Tertiary lymphoid structures (lymphoid neogenesis)

Abstract

Secondary lymphoid organs are integral to initiation and execution of adaptive immune responses. These organs provide a setting for interactions between antigen-specific lymphocytes and antigen-presenting cells recruited from local infected or inflamed tissues. Secondary lymphoid organs develop as a part of a genetically preprogrammed process during embryogenesis. However, organogenesis of secondary lymphoid tissues can also be recapitulated in adulthood during de novo lymphoid neogenesis of tertiary lymphoid structures (TLSs). These ectopic lymphoid-like structures form in the inflamed tissues afflicted by various pathological conditions, including cancer, autoimmunity, infection, or allograft rejection. Studies are beginning to shed light on the function of such structures in different disease settings, raising important questions regarding their contribution to progression or resolution of disease. Data show an association between the tumor-associated TLSs and a favorable prognosis in various types of human cancer, attracting the speculation that TLSs support effective local antitumor immune responses. However, definitive evidence for the role for TLSs in fostering immune responses in vivo are lacking, with current data remaining largely correlative by nature. In this review, we will compare and contrast TLS neogenesis with the development of a prototypic SLO, the LN. Importantly, we will discuss current knowledge surrounding the function of TLSs, specifically within cancer. Scopus, Web of Science, MedLine, The Cochrane Library, EMBASE, Global Health, CyberLeninka, RISC were used to search for the required literature.

Keywords:review; secondary lymphoid organs; tertiary lymphoid structures; lymphoid neogenesis

Received 20.11.2018. Accepted 16.02.2019.

For citation: Gantsev Sh.Kh., Rustamkhanov R.A., Gantsev K.Sh., Kzyrgalin Sh.R. Tertiary lymphoid structures (lymphoid neogenesis). Immunologiya. 2019; 40 (2): 58-63. doi: 10.24411/0206-4952-2019-12008. (in Russian)

Acknowledgments. The study had no sponsorship.

Conflict of interest. The authors declare no conflict of interest.

References

1. Pavlenko V.I., Sayapina I.Yu. Cells and organs of the immune system: manual. Blagoveshchensk, 2017. (in Russian)

2. Van de Pavert S.A., Mebius R.E. New insights into the development of lymphoid tissues. Nat. Rev. Immunol. 2010; 10: 664-74. doi: 10.1038/nri2832.

3. Randall T.D., Carragher D.M., Rangel-Moreno J. Development of secondary lymphoid organs. Annu. Rev. Immunol. 2008; 26: 627-50. doi: 10.1146/annurev.immunol.26.021607.090257.

4. Drayton D.L., Liao S., Mounzer R.H., Ruddle N.H. Lymphoid organ development: from ontogeny to neogenesis. Nat. Immunol. 2006; 7: 344-53. doi: 10.1038/ni1330.

5. Dieu-Nosjean M.-C., Goc J., Giraldo N.A., Sautes-Fridman C., et al. Tertiary lymphoid structures in cancer and beyond. Trends Immunol. 2014; 35: 571-80. doi: 10.1016/j.it.2014.09.006.

6. Sautes-Fridman C., Lawand M., Giraldo N.A., et al. Tertiary lymphoid structures in cancers: prognostic value, regulation, and manipulation for therapeutic intervention. Front. Immunol. 2016; 7: 407. doi: 10.3389/fimmu.2016.00407.

7. Pitzalis C., Jones G.W., Bombardieri M., Jones S.A. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat. Rev. Immunol. 2014; 14: 447-62. doi: 10.1038/nri3700.

8. Neyt K., Perros F., GeurtsvanKessel C.H., Hammad H., et al. Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol. 2012; 33: 297-305. doi: 10.1016/j.it.2012.04.006.

9. Lu T.T., Browning J.L. Role of the lymphotoxin/LIGHT system in the development and maintenance of reticular networks and vasculature in lymphoid tissues. Front. Immunol. 2014; 5: 47. doi: 10.3389/fimmu.2014.00047.

10. Stranford S., Ruddle N.H. Follicular dendritic cells, conduits, lymphatic vessels, and high endothelial venules in tertiary lymphoid organs: parallels with lymph node stroma. Front. Immunol. 2012; 3: 350. doi: 10.3389/fimmu.2012.00350.

11. Aloisi F., Pujol-Borrell R. Lymphoid neogenesis in chronic inflammatory diseases. Nat. Rev. Immunol. 2006; 6: 205-17. doi: 10.1038/nri1786.

12. Carragher D.M., Rangel-Moreno J., Randall T.D. Ectopic lymphoid tissues and local immunity. Semin. Immunol. 2008; 20: 26-42. doi: 10.1016/j.smim.2007.12.004.

