Skip to main content
Top
Published in: BMC Cancer 1/2023

Open Access 01-12-2023 | Pancreatic Cancer | Research

Prognostic value of tertiary lymphoid structures (TLS) in digestive system cancers: a systematic review and meta-analysis

Authors: Hao Sun, Yuanyu Shi, Hailiang Ran, Junwei Peng, Qiongxian Li, Guiqing Zheng, Yandie He, Shuqing Liu, Wei Chang, Yuanyuan Xiao

Published in: BMC Cancer | Issue 1/2023

Login to get access

Abstract

Background

Existing literature suggests that tertiary lymphatic structure (TLS) is associated with the progression of cancer. However, the prognostic role of TLS in digestive system cancers remains controversial. This meta-analysis aims to synthesize currently available evidence in the association between TLS and the survival of digestive system cancers.

Methods

We systematically searched three digital databases (PubMed, Embase, Web of Science) for articles published from database inception to December 23, 2022. Study selection criteria are based on PECO framework: P (population: patients with digestive system cancers), E (exposure: presence of TLS), C (comparator: absence of TLS), O (outcome: overall survival, OS; recurrence-free survival, RFS; disease-free survival, DFS). The Quality in Prognostic Studies (QUIPS) tool was used to assess risk of bias for included studies. The study protocol was registered with PROSPERO (CRD42023416307).

Results

A total of 25 studies with 6910 patients were included into the final meta-analysis. Random-effects models revealed that the absence of TLS was associated with compromised OS, RFS, and DFS of digestive system cancers, with pooled hazard ratios (HRs) of 1.74 (95% CI: 1.50–2.03), 1.96 (95% CI: 1.58–2.44), and 1.81 (95% CI: 1.49–2.19), respectively. Subgroup analyses disclosed a stronger TLS-survival association for pancreatic cancer, compared with other digestive system cancers.

