AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (437.1 KB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review | Open Access

Insights into tertiary lymphoid structures in the solid tumor microenvironment: anti-tumor mechanism, functional regulation, and immunotherapeutic strategies

Hua Zhao1Hao Wang1Qiuru Zhou1Xiubao Ren1,2 ( )
Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China
Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China
Show Author Information

Abstract

Tertiary lymphoid structures (TLSs) are ectopic immune cell aggregations that develop in peripheral tissues in response to a wide range of chronic inflammatory conditions, including infection, autoimmune disease, and cancer. In the tumor microenvironment (TME), the structures of TLSs, including B-cell- and T-cell-enriched areas indicate that the TLSs might be the local site during the initiation and maintenance of humoral and cellular immune responses against cancers. Numerous studies have evaluated the expression of TLSs in different cancer patients and their association with prognoses of cancer patients. It was shown that well-developed TLSs characterized by mature B cells synthesized tumor specific antibodies, which were considered as specific markers for a good prognosis. However, there are still some immunosuppressive factors existing in the TLSs that may affect anti-tumor responses. These factors include dysfunctional B cells, regulatory T cells, and T follicular regulatory cells. The complexity and heterogeneity of the TLS composition may affect the function and activity of TLSs; it is therefore essential to fully understand the function and influencing factors in TLSs. It has been reported that checkpoint inhibitors and vaccines are currently being developed to reprogram the TME by establishing mature TLSs to improve cancer immunotherapies. In this review, we focused on recent advances in TLSs in human solid tumors, including structural characteristics and classes, antitumor mechanisms, immunosuppressive factors, and TLS-based therapeutic approaches.

References

1

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

2

Arneth B. Tumor microenvironment. Medicina (Kaunas). 2020; 56: 15.

3

Bruni D, Angell HK, Galon J. The immune contexture and immunoscore in cancer prognosis and therapeutic efficacy. Nat Rev Cancer. 2020; 20(11): 662-80.

4

Szeto GL, Finley SD. Integrative approaches to cancer immunotherapy. Trends Cancer. 2019; 5: 400-10.

5

Sautès-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019; 19(6): 307-25.

6

Ruddle NH. Basics of inducible lymphoid organs. Curr Top Microbiol Immunol. 2020; 426: 1-19.

7

Pipi E, Nayar S, Gardner DH, Colafrancesco S, Smith C, Barone F. Tertiary lymphoid structures: autoimmunity goes local. Front Immunol. 2018; 9: 1952.

8

Leur K, Clahsen-van Groningen MC, Bosch TPP, Graav GN, Hesselink DA, Samsom JN, et al. Characterization of ectopic lymphoid structures in different types of acute renal allograft rejection. Clin Exp Immunol. 2018; 192(2): 224-32.

9

Martinet L, Garrido I, Filleron T, Guellec SL, Bellard E, Fournie JJ, 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.

10

Cabrita R, Lauss M, Sanna A, Donia M, Larsen MS, Mitra S, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020; 577: 561-5.

11

Petitprez F, Reyniès A, Keung EZ, Wu Chen TW, Sun CM, Calderaro J, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020; 577: 556-60.

12

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: 549-55.

13

Steele KE, Brown C. Multiplex immunohistochemistry for image analysis of tertiary lymphoid structures in cancer. Methods Mol Biol. 2018; 1845: 87-98.

14

Tang H, Qiu X, Timmerman C, Fu YX. Targeting tertiary lymphoid structures for tumor immunotherapy. Methods Mol Biol. 2018; 1845: 275-86.

15

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: 4410-7.

16

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: 3005-15.

17

Gu-Trantien C, Migliori E, Buisseret L, Wind A, Brohée S, Garaud S, et al. CXCL13-producing TFH cells link immune suppression and adaptive memory in human breast cancer. JCI Insight. 2017; 2: e91487.

18

Li K, Guo Q, Zhang X, Dong X, Liu W, Zhang A, et al. Oral cancer-associated tertiary lymphoid structures: gene expression profile and prognostic value. Clin Exp Immunol. 2020; 199: 172-81.

19

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: 1733-45.

20

Rodriguez AB, Engelhard VH. Insights into tumor-associated tertiary lymphoid structures: novel targets for antitumor immunity and cancer immunotherapy. Cancer Immunol Res. 2020; 8: 1338-45.

21

Calderaro J, Petitprez F, Becht E, Laurent A, Hirsch TZ, Rousseau B, et al. Intra-tumoral tertiary lymphoid structures are associated with a low risk of early recurrence of hepatocellular carcinoma. J Hepatol. 2019; 70: 58-65.

22

Munoz-Erazo L, Rhodes JL, Marion VC, Kemp RA. Tertiary lymphoid structures in cancer-considerations for patient prognosis. Cell Mol Immunol. 2020; 17: 570-5.

