254
Views
0
CrossRef citations to date
0
Altmetric
Articles

Tertiary Lymphoid Structures, Immune Response, and Prognostic Relevance in Non-Small Cell Lung Cancer

, , &
Pages 48-57 | Received 30 Jan 2022, Accepted 12 Oct 2022, Published online: 02 Nov 2022

References

  • Bashford EF, Murray JA, Cramer W. The natural and induced resistance of mice to the growth of cancer. Proc R Soc Lond B. 1907;79:164–187.
  • Garon EB, Hellmann MD, Rizvi NA, Carcereny E, Leighl NB, Ahn MJ, et al. Five-year overall survival for patients with advanced non–small-cell lung cancer treated with pembrolizumab: results from the phase I KEYNOTE-001 study. J Clin Oncol. 2019;37(28):2518–2527.
  • Carroll R, Bortolini M, Calleja A, Munro R, Kong S, Daumont MJ, et al. Trends in treatment patterns and survival outcomes in advanced non-small cell lung cancer: a Canadian population-based real-world analysis. BMC Cancer. 2022;22(1):255. doi:10.1186/s12885-022-09342-5.
  • De Giglio A, Di Federico A, Nuvola G, Deiana C, Gelsomino F. The landscape of immunotherapy in advanced NSCLC: driving beyond PD-1/PD-L1 inhibitors (CTLA-4, LAG3, IDO, OX40, TIGIT, vaccines). Curr Oncol Rep. 2021;23(11):126. doi:10.1007/s11912-021-01124-9.
  • Moyron-Quiroz JE, Rangel-Moreno J, Kusser K, Hartson L, Sprague F, Goodrich S, et al. Role of inducible bronchus-associated lymphoid tissue (iBALT) in respiratory immunity. Nat Med. 2004;10(9):927–934. doi:10.1038/nm1091.
  • Ager A. High endothelial venules and other blood vessels: critical regulators of lymphoid organ development and function. Front Immunol. 2017;8:45. doi:10.3389/fimmu.2017.00045.
  • Hiraoka N, Ino Y, Yamazaki-Itoh R. Tertiary lymphoid organs in cancer tissues. Front Immunol. 2016;7:244. doi:10.3389/fimmu.2016.00244.
  • Gago da Graça C, van Baarsen LGM, Mebius RE. Tertiary lymphoid structures: diversity in their development, composition, and role. J Immunol. 2021;206(2):273–281. doi:10.4049/jimmunol.2000873.
  • Lin L, Hu X, Zhang H, Hu H. Tertiary lymphoid organs in cancer immunology: mechanisms and the new strategy for immunotherapy. Front Immunol. 2019;10:1398. doi:10.3389/fimmu.2019.01398.
  • Linares JF, Cid-Diaz T, Duran A, Osrodek M, Martinez-Ordoñez A, Reina-Campos M, et al. The lactate-NAD+ axis activates cancer-associated fibroblasts by downregulating p62. Cell Rep. 2022;39(6):110792. doi:10.1016/j.celrep.2022.110792.
  • Sivridis E, Giatromanolaki A, Koukourakis MI. Proliferating fibroblasts at the invading tumour edge of colorectal adenocarcinomas are associated with endogenous markers of hypoxia, acidity, and oxidative stress. J Clin Pathol. 2005;58(10):1033–1038. doi:10.1136/jcp.2005.026260.
  • Giatromanolaki A, Koukourakis IM, Chatzipantelis P, Kouroupi M, Balaska K, Koukourakis MI. Rectal cancer induces a regulatory lymphocytic phenotype in the tumor-draining lymph nodes to promote cancer cell installation. Immunol Res. 2020;68(6):363–372. doi:10.1007/s12026-020-09161-5.
  • Giatromanolaki A, Mitrakas A, Anestopoulos I, Kontosis A, Koukourakis IM, Pappa A, et al. Expression of CD47 and SIRPα macrophage immune-checkpoint pathway in non-small-cell lung cancer. Cancers. 2022;14(7):1801. doi:10.3390/cancers14071801.
  • Giatromanolaki A, Banham AH, Harris AL, Koukourakis MI. FOXP3 infiltrating lymphocyte density and PD-L1 expression inoperable non-small cell lung carcinoma. Exp Lung Res. 2019;45(3-4):76–83. doi:10.1080/01902148.2019.1617371.
  • Giatromanolaki A, Harris AL, Banham AH, Contrafouris CA, Koukourakis MI. Carbonic anhydrase 9 (CA9) expression in non-small-cell lung cancer: correlation with regulatory FOXP3 + T-cell tumour stroma infiltration. Br J Cancer. 2020;122(8):1205–1210. doi:10.1038/s41416-020-0756-3.
  • Mebius RE. Organogenesis of lymphoid tissues. Nat Rev Immunol. 2003;3(4):292–303. doi:10.1038/nri1054.
  • Rangel-Moreno J, Carragher DM, de la Luz Garcia-Hernandez M, Hwang JY, Kusser K, Hartson L, et al. The development of inducible bronchus-associated lymphoid tissue depends on IL-17. Nat Immunol. 2011;12(7):639–646. doi:10.1038/ni.2053.
  • Watanabe M, Ueno Y, Yajima T, Okamoto S, Hayashi T, Yamazaki M, et al. Interleukin 7 transgenic mice develop chronic colitis with decreased interleukin 7 protein accumulation in the colonic mucosa. J Exp Med. 1998;187(3):389–402. doi:10.1084/jem.187.3.389.
  • Lee HJ, Kim JY, Park IA, Song IH, Yu JH, Ahn JH, Gong G. Prognostic significance of tumor-infiltrating lymphocytes and the tertiary lymphoid structures in HER2-positive breast cancer treated with adjuvant trastuzumab. Am J Clin Pathol. 2015;144(2):278–288. doi:10.1309/AJCPIXUYDVZ0RZ3G.