Conclusion

TLS may be of prognostic significance for digestive system cancers. More original studies are needed to further corroborate this finding.
Appendix
Available only for authorised users
Literature
1.
go back to reference Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef
2.
go back to reference Rumgay H, Arnold M, Ferlay J, Lesi O, Cabasag CJ, Vignat J, et al. Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol. 2022;77(6):1598–606.PubMedPubMedCentralCrossRef Rumgay H, Arnold M, Ferlay J, Lesi O, Cabasag CJ, Vignat J, et al. Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol. 2022;77(6):1598–606.PubMedPubMedCentralCrossRef
3.
go back to reference Morgan E, Soerjomataram I, Rumgay H, Coleman HG, Thrift AP, Vignat J, et al. The Global Landscape of Esophageal Squamous Cell Carcinoma and Esophageal Adenocarcinoma Incidence and Mortality in 2020 and Projections to 2040: New Estimates From GLOBOCAN 2020. Gastroenterology. 2022;163(3):649–58.PubMedCrossRef Morgan E, Soerjomataram I, Rumgay H, Coleman HG, Thrift AP, Vignat J, et al. The Global Landscape of Esophageal Squamous Cell Carcinoma and Esophageal Adenocarcinoma Incidence and Mortality in 2020 and Projections to 2040: New Estimates From GLOBOCAN 2020. Gastroenterology. 2022;163(3):649–58.PubMedCrossRef
4.
go back to reference Morgan E, Arnold M, Gini A, Lorenzoni V, Cabasag CJ, Laversanne M, et al. Global burden of colorectal cancer in 2020 and 2040: incidence and mortality estimates from GLOBOCAN. Gut. 2023;72(2):338–44.PubMedCrossRef Morgan E, Arnold M, Gini A, Lorenzoni V, Cabasag CJ, Laversanne M, et al. Global burden of colorectal cancer in 2020 and 2040: incidence and mortality estimates from GLOBOCAN. Gut. 2023;72(2):338–44.PubMedCrossRef
8.
go back to reference Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science. 2022;375(6576):eabf9419.PubMedCrossRef Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science. 2022;375(6576):eabf9419.PubMedCrossRef
9.
go back to reference Drayton DL, Liao S, Mounzer RH, Ruddle NH. Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol. 2006;7(4):344–53.PubMedCrossRef Drayton DL, Liao S, Mounzer RH, Ruddle NH. Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol. 2006;7(4):344–53.PubMedCrossRef
10.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
11.
go back to reference Picker LJ, Butcher EC. Physiological and molecular mechanisms of lymphocyte homing. Annu Rev Immunol. 1992;10:561–91.PubMedCrossRef Picker LJ, Butcher EC. Physiological and molecular mechanisms of lymphocyte homing. Annu Rev Immunol. 1992;10:561–91.PubMedCrossRef
12.
go back to reference Neyt K, Perros F, GeurtsvanKessel CH, Hammad H, Lambrecht BN. Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol. 2012;33(6):297–305.PubMedPubMedCentralCrossRef Neyt K, Perros F, GeurtsvanKessel CH, Hammad H, Lambrecht BN. Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol. 2012;33(6):297–305.PubMedPubMedCentralCrossRef
13.
go back to reference Aloisi F, Pujol-Borrell R. Lymphoid neogenesis in chronic inflammatory diseases. Nat Rev Immunol. 2006;6(3):205–17.PubMedCrossRef Aloisi F, Pujol-Borrell R. Lymphoid neogenesis in chronic inflammatory diseases. Nat Rev Immunol. 2006;6(3):205–17.PubMedCrossRef
14.
go back to reference 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(3):579–90.PubMedPubMedCentralCrossRef 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(3):579–90.PubMedPubMedCentralCrossRef
15.