23

Germain C, Gnjatic S, Tamzalit F, Knockaert S, Remark R, Goc J, 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.

24

Posch F, Silina K, Leibl S, Mündlein A, Moch H, Siebenhüner A, et al. Maturation of tertiary lymphoid structures and recurrence of stage Ⅱ and Ⅲ colorectal cancer. Oncoimmunology. 2017; 7: e1378844.

25

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: 1308-20.

26

Vinuesa CG, Linterman MA, Yu D, MacLennan IC. Follicular helper T cells. Annu Rev Immunol. 2016; 34: 335-68.

27

Crotty S. T Follicular helper cell biology: a decade of discovery and diseases. Immunity. 2019; 50: 1132-48.

28

Sharonov GV, Serebrovskaya EO, Yuzhakova DV, Britanova OV, Chudakov DM. B cells, plasma cells and antibody repertoires in the tumour microenvironment. Nat Rev Immunol. 2020; 20: 294-307.

29

Katoh H, Komura D, Konishi H, Suzuki R, Yamamoto A, Kakiuchi M, et al. Immunogenetic profiling for gastric cancers identifies sulfated glycosaminoglycans as major and functional B cell antigens in human malignancies. Cell Rep. 2017; 20: 1073-87.

30

Koscsó B, Kurapati S, Rodrigues RR, Nedjic J, Gowda K, Shin C, et al. Gut-resident CX3CR1 hi macrophages induce tertiary lymphoid structures and IgA response in situ. Sci Immunol. 2020; 5: eaax0062.

31

Schlößer HA, Thelen M, Lechner A, Wennhold K, Garcia-Marquez MA, Rothschild SI, et al. B cells in esophago-gastric adenocarcinoma are highly differentiated, organize in tertiary lymphoid structures and produce tumor-specific antibodies. Oncoimmunology. 2018; 8: e1512458.

32

Montfort A, Pearce O, Maniati E, Vincent BG, Bixby L, Böhm S, et al. A strong B-cell response is part of the immune landscape in human high-grade serous ovarian metastases. Clin Cancer Res. 2017; 23: 250-62.

33

Wang X, Mathieu M, Brezski RJ. IgG Fc engineering to modulate antibody effector functions. Protein Cell. 2018; 9: 63-73.

34

Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front Immunol. 2014; 5: 520.

35

Mizukami M, Hanagiri T, Yasuda M, Kuroda K, Shigematsu Y, Baba T, et al. Antitumor effect of antibody against a SEREX-defined antigen (UOEH-LC-1) on lung cancer xenotransplanted into severe combined immunodeficiency mice. Cancer Res. 2007; 67: 8351-7.

36

Lechner A, Schlößer HA, Thelen M, Wennhold K, Rothschild SI, Gilles R, et al. Tumor-associated B cells and humoral immune response in head and neck squamous cell carcinoma. Oncoimmunology. 2019; 8: 1535293.

37

Wang TT, Ravetch JV. Functional diversification of IgGs through Fc glycosylation. J Clin Invest. 2019; 129: 3492-8.

38

Bindon CI, Hale G, Brüggemann M, Waldmann H. Human monoclonal IgG isotypes differ in complement activating function at the level of C4 as well as C1q. J Exp Med. 1988; 168: 127-42.

39

Shi JY, Gao Q, Wang ZC, Zhou J, Wang XY, Min ZH, et al. Margin-infiltrating CD20(+) B cells display an atypical memory phenotype and correlate with favorable prognosis in hepatocellular carcinoma. Clin Cancer Res. 2013; 19: 5994-6005.

40

Rao DA. T cells that help B cells in chronically inflamed tissues. Front Immunol. 2018; 9: 1924.

41

Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, Wind A, et al. CD4+ follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest. 2013; 123: 2873-92.

42

Hatzi K, Nance JP, Kroenke MA, Bothwell M, Haddad EK, Melnick A, et al. BCL6 orchestrates Tfh cell differentiation via multiple distinct mechanisms. J Exp Med. 2015; 212: 539-53.

43

Shi J, Hou S, Fang Q, Liu X, Liu X, Qi H. PD-1 controls follicular T helper cell positioning and function. Immunity. 2018; 49: 264-74.e4.

44

Hutloff A. T follicular helper-like cells in inflamed non-lymphoid tissues. Front Immunol. 2018; 9: 1707.

45

Kawamoto S, Tran TH, Maruya M, Suzuki K, DoiY, Tsutsui Y, et al. The inhibitory receptor PD-1 regulates IgA selection and bacterial composition in the gut. Science. 2012; 336: 485-9.

46

Behr DS, Peitsch WK, Hametner C, Lasitschka F, Houben R, Schönhaar K, et al. Prognostic value of immune cell infiltration, tertiary lymphoid structures and PD-L1 expression in merkel cell carcinomas. Int J Clin Exp Pathol. 2014; 7: 7610-21.