13. Takemura S., Braun A., Crowson C., Kurtin PJ., et al. Lymphoid neogenesis in rheumatoid synovitis. J. Immunol. 2001; 167: 1072-80. doi: 10.4049/jimmunol.167.2.1072.

14. Manzo A., Paoletti S., Carulli M., Blades M.C., et al. Systematic micro anatomical analysis of CXCL13 and CCL21 in situ production and progressive lymphoid organization in rheumatoid synovitis. Eur. J. Immunol. 2005; 35: 1347-59. doi: 10.1002/eji.200425830.

15. Cantaert T., Kolln J., Timmer T., van der Pouw Kraan T.C., et al. B lymphocyte autoimmunity in rheumatoid synovitis is independent of ectopic lymphoid neogenesis. J. Immunol. 2008; 181: 785-94. doi: 10.4049/jimmunol.181.1.785.

16. Guo J., Qian J., Zhang R. The pathological features of ectopic lymphoid neogenesis in idiopathic dacryoadenitis. BMC Ophthalmol. 2016; 16: 66. doi: 10.1186/s12886-016-0250-0.

17. Colbeck E.J., Ager A., Gallimore A., Jones G.W. Tertiary Lymphoid Structures in Cancer: Drivers of Antitumor Immunity, Immunosuppression, or Bystander Sentinels in Disease? Front. Immunol. 2017; 8: 1830. doi: 10.3389/fimmu.2017.01830.

18. Konenkov V.I., Borodin Yu.I., Lyubarsky M.S. Lymphology. Novosibirsk: Manuscript, 2012: 1096 p. (in Russian)

19. Aloisi F., Borrell P-R. Lymphoid neogenesis in chronic inflammatory diseases. Nat. Rev. Immunol. 2006; 6: 205-17.

20. Lee J.-W., Epardaud M., Sun J., Becker J.E., et al. Peripheral antigen display by lymph node stroma promotes T cell tolerance to intestinal self. Nat. Immunol. 2007; 8: 181-90. doi: 10.1038/ni1427.

21. Goc J., Fridman W.-H., Sautes-Fridman C., Dieu-Nosjean M.-C. Characteristics of tertiary lymphoid structures in primary cancers. Oncoimmunology. 2013; 2: e26836. doi: 10.4161/onci.26836.

22. Joyce J.A., Fearon D.T. T cell exclusion, immune privilege, and the tumor microenvironment. Science. 2015; 348: 74-80. doi: 10.1126/science.aaa6204.

23. Martinet L., Garrido I., Filleron T., Le Guellec S., et al. Human solid tumors contain high endothelial venules: association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res. 2011; 71: 5678-87. doi: 10.1158/0008-5472.CAN-11-0431.

24. de Chaisemartin L., Goc J., Damotte D., Validire P, 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: 6391-9. doi: 10.1158/0008-5472.CAN-11-0952.

25. Goc J., Germain C., Vo-Bourgais T.K.D., Lupo A., 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: 705-15. doi: 10.1158/0008-5472.CAN-13-1342.

26. Bento D.C., Jones E., Junaid S., Tull J., et al. High endothelial venules are rare in colorectal cancers but accumulate in extra-tumoral areas with disease progression. Oncoimmunology. 2015; 4: e974374. doi: 10.4161/2162402X.2014.974374.

27. Di Caro G., Bergomas F., Grizzi F., Doni 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: 2147-58. doi: 10.1158/1078-0432.CCR-13-2590.

28. Coppola D., Nebozhyn M., Khalil F., Dai H., et al. Unique ectopic lymph node-like structures present in human primary colorectal carcinoma are identified by immune gene array profiling. Am. J. Pathol. 2011; 179: 37^5. doi: 10.1016/j.ajpath.2011.03.007.

29. Remark R., Alifano M., Cremer I., Lupo A., 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: 4079-91. doi: 10.1158/1078-0432.CCR-12-3847.

30. Gu-Trantien C., Loi S., Garaud S., Equeter C., et al. CD4? follicular helper T cell infiltration predicts breast cancer survival. J. Clin. Invest. 2013; 123: 2873-92. doi: 10.1172/JCI67428.

31. Martinet L., Filleron T., Le Guellec S., Rochaix P, et al. High endothelial venule blood vessels for tumor-infiltrating lymphocytes are associated with lymphotoxin р-producing dendritic cells in human breast cancer. J. Immunol. 2013; 191: 2001-8. doi: 10.4049/jimmunol.1300872.

32. Figenschau S.L., Fismen S., Fenton K.A., Fenton C., et al. Tertiary lymphoid structures are associated with higher tumor grade in primary operable breast cancer patients. BMC Cancer. 2015; 15: 101. doi: 10.1186/s12885-015-1116-1.