go back to reference Chaurio RA, Anadon CM, Lee Costich T, Payne KK, Biswas S, Harro CM, et al. TGF-β-mediated silencing of genomic organizer SATB1 promotes Tfh cell differentiation and formation of intra-tumoral tertiary lymphoid structures. Immunity. 2022;55(1):115–28.PubMedPubMedCentralCrossRef Chaurio RA, Anadon CM, Lee Costich T, Payne KK, Biswas S, Harro CM, et al. TGF-β-mediated silencing of genomic organizer SATB1 promotes Tfh cell differentiation and formation of intra-tumoral tertiary lymphoid structures. Immunity. 2022;55(1):115–28.PubMedPubMedCentralCrossRef
16.
go back to reference Messina JL, Fenstermacher DA, Eschrich S, Qu X, Berglund AE, Lloyd MC, et al. 12-Chemokine Gene Signature Identifies Lymph Node-like Structures in Melanoma: Potential for Patient Selection for Immunotherapy? Sci Rep. 2012;2:765.PubMedPubMedCentralCrossRef Messina JL, Fenstermacher DA, Eschrich S, Qu X, Berglund AE, Lloyd MC, et al. 12-Chemokine Gene Signature Identifies Lymph Node-like Structures in Melanoma: Potential for Patient Selection for Immunotherapy? Sci Rep. 2012;2:765.PubMedPubMedCentralCrossRef
17.
go back to reference Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, de Wind A, et al. CD4+ follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest. 2013;123(7):2873–92.PubMedPubMedCentralCrossRef Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, de Wind A, et al. CD4+ follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest. 2013;123(7):2873–92.PubMedPubMedCentralCrossRef
18.
go back to reference 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):705–15.PubMedCrossRef 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):705–15.PubMedCrossRef
19.
go back to reference Bento DC, Jones E, Junaid S, Tull J, Williams GT, Godkin A, et al. High endothelial venules are rare in colorectal cancers but accumulate in extra-tumoral areas with disease progression. OncoImmunology. 2015;4(3): e974374.PubMedPubMedCentralCrossRef Bento DC, Jones E, Junaid S, Tull J, Williams GT, Godkin A, et al. High endothelial venules are rare in colorectal cancers but accumulate in extra-tumoral areas with disease progression. OncoImmunology. 2015;4(3): e974374.PubMedPubMedCentralCrossRef
20.
go back to reference 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. 2018;7(2): e1378844.PubMedCrossRef 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. 2018;7(2): e1378844.PubMedCrossRef
21.
go back to reference Finkin S, Yuan D, Stein I, Taniguchi K, Weber A, Unger K, et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat Immunol. 2015;16(12):1235–44.PubMedPubMedCentralCrossRef Finkin S, Yuan D, Stein I, Taniguchi K, Weber A, Unger K, et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat Immunol. 2015;16(12):1235–44.PubMedPubMedCentralCrossRef
22.
go back to reference 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(1):58–65.PubMedCrossRef 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(1):58–65.PubMedCrossRef
23.
go back to reference Page MJ, McKenzie JE, Bossuyt PM, Bossuyt PM, Boutron I, Hoffmann TC, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;134:178–89. Page MJ, McKenzie JE, Bossuyt PM, Bossuyt PM, Boutron I, Hoffmann TC, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;134:178–89.
24.
go back to reference Morgan RL, Whaley P, Thayer KA, Schünemann HJ. Identifying the PECO: A framework for formulating good questions to explore the association of environmental and other exposures with health outcomes. Environ Int. 2018;121(Pt 1):1027–31.PubMedPubMedCentralCrossRef Morgan RL, Whaley P, Thayer KA, Schünemann HJ. Identifying the PECO: A framework for formulating good questions to explore the association of environmental and other exposures with health outcomes. Environ Int. 2018;121(Pt 1):1027–31.PubMedPubMedCentralCrossRef
25.
go back to reference Hayden JA, van der Windt DA, Cartwright JL, Côté P, Bombardier C. Assessing bias in studies of prognostic factors. Ann Intern Med. 2013;158(4):280–6.PubMedCrossRef Hayden JA, van der Windt DA, Cartwright JL, Côté P, Bombardier C. Assessing bias in studies of prognostic factors. Ann Intern Med. 2013;158(4):280–6.PubMedCrossRef