47

Goc J, Germain C, Vo-Bourgais TKD, 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: 705-15.

48

Li Q, Zhang D, He W, Chen T, Yan Z, Gao X, et al. CD8+ T cells located in tertiary lymphoid structures are associated with improved prognosis in patients with gastric cancer. Oncol Lett. 2020; 20: 2655-64.

49

Truxova I, Kasikova L, Hensler M, Skapa P, Laco J, Pecen L, et al. Mature dendritic cells correlate with favorable immune infiltrate and improved prognosis in ovarian carcinoma patients. J Immunother Cancer. 2018; 6: 139.

50

Thomas R, Al-Khadairi G, Roelands J, Hendrickx W, Dermime S, Bedognetti D, et al. NY-ESO-1 based immunotherapy of cancer: current perspectives. Front Immunol. 2018; 9: 947.

51

Gnjatic S, Atanackovic D, Jäger E, Matsuo M, Selvakumar A, Altorki NK, et al. Survey of naturally occurring CD4+ T cell responses against NY-ESO-1 in cancer patients: correlation with antibody responses. Proc Natl Acad Sci USA. 2003; 100: 8862-7.

52

Shedlock DJ, Shen H. Requirement for CD4 T cell help in generating functional CD8 T cell memory. Science. 2003; 300: 337-9.

53

Hennequin A, Derangère V, Boidot R, Apetoh L, Vincent J, Orry D, et al. Tumor infiltration by Tbet+ effector T cells and CD20+ B cells is associated with survival in gastric cancer patients. Oncoimmunology. 2015; 5: e1054598.

54

Bruno TC, Ebner PJ, Moore BL, Squalls OG, Waugh KA, Eruslanov EB, et al. Antigen-presenting intratumoral B cells affect CD4+ TIL phenotypes in non-small cell lung cancer patients. Cancer Immunol Res. 2017; 5: 898-907.

55

Yamakoshi Y, Tanaka H, Sakimura C, Deguchi S, Mori T, Tamura T, et al. Immunological potential of tertiary lymphoid structures surrounding the primary tumor in gastric cancer. Int J Oncol. 2020; 57: 171-82.

56

Garaud S, Zayakin P, Buisseret L, Rulle U, Silina K, Wind A, et al. Antigen specificity and clinical significance of IgG and IgA autoantibodies produced in situ by Tumor-Infiltrating B cells in breast cancer. Front Immunol. 2018; 9: 2660.

57

Isaeva OI, Sharonov GV, Serebrovskaya EO, Turchaninova MA, Zaretsky AR, Shugay M, et al. Intratumoral immunoglobulin isotypes predict survival in lung adenocarcinoma subtypes. J Immunother Cancer. 2019; 7: 279.

58

Shalapour S, Font-Burgada J, Caro GD, Zhong Z, Sanchez-Lopez E, Dhar D, et al. Immunosuppressive plasma cells impede T cell-dependent immunogenic chemotherapy. Nature. 2015; 521: 94-8.

59

Schietinger A, Greenberg PD. Tolerance and exhaustion: defining mechanisms of T cell dysfunction. Trends Immunol. 2014; 35: 51-60.

60

Shah N. Diagnostic significance of levels of immunoglobulin A in seminal fluid of patients with prostatic disease. Urology. 1976; 8: 270-2.

61

Chen L, Oke T, Siegel N, Cojocaru G, Tam AJ, Blosser RL, et al. The immunosuppressive niche of soft-tissue sarcomas is sustained by tumor-associated macrophages and characterized by intratumoral tertiary lymphoid structures. Clin Cancer Res. 2020; 26: 4018-30.

62

García-Hernández ML, Uribe-Uribe NO, Espinosa-González R, Kast WM, Khader SA, Rangel-Moreno J. A unique cellular and molecular microenvironment is present in tertiary lymphoid organs of patients with spontaneous prostate cancer regression. Front Immunol. 2017; 8: 563.

63

Schweiger T, Berghoff AS, Glogner C, Glueck O, Rajky O, Traxler D, et al. Tumor-infiltrating lymphocyte subsets and tertiary lymphoid structures in pulmonary metastases from colorectal cancer. Clin Exp Metastasis. 2016; 33: 727-39.

64

Hiraoka N, Ino Y, Yamazaki-Itoh R, Kanai Y, Kosuge T, Shimada K. Intratumoral tertiary lymphoid organ is a favourable prognosticator in patients with pancreatic cancer. Br J Cancer. 2015; 112: 1782-90.

65

Joshi NS, Akama-Garren EH, Lu Y, Lee DY, Chang GP, Li A, et al. Regulatory T cells in tumor-associated tertiary lymphoid structures suppress anti-tumor T cell responses. Immunity. 2015; 43: 579-90.

