastatic niche. Cancer Discov 2015;5:168–181. [CrossRef]