28.
go back to reference Begg CB, Mazumdar M. Operating characteristics of a rank correlation test for publication bias. Biometrics. 1994;50:1088–101.PubMedCrossRef Begg CB, Mazumdar M. Operating characteristics of a rank correlation test for publication bias. Biometrics. 1994;50:1088–101.PubMedCrossRef
29.
go back to reference Deguchi S, Tanaka H, Suzuki S, Natsuki S, Mori T, Miki Y, et al. Clinical relevance of tertiary lymphoid structures in esophageal squamous cell carcinoma. BMC Cancer. 2022;22:699.PubMedPubMedCentralCrossRef Deguchi S, Tanaka H, Suzuki S, Natsuki S, Mori T, Miki Y, et al. Clinical relevance of tertiary lymphoid structures in esophageal squamous cell carcinoma. BMC Cancer. 2022;22:699.PubMedPubMedCentralCrossRef
30.
go back to reference Li RT, Huang X, Yang W, Wang J, Liang Y, Zhang T, et al. Tertiary lymphoid structures favor outcome in resected esophageal squamous cell carcinoma. J Pathol Clin Res. 2022;8(5):422–35.PubMedPubMedCentralCrossRef Li RT, Huang X, Yang W, Wang J, Liang Y, Zhang T, et al. Tertiary lymphoid structures favor outcome in resected esophageal squamous cell carcinoma. J Pathol Clin Res. 2022;8(5):422–35.PubMedPubMedCentralCrossRef
31.
go back to reference Ling YH, Zhong J, Weng Z, Lin G, Liu C, Pan C, et al. The prognostic value and molecular properties of tertiary lymphoid structures in oesophageal squamous cell carcinoma. Clin Transl Med. 2022;12(10):e1074.PubMedPubMedCentralCrossRef Ling YH, Zhong J, Weng Z, Lin G, Liu C, Pan C, et al. The prognostic value and molecular properties of tertiary lymphoid structures in oesophageal squamous cell carcinoma. Clin Transl Med. 2022;12(10):e1074.PubMedPubMedCentralCrossRef
32.
go back to reference Jiang Q, Tian C, Wu H, Min L, Chen H, Chen L, et al. Tertiary lymphoid structure patterns predicted anti-PD1 therapeutic responses in gastric cancer. Chin J Cancer Res. 2022;34(4):365–82.PubMedPubMedCentralCrossRef Jiang Q, Tian C, Wu H, Min L, Chen H, Chen L, et al. Tertiary lymphoid structure patterns predicted anti-PD1 therapeutic responses in gastric cancer. Chin J Cancer Res. 2022;34(4):365–82.PubMedPubMedCentralCrossRef
33.
go back to reference Kemi N, Ylitalo O, Väyrynen JP, Helminen O, Junttila A, Mrena J, Böhm J, Kauppila JH. Tertiary lymphoid structures and gastric cancer prognosis. APMIS. 2023;131(1):19–25.PubMedCrossRef Kemi N, Ylitalo O, Väyrynen JP, Helminen O, Junttila A, Mrena J, Böhm J, Kauppila JH. Tertiary lymphoid structures and gastric cancer prognosis. APMIS. 2023;131(1):19–25.PubMedCrossRef
34.
go back to reference Zhan Z, Shi-Jin L, Yi-Ran Z, Zhi-Long L, Xiao-Xu Z, Hui D, et al. High endothelial venules proportion in tertiary lymphoid structure is a prognostic marker and correlated with anti-tumor immune microenvironment in colorectal cancer. Ann Med. 2023;55(1):114–26.PubMedCrossRef Zhan Z, Shi-Jin L, Yi-Ran Z, Zhi-Long L, Xiao-Xu Z, Hui D, et al. High endothelial venules proportion in tertiary lymphoid structure is a prognostic marker and correlated with anti-tumor immune microenvironment in colorectal cancer. Ann Med. 2023;55(1):114–26.PubMedCrossRef
35.
go back to reference Li JH, Nie Y, Jia W, Wu W, Song W, Li Y. Effect of Tertiary Lymphoid Structures on Prognosis of Patients with Hepatocellular Carcinoma and Preliminary Exploration of Its Formation Mechanism. Cancers (Basel). 2022;14(20):5157.PubMedCrossRef Li JH, Nie Y, Jia W, Wu W, Song W, Li Y. Effect of Tertiary Lymphoid Structures on Prognosis of Patients with Hepatocellular Carcinoma and Preliminary Exploration of Its Formation Mechanism. Cancers (Basel). 2022;14(20):5157.PubMedCrossRef