  • Sofopoulos M, Fortis SP, Vaxevanis CK, Sotiriadou NN, Arnogiannaki N, Ardavanis A, et al. The prognostic significance of peritumoral tertiary lymphoid structures in breast cancer. Cancer Immunol Immunother. 2019;68(11):1733–1745. doi:10.1007/s00262-019-02407-8.
  • Liu X, Tsang JYS, Hlaing T, Hu J, Ni YB, Chan SK, et al. Distinct tertiary lymphoid structure associations and their prognostic relevance in HER2 positive and negative breast cancers. Oncologist. 2017;22(11):1316–1324. doi:10.1634/theoncologist.2017-0029.
  • Lee HJ, Park IA, Song IH, Shin SJ, Kim JY, Yu JH, Gong G. Tertiary lymphoid structures: prognostic significance and relationship with tumour-infiltrating lymphocytes in triple-negative breast cancer. J Clin Pathol. 2016;69(5):422–430. doi:10.1136/jclinpath-2015-203089.
  • Di Caro G, Bergomas F, Grizzi F, Doni A, Bianchi P, Malesci A, et al. Occurrence of tertiary lymphoid tissue is associated with T-cell infiltration and predicts better prognosis in early-stage colorectal cancers. Clin Cancer Res. 2014;20(8):2147–2158. doi:10.1158/1078-0432.CCR-13-2590.
  • Ahmed A, Halama N. Tertiary lymphoid structures in colorectal cancer liver metastases: association with immunological and clinical parameters and chemotherapy response. Anticancer Res. 2020;40(11):6367–6373. doi:10.21873/anticanres.14657.
  • Rakaee M, Kilvaer TK, Jamaly S, Berg T, Paulsen EE, Berglund M, et al. Tertiary lymphoid structure score: a promising approach to refine the TNM staging in resected non-small cell lung cancer. Br J Cancer. 2021;124(10):1680–1689. doi:10.1038/s41416-021-01307-y.
  • Siliņa K, Soltermann A, Attar FM, Casanova R, Uckeley ZM, Thut H, et al. Germinal centers determine the prognostic relevance of tertiary lymphoid structures and are impaired by corticosteroids in lung squamous cell carcinoma. Cancer Res. 2018;78(5):1308–1320. doi:10.1158/0008-5472.CAN-17-1987.
  • Dieu-Nosjean MC, Antoine M, Danel C, Heudes D, Wislez M, Poulot V, et al. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol. 2008;26(27):4410–4417. doi:10.1200/JCO.2007.15.0284.
  • Germain C, Gnjatic S, Tamzalit F, Knockaert S, Remark R, Goc J, et al. The 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(7):832–844. doi:10.1164/rccm.201309-1611OC.
  • Helmink BA, Reddy SM, Gao J, Zhang S, Basar R, Thakur R, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 2020;577(7791):549–555. doi:10.1038/s41586-019-1922-8.
  • Liu Y, Xiang F, Huang Y, Shi L, Hu C, Yang Y, et al. Interleukin-22 promotes aerobic glycolysis associated with tumor progression via targeting hexokinase-2 in human colon cancer cells. Oncotarget. 2017;8(15):25372–25383. doi:10.18632/oncotarget.15913.
  • Straus DS. TNFα and IL-17 cooperatively stimulate glucose metabolism and growth factor production in human colorectal cancer cells. Mol Cancer. 2013;12:78. doi:10.1186/1476-4598-12-78.
  • Calcinotto A, Filipazzi P, Grioni M, Iero M, De Milito A, Ricupito A, et al. Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes. Cancer Res. 2012;72(11):2746–2756.
  • Caslin HL, Abebayehu D, Abdul Qayum A, Haque TT, Taruselli MT, Paez PA, et al. Lactic acid inhibits lipopolysaccharide-induced mast cell function by limiting glycolysis and ATP availability. J Immunol. 2019;203(2):453–464. doi:10.4049/jimmunol.1801005.
  • Kroeger DR, Milne K, Nelson BH. Tumor-infiltrating plasma cells are associated with tertiary lymphoid structures, cytolytic T-cell responses, and superior prognosis in ovarian cancer. Clin Cancer Res. 2016;22(12):3005–3015. doi:10.1158/1078-0432.CCR-15-2762.
  • Goc J, Germain C, Vo-Bourgais TK, Lupo A, Klein C, Knockaert S, et al. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res. 2014;74(3):705–715. doi:10.1158/0008-5472.CAN-13-1342.
  • Vanhersecke L, Brunet M, Guégan JP, Rey C, Bougouin A, Cousin S, et al. Mature tertiary lymphoid structures predict immune checkpoint inhibitor efficacy in solid tumors independently of PD-L1 expression. Nat Cancer. 2021;2(8):794–802. doi:10.1038/s43018-021-00232-6.
  • Ikeda A, Ogino T, Kayama H, Okuzaki D, Nishimura J, Fujino S, et al. Human NKp44+ group 3 innate lymphoid cells associate with tumor-associated tertiary lymphoid structures in colorectal cancer. Cancer Immunol Res. 2020;8(6):724–731. doi:10.1158/2326-6066.CIR-19-0775.
  • Bruchard M, Ghiringhelli F. Deciphering the roles of innate lymphoid cells in cancer. Front Immunol. 2019;10:656. doi:10.3389/fimmu.2019.00656.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.