33. Song I.H., Heo S.-H., Bang W.S., Park H.S., et al. Predictive value of tertiary lymphoid structures assessed by high endothelial venule counts in the neoadjuvant setting of triple-negative breast cancer. Cancer Res Treat. 2017; 49: 399-407. doi: 10.4143/crt.2016.215.

34. Martinet L., Le Guellec S., Filleron T., Lamant L., et al. High endothelial venules (HEVs) in human melanoma lesions: major gateways for tumor-infiltrating lymphocytes. Oncoimmunology. 2012; 1: 829-39. doi: 10.4161/onci.20492.

35. Messina J.L., Fenstermacher D.A., Eschrich S., Qu X., et al. 12-Chemokine gene signature identifies lymph node-like structures in melanoma: potential for patient selection for immunotherapy? Sci. Rep. 2012; 2: 765. doi: 10.1038/srep00765.

36. Avram G., Sanchez-Sendra B., Martin J.M., Terradez L., et al. The density and type of MECA-79-positive high endothelial venules correlate with lymphocytic infiltration and tumour regression in primary cutaneous melanoma. Histopathology. 2013; 63: 852-61. doi: 10.1111/his.12235.

37. Garcia-Hernandez M.L., Uribe-Uribe N.O., Espinosa-Gonzalez R., Kast W.M., et al. A unique cellular and molecular microenvironment is present in tertiary lymphoid organs of patients with spontaneous prostate cancer regression. Front. Immunol. 2017; 8: 563. doi: 10.3389/fimmu.2017.00563.

38. Cipponi A., Mercier M., Seremet T., Baurain J.-F., et al. Neogenesis of lymphoid structures and antibody responses occur in human melanoma metastases. Cancer Res. 2012; 72: 3997^007. doi: 10.1158/0008-5472.CAN-12-1377.

39. Nielsen J.S., Sahota R.A., Milne K., Kost S.E., et al. CD20+ tumor-infiltrating lymphocytes have an atypical CD27- memory phenotype and together with CD8+ T cells promote favorable prognosis in ovarian cancer. Clin. Cancer Res. 2012; 18: 3281-92. doi: 10.1158/1078-0432.CCR-12-0234.

40. Kroeger D.R., Milne K., Nelson B.H. 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: 3005-15. doi: 10.1158/1078-0432.CCR-15-2762.

41. Hiraoka N., Ino Y., Yamazaki-Itoh R., Kanai Y., et al. Intratumoral tertiary lymphoid organ is a favourable prognosticator in patients with pancreatic cancer. Br. J. Cancer. 2015; 112: 1782-90. doi: 10.1038/bjc.2015.145.

42. Finkin S., Yuan D., Stein I., Taniguchi K., et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat. Immunol. 2015; 16: 1235-44. doi: 10.1038/ni.3290.

43. Sakai Y., Hoshino H., Kitazawa R., Kobayashi M. High endothelial venule-like vessels and lymphocyte recruitment in testicular seminoma. Andrology. 2014; 2: 282-9. doi: 10.1111/j.2047-2927.2014.00192.x.

44. Giraldo N.A., Becht E., Pages F., Skliris G., et al. Orchestration and prognostic significance of immune checkpoints in the microenvironment of primary and metastatic renal cell cancer. Clin. Cancer Res. 2015; 21: 3031^0. doi: 10.1158/1078-0432.CCR-14-2926.

45. Low S., Sakai Y., Hoshino H., Hirokawa M., et al. High endothelial venule-like vessels and lymphocyte recruitment in diffuse sclerosing variant of papillary thyroid carcinoma. Pathology. Doi: 10.1016/j.pathol.2016.08.002.

46. Gants ev S.K. New technologies of diagnosis and treatment of breast cancer. Creativnaya khirurgiya i onkologiya. 2009; 1: 6-9. (in Russian)

47. Gantsev S.K., Umezawa K., Islamgulov D.V., et al. The role of inflammatory chemokines in lymphoid neoorganogenesis in breast cancer. Biomed. Pharmacother. 2013; 67 (5): 363-6.

48. Gantsev S.K. Axillary lymphatic apparatus in breast cancer: Atlas. Moscow: MIA, 2011: 194 p. (in Russian)

49. Gantsev S.K. New to the theory of cancer metastasis and approaches to its treatment. Creativnaya khirurgiya i onkologiya. 2010; 4: 5-11. (in Russian)

50. Borodin Yu.I., Gorchakova O.V., Gorchakov V.N. Peripheral lymphoid structures: formation and function. Morfologiya. 2016; 4: 90-6. (in Russian)

51. Dieu-Nosjean M.C., Giraldo N.A., Kaplon H., Germain C., et al. Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol. Rev. 2016; 271 (1): 260-75.

All articles in our journal are distributed under the Creative Commons Attribution 4.0 International License (CC BY 4.0 license)


JOURNALS of «GEOTAR-Media»