36.
go back to reference Nie Y, Fan H, Li J, Lei X, Zhang T, Wang Y, et al. Tertiary lymphoid structures: Associated multiple immune cells and analysis their formation in hepatocellular carcinoma. FASEB J. 2022;36(11):e22586.PubMedCrossRef Nie Y, Fan H, Li J, Lei X, Zhang T, Wang Y, et al. Tertiary lymphoid structures: Associated multiple immune cells and analysis their formation in hepatocellular carcinoma. FASEB J. 2022;36(11):e22586.PubMedCrossRef
37.
go back to reference Wen SD, Chen Y, Hu C, Du X, Xia J, Wang X, et al. Combination of Tertiary Lymphoid Structure and Neutrophil-to-Lymphocyte Ratio Predicts Survival in Patients With Hepatocellular Carcinoma. Front Immunol. 2021;12:788640.PubMedCrossRef Wen SD, Chen Y, Hu C, Du X, Xia J, Wang X, et al. Combination of Tertiary Lymphoid Structure and Neutrophil-to-Lymphocyte Ratio Predicts Survival in Patients With Hepatocellular Carcinoma. Front Immunol. 2021;12:788640.PubMedCrossRef
38.
go back to reference Yu JS, Huang WB, Zhang YH, Chen J, Li J, Fu HF, et al. The association of immune cell infiltration and prognostic value of tertiary lymphoid structures in gastric cancer. Neoplasma. 2022;69(4):886–98.PubMedCrossRef Yu JS, Huang WB, Zhang YH, Chen J, Li J, Fu HF, et al. The association of immune cell infiltration and prognostic value of tertiary lymphoid structures in gastric cancer. Neoplasma. 2022;69(4):886–98.PubMedCrossRef
39.
go back to reference Cheng N, Li P, Cheng H, Zhao X, Dong M, Zhang Y, et al. Prognostic Value of Tumor-Infiltrating Lymphocytes and Tertiary Lymphoid Structures in Epstein-Barr Virus-Associated and -Negative Gastric Carcinoma. Front Immunol. 2021;12:692859.PubMedPubMedCentralCrossRef Cheng N, Li P, Cheng H, Zhao X, Dong M, Zhang Y, et al. Prognostic Value of Tumor-Infiltrating Lymphocytes and Tertiary Lymphoid Structures in Epstein-Barr Virus-Associated and -Negative Gastric Carcinoma. Front Immunol. 2021;12:692859.PubMedPubMedCentralCrossRef
40.
go back to reference Yamakoshi Y, Tanaka H, Sakimura C, Mori T, Deguchi S, Yoshii M, et al. Association between the preoperative neutrophil-to-lymphocyte ratio and tertiary lymphoid structures surrounding tumor in gastric cancer. Mol Clin Oncol. 2021;14(4):76.PubMedPubMedCentralCrossRef Yamakoshi Y, Tanaka H, Sakimura C, Mori T, Deguchi S, Yoshii M, et al. Association between the preoperative neutrophil-to-lymphocyte ratio and tertiary lymphoid structures surrounding tumor in gastric cancer. Mol Clin Oncol. 2021;14(4):76.PubMedPubMedCentralCrossRef
41.
go back to reference Gunderson AJ, Rajamanickam V, Bui C, Bernard B, Pucilowska J, Ballesteros-Merino C, et al. Germinal center reactions in tertiary lymphoid structures associate with neoantigen burden, humoral immunity and long-term survivorship in pancreatic cancer. Oncoimmunology. 2021;10(1):1900635. Gunderson AJ, Rajamanickam V, Bui C, Bernard B, Pucilowska J, Ballesteros-Merino C, et al. Germinal center reactions in tertiary lymphoid structures associate with neoantigen burden, humoral immunity and long-term survivorship in pancreatic cancer. Oncoimmunology. 2021;10(1):1900635.
42.
go back to reference Mori T, Tanaka H, Deguchi S, Yamakoshi Y, Miki Y, Yoshii M, et al. Clinical efficacy of nivolumab is associated with tertiary lymphoid structures in surgically resected primary tumors of recurrent gastric cancer. PLoS ONE. 2022;17(1):e0262455.PubMedPubMedCentralCrossRef Mori T, Tanaka H, Deguchi S, Yamakoshi Y, Miki Y, Yoshii M, et al. Clinical efficacy of nivolumab is associated with tertiary lymphoid structures in surgically resected primary tumors of recurrent gastric cancer. PLoS ONE. 2022;17(1):e0262455.PubMedPubMedCentralCrossRef