66

Yamaguchi K, Ito M, Ohmura H, Hanamura F, Nakano M, Tsuchihashi K, et al. Helper T cell-dominant tertiary lymphoid structures are associated with disease relapse of advanced colorectal cancer. Oncoimmunology. 2020; 9: 1724763.

67

Li L, Ma Y, Xu Y, Maerkeya K. TIM-3 expression identifies a distinctive PD-1+ follicular helper T cell subset, with reduced interleukin 21 production and B cell help function in ovarian cancer patients. Int Immunopharmacol. 2018; 57: 139-46.

68

Zhang M, Wu Y, Bastian D, Iamsawat S, Chang J, Daenthanasanmak A, et al. Inducible T-cell co-stimulator impacts chronic graft-versus-host disease by regulating both pathogenic and regulatory T Cells. Front Immunol. 2018; 9: 1461.

69

Song H, Liu A, Liu G, Wu F, Li Z. T follicular regulatory cells suppress Tfh-mediated B cell help and synergistically increase IL-10-producing B cells in breast carcinoma. Immunol Res. 2019; 67: 416-23.

70

Li L, Ma Y, Xu Y. Follicular regulatory T cells infiltrated the ovarian carcinoma and resulted in CD8 T cell dysfunction dependent on IL-10 pathway. Int Immunopharmacol. 2019; 68: 81-7.

71

Cottrell TR, Thompson ED, Forde PM, Stein JE, Duffield AS, Anagnostou V, et al. Pathologic features of response to neoadjuvant anti-PD-1 in resected non-small-cell lung carcinoma: a proposal for quantitative immune-related pathologic response criteria (irPRC). Ann Oncol. 2018; 29: 1853-60.

72

Morcrette G, Hirsch TZ, Badour E, Pilet J, Caruso S, Calderaro J, et al. APC germline hepatoblastomas demonstrate cisplatin-induced intratumor tertiary lymphoid structures. Oncoimmunology. 2019; 8: e1583547.

73

Shimizu K, Yamasaki S, Shinga J, Sato Y, Watanabe T, Ohara O, et al. Systemic DC activation modulates the tumor microenvironment and shapes the long-lived tumor-specific memory mediated by CD8+ T Cells. Cancer Res. 2016; 76: 3756-66.

74

Huang Y, Chen Y, Zhou S, Chen L, Wang J, Pei Y, et al. Dual-mechanism based CTLs infiltration enhancement initiated by Nano-sapper potentiates immunotherapy against immune-excluded tumors. Nat Commun. 2020; 11: 622.

75

Johansson-Percival A, He B, Li ZJ, Kjellén A, Russell K, Li J, et al. De novo induction of intratumoral lymphoid structures and vessel normalization enhances immunotherapy in resistant tumors. Nat Immunol. 2017; 18: 1207-17.

76

Chelvanambi M, Fecek RJ, Taylor JL, Storkus WJ. STING agonist-based treatment promotes vascular normalization and tertiary lymphoid structure formation in the therapeutic melanoma microenvironment. J Immunother Cancer. 2021; 9: e001906.

77

Lutz ER, Wu AA, Bigelow E, Sharma R, Mo G, Soares K, et al. Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol Res. 2014; 2: 616-31.

78

Pollack KE, Meneveau MO, Melssen MM, Lynch KT, Koeppel AF, Young SJ, et al. Incomplete Freund’s adjuvant reduces arginase and enhances Th1 dominance, TLR signaling and CD40 ligand expression in the vaccine site microenvironment. J Immunother Cancer. 2020; 8: e000544.

79

Maldonado L, Teague JE, Morrow MP, Jotova I, Wu TC, Wang C, et al. Intramuscular therapeutic vaccination targeting HPV16 induces T cell responses that localize in mucosal lesions. Sci Transl Med. 2014; 6: 221ra13.

80

Jones E, Gallimore A, Ager A. Defining high endothelial venules and tertiary lymphoid structures in cancer. Methods Mol Biol. 2018; 1845: 99-118.

81

Song IH, Heo SH, Bang WS, Park HS, Park IA, Kim YA, 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.

82

Allen E, Jabouille A, Rivera LB, Lodewijckx I, Missiaen R, Steri V, et al. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med. 2017; 9: eaak9679.

Cancer Biology & Medicine
Pages 981-991
Cite this article:
Zhao H, Wang H, Zhou Q, et al. Insights into tertiary lymphoid structures in the solid tumor microenvironment: anti-tumor mechanism, functional regulation, and immunotherapeutic strategies. Cancer Biology & Medicine, 2021, 18(4): 981-991. https://doi.org/10.20892/j.issn.2095-3941.2021.0029

66

Views

0

Downloads

15

Crossref

17

Web of Science

16

Scopus

Altmetrics

Received: 12 January 2021
Accepted: 08 June 2021
Published: 01 November 2021
©2021 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return