43.
go back to reference Zhao YY, Xu E, Yang X, Zhang Y, Chen H, Wang Y, Jin M. Tumor infiltrative growth pattern correlates with the immune microenvironment and is an independent factor for lymph node metastasis and prognosis in stage T1 esophageal squamous cell carcinoma. Virchows Arch. 2020;477(3):401–8.PubMedCrossRef Zhao YY, Xu E, Yang X, Zhang Y, Chen H, Wang Y, Jin M. Tumor infiltrative growth pattern correlates with the immune microenvironment and is an independent factor for lymph node metastasis and prognosis in stage T1 esophageal squamous cell carcinoma. Virchows Arch. 2020;477(3):401–8.PubMedCrossRef
44.
go back to reference He WT, Zhang D, Liu H, Chen T, Xie J, Peng L, et al. The High Level of Tertiary Lymphoid Structure Is Correlated With Superior Survival in Patients With Advanced Gastric Cancer. Front Oncol. 2020;10:980.PubMedPubMedCentralCrossRef He WT, Zhang D, Liu H, Chen T, Xie J, Peng L, et al. The High Level of Tertiary Lymphoid Structure Is Correlated With Superior Survival in Patients With Advanced Gastric Cancer. Front Oncol. 2020;10:980.PubMedPubMedCentralCrossRef
45.
go back to reference 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(3):2655–64.PubMedPubMedCentralCrossRef 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(3):2655–64.PubMedPubMedCentralCrossRef
46.
go back to reference Li H, Wang J, Liu H, Lan T, Xu L, Wang G, et al. Existence of intratumoral tertiary lymphoid structures is associated with immune cells infiltration and predicts better prognosis in early-stage hepatocellular carcinoma. Aging (Albany NY). 2020;12(4):3451–72.PubMedCrossRef Li H, Wang J, Liu H, Lan T, Xu L, Wang G, et al. Existence of intratumoral tertiary lymphoid structures is associated with immune cells infiltration and predicts better prognosis in early-stage hepatocellular carcinoma. Aging (Albany NY). 2020;12(4):3451–72.PubMedCrossRef
47.
go back to reference Zhang WH, Wang WQ, Han X, Gao HL, Xu SS, Li S, et al. Infiltrating pattern and prognostic value of tertiary lymphoid structures in resected non-functional pancreatic neuroendocrine tumors. J Immunother Cancer. 2020;8(2):e001188.PubMedPubMedCentralCrossRef Zhang WH, Wang WQ, Han X, Gao HL, Xu SS, Li S, et al. Infiltrating pattern and prognostic value of tertiary lymphoid structures in resected non-functional pancreatic neuroendocrine tumors. J Immunother Cancer. 2020;8(2):e001188.PubMedPubMedCentralCrossRef
48.
go back to reference Kuwabara S, Tsuchikawa T, Nakamura T, Hatanaka Y, Hatanaka KC, Sasaki K, et al. Prognostic relevance of tertiary lymphoid organs following neoadjuvant chemoradiotherapy in pancreatic ductal adenocarcinoma. Cancer Sci. 2019;110(6):1853–62.PubMedPubMedCentralCrossRef Kuwabara S, Tsuchikawa T, Nakamura T, Hatanaka Y, Hatanaka KC, Sasaki K, et al. Prognostic relevance of tertiary lymphoid organs following neoadjuvant chemoradiotherapy in pancreatic ductal adenocarcinoma. Cancer Sci. 2019;110(6):1853–62.PubMedPubMedCentralCrossRef
49.
go back to reference 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(7):727–39.PubMedPubMedCentralCrossRef 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(7):727–39.PubMedPubMedCentralCrossRef
50.
go back to reference 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(11):1782–90.PubMedPubMedCentralCrossRef 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(11):1782–90.PubMedPubMedCentralCrossRef
51.
go back to reference 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):2147–58.PubMedCrossRef 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):2147–58.PubMedCrossRef
52.
go back to reference Paijens ST, Vledder A, de Bruyn M, Nijman HW. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021;18(4):842–59.PubMedCrossRef Paijens ST, Vledder A, de Bruyn M, Nijman HW. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021;18(4):842–59.PubMedCrossRef
53.
go back to reference Overacre-Delgoffe AE, Bumgarner HJ, Cillo AR, Burr AHP, Tometich JT, Bhattacharjee A, et al. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity. 2021;54(12):2812–24.PubMedPubMedCentralCrossRef Overacre-Delgoffe AE, Bumgarner HJ, Cillo AR, Burr AHP, Tometich JT, Bhattacharjee A, et al. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity. 2021;54(12):2812–24.PubMedPubMedCentralCrossRef
54.
go back to reference Hill DG, Yu L, Gao H, Balic JJ, West A, Oshima H, et al. Hyperactive gp130/STAT3-driven gastric tumourigenesis promotes submucosal tertiary lymphoid structure development. Int J Cancer. 2018;143(1):167–78.PubMedPubMedCentralCrossRef Hill DG, Yu L, Gao H, Balic JJ, West A, Oshima H, et al. Hyperactive gp130/STAT3-driven gastric tumourigenesis promotes submucosal tertiary lymphoid structure development. Int J Cancer. 2018;143(1):167–78.PubMedPubMedCentralCrossRef
55.
go back to reference 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(385):eaak9679.PubMedPubMedCentralCrossRef 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(385):eaak9679.PubMedPubMedCentralCrossRef
56.
go back to reference Lee M, Heo SH, Song IH, Rajayi H, Park HS, Park IA, et al. Presence of tertiary lymphoid structures determines the level of tumor-infiltrating lymphocytes in primary breast cancer and metastasis. Mod Pathol. 2019;32(1):70–80.PubMedCrossRef Lee M, Heo SH, Song IH, Rajayi H, Park HS, Park IA, et al. Presence of tertiary lymphoid structures determines the level of tumor-infiltrating lymphocytes in primary breast cancer and metastasis. Mod Pathol. 2019;32(1):70–80.PubMedCrossRef
57.
go back to reference 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(1):250–62.PubMedCrossRef 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(1):250–62.PubMedCrossRef
58.
go back to reference Lynch KT, Young SJ, Meneveau MO, Wages NA, Engelhard VH, Slingluff CL Jr, et al. Heterogeneity in tertiary lymphoid structure B-cells correlates with patient survival in metastatic melanoma. J Immunother Cancer. 2021;9(6):e002273.PubMedPubMedCentralCrossRef Lynch KT, Young SJ, Meneveau MO, Wages NA, Engelhard VH, Slingluff CL Jr, et al. Heterogeneity in tertiary lymphoid structure B-cells correlates with patient survival in metastatic melanoma. J Immunother Cancer. 2021;9(6):e002273.PubMedPubMedCentralCrossRef
59.
go back to reference Brummel K, Eerkens AL, de Bruyn M, Nijman HW. Tumour-infiltrating lymphocytes: from prognosis to treatment selection. Br J Cancer. 2023;128(3):451–8.PubMedCrossRef Brummel K, Eerkens AL, de Bruyn M, Nijman HW. Tumour-infiltrating lymphocytes: from prognosis to treatment selection. Br J Cancer. 2023;128(3):451–8.PubMedCrossRef
60.
go back to reference Ene-Obong A, Clear AJ, Watt J, Wang J, Fatah R, Riches JC, et al. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology. 2013;145(5):1121–32.PubMedCrossRef Ene-Obong A, Clear AJ, Watt J, Wang J, Fatah R, Riches JC, et al. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology. 2013;145(5):1121–32.PubMedCrossRef
Metadata
Title
Prognostic value of tertiary lymphoid structures (TLS) in digestive system cancers: a systematic review and meta-analysis
Authors
Hao Sun
Yuanyu Shi
Hailiang Ran
Junwei Peng
Qiongxian Li
Guiqing Zheng
Yandie He
Shuqing Liu
Wei Chang
Yuanyuan Xiao
Publication date
01-12-2023
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2023
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-11738-w

Other articles of this Issue 1/2023

BMC Cancer 1/2023 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine