PDF
Abstract
In the tumour microenvironment (TME) of renal cell carcinoma (RCC), tertiary lymphoid structures (TLS) play a crucial role in anti-tumour immune responses. Resembling secondary lymphoid organs, TLS comprises B cells, T cell zones, high endothelial venules, and antigen-presenting cells, facilitating local immune activation. While TLS has shown correlations with improved immune checkpoint inhibitors (ICIs) outcomes in other cancers, its role in RCC is still under investigation. Emerging evidence indicates that mature TLS enhances anti-tumour activity by activating T and B cells, whereas immature TLS may contribute to immune suppression. The RCC TME is highly immunosuppressive, marked by regulatory T cells, myeloid-derived suppressor cells, and elevated pro-angiogenic and immunosuppressive cytokines. In this context, TLS, particularly mature TLS, can counteract immunosuppression, boost local immune responses, and improve ICIs efficacy. However, TLS in RCC is heterogeneous, with their formation and function affected by factors like CXCL13 expression. The presence, maturity, and functionality of TLS may serve as valuable predictors of ICIs response and patient prognosis. Further research is required to understand TLS regulation and leverage their potential to enhance personalised immunotherapy for RCC.
Keywords
immune checkpoint inhibitors
/
individualised therapy
/
renal cell carcinoma
/
tertiary lymphoid structures
/
tumour microenvironment
Cite this article
Download citation ▾
Lin Yang, Wentai Shangguan, Weijia Li, Wenxue Huang, Zhuohang Li, Boyuan Sun, Cunzhen Ma, Xunguo Yang, Bisheng Cheng, Peng Wu.
Role of tertiary lymphoid structures in the tumour microenvironment and immunotherapy response of renal cell carcinoma.
Clinical and Translational Medicine, 2025, 15(10): e70455 DOI:10.1002/ctm2.70455
| [1] |
Dieu-Nosjean M-C, Giraldo NA, Kaplon H, Germain C, Fridman WH, Sautès-Fridman C. Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol Rev. 2016; 271(1): 260-275.
|
| [2] |
Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science. 2022; 375(6576): eabf9419.
|
| [3] |
Sautès-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019; 19(6): 307-325.
|
| [4] |
Helmink BA, Reddy SM, Gao J, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 2020; 577(7791): 549-555.
|
| [5] |
Bukavina L, Bensalah K, Bray F, et al. Epidemiology of renal cell carcinoma: 2022 update. Euro Urol. 2022; 82(5): 529-542.
|
| [6] |
Chevrier S, Levine JH, Zanotelli VRT, et al. An immune Atlas of clear cell renal cell carcinoma. Cell. 2017; 169(4): 736-749.
|
| [7] |
Vuong L, Kotecha RR, Voss MH, Hakimi AA. Tumor microenvironment dynamics in clear-cell renal cell carcinoma. Cancer Discov. 2019; 9(10): 1349-1357.
|
| [8] |
Schödel J, Grampp S, Maher ER, et al. Hypoxia, hypoxia-inducible transcription factors, and renal cancer. Euro Urol. 2016; 69(4): 646-657.
|
| [9] |
Wettersten HI, Aboud OA, Lara PN, Weiss RH. Metabolic reprogramming in clear cell renal cell carcinoma. Nat Rev Nephrol. 2017; 13(7): 410-419.
|
| [10] |
Hu J, Wang S-G, Hou Y, et al. Multi-omic profiling of clear cell renal cell carcinoma identifies metabolic reprogramming associated with disease progression. Nat Genet. 2024; 56(3): 442-457.
|
| [11] |
Au L, Hatipoglu E, Robert de Massy M, et al. Determinants of anti-PD-1 response and resistance in clear cell renal cell carcinoma. Cancer Cell. 2021; 39(11): 1497-1518.
|
| [12] |
Barata PC, Rini BI. Treatment of renal cell carcinoma: current status and future directions. CA Cancer J Clin. 2017; 67(6): 507-524.
|
| [13] |
Motzer RJ, Tannir NM, McDermott DF, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018; 378(14): 1277-1290.
|
| [14] |
Rodriguez AB, Engelhard VH. Insights into tumor-associated tertiary lymphoid structures: novel targets for antitumor immunity and cancer immunotherapy. Cancer Immunol Res. 2020; 8(11): 1338-1345.
|
| [15] |
Vanhersecke L, Brunet M, Guégan J-P, 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.
|
| [16] |
Teillaud J-L, Houel A, Panouillot M, Riffard C, Dieu-Nosjean M-C. Tertiary lymphoid structures in anticancer immunity. Nat Rev Cancer. 2024; 24(9): 629-646.
|
| [17] |
Cabrita R, Lauss M, Sanna A, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020; 577(7791): 561-565.
|
| [18] |
Petroni G, Pillozzi S, Antonuzzo L. Exploiting tertiary lymphoid structures to stimulate antitumor immunity and improve immunotherapy efficacy. Cancer Res. 2024; 84(8): 1199-1209.
|
| [19] |
Groen-van Schooten TS, Franco Fernandez R, et al. Mapping the complexity and diversity of tertiary lymphoid structures in primary and peritoneal metastatic gastric cancer. J Immunotherap Cancer. 2024; 12(7): e009243.
|
| [20] |
Lauss M, Donia M, Svane IM, Jönsson G. B cells and tertiary lymphoid structures: friends or foes in cancer immunotherapy? Clin Cancer Res. 2022; 28(9): 1751-1758.
|
| [21] |
Masuda T, Tanaka N, Takamatsu K, et al. Unique characteristics of tertiary lymphoid structures in kidney clear cell carcinoma: prognostic outcome and comparison with bladder cancer. J Immunother Cancer. 2022; 10(3): e003883.
|
| [22] |
Meylan M, Petitprez F, Becht E, et al. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. Immunity. 2022; 55(3): 527-541.
|
| [23] |
Xu W, Lu J, Liu W-R, et al. Heterogeneity in tertiary lymphoid structures predicts distinct prognosis and immune microenvironment characterizations of clear cell renal cell carcinoma. J Immunother Cancer. 2023; 11(12): e006667.
|
| [24] |
Wang Y-Q, Chen W-J, Zhou W, et al. Integrated analysis of tertiary lymphoid structures and immune infiltration in ccRCC microenvironment revealed their clinical significances: a multicenter cohort study. J Immunother Cancer. 2024; 12(6): e008613.
|
| [25] |
Sato Y, Silina K, van den Broek M, Hirahara K, Yanagita M. The roles of tertiary lymphoid structures in chronic diseases. Nat Rev Nephrol. 2023; 19(8): 525-537.
|
| [26] |
Onder L, Papadopoulou C, Lütge A, et al. Fibroblastic reticular cells generate protective intratumoral T cell environments in lung cancer. Cell. 2025; 188(2): 430-446.
|
| [27] |
Blanchard L, Girard J-P. High endothelial venules (HEVs) in immunity, inflammation and cancer. Angiogenesis. 2021; 24(4): 719-753.
|
| [28] |
Vella G, Hua Y, Bergers G. High endothelial venules in cancer: regulation, function, and therapeutic implication. Cancer Cell. 2023; 41(3): 527-545.
|
| [29] |
Hu L, Li T, Deng S, et al. Tertiary lymphoid structure formation induced by LIGHT-engineered and photosensitive nanoparticles-decorated bacteria enhances immune response against colorectal cancer. Biomaterials. 2025; 314: 122846.
|
| [30] |
Wang X, Li X, Zhao J, et al. Rapid generation of hPSC-derived high endothelial venule organoids with in vivo ectopic lymphoid tissue capabilities. Adv Mater. 2024; 36(15): e2308760.
|
| [31] |
Tang H, Zhu M, Qiao J, Fu Y-X. Lymphotoxin signalling in tertiary lymphoid structures and immunotherapy. Cell Mol Immunol. 2017; 14(10): 809-818.
|
| [32] |
Clubb JHA, Kudling TV, Heiniö C, et al. Adenovirus encoding tumor necrosis factor alpha and interleukin 2 induces a tertiary lymphoid structure signature in immune checkpoint inhibitor refractory head and neck cancer. Front Immunol. 2022; 13: 794251.
|
| [33] |
Sadeghirad H, Monkman J, Tan CW, et al. Spatial dynamics of tertiary lymphoid aggregates in head and neck cancer: insights into immunotherapy response. J Transl Med. 2024; 22(1): 677.
|
| [34] |
Vahidian F, Lamaze FC, Bouffard C, et al. CXCL13 positive cells localization predict response to anti-PD-1/PD-L1 in pulmonary non-small cell carcinoma. Cancers (Basel). 2024; 16(4): 708.
|
| [35] |
Bao X, Lin X, Xie M, et al. Mature tertiary lymphoid structures: important contributors to anti-tumor immune efficacy. Front Immunol. 2024; 15: 1413067.
|
| [36] |
Sun X, Liu W, Sun L, et al. Maturation and abundance of tertiary lymphoid structures are associated with the efficacy of neoadjuvant chemoimmunotherapy in resectable non-small cell lung cancer. J Immunotherap Cancer. 2022; 10(11): e005531.
|
| [37] |
Giatromanolaki A, Chatzipantelis P, Contrafouris CA, Koukourakis MI. Tertiary lymphoid structures, immune response, and prognostic relevance in non-small cell lung cancer. Cancer Invest. 2023; 41(1): 48-57.
|
| [38] |
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 Immunotherap Cancer. 2021; 9(2): e001906.
|
| [39] |
Zhao Y-Y, Fan Z, Tao B-R, Du Z-G, Shi Z-F. Density of tertiary lymphoid structures predicts clinical outcome in breast cancer brain metastasis. J Immunotherap Cancer. 2024; 12(7): e009232.
|
| [40] |
Lee HJ, Park IA, Song IH, et al. Tertiary lymphoid structures: prognostic significance and relationship with tumour-infiltrating lymphocytes in triple-negative breast cancer. J Clin Pathol. 2016; 69(5): 422-430.
|
| [41] |
Zou X, Lin X, Cheng H, et al. Characterization of intratumoral tertiary lymphoid structures in pancreatic ductal adenocarcinoma: cellular properties and prognostic significance. J Immunotherap Cancer. 2023; 11(6): e006698.
|
| [42] |
Kinker GS, Vitiello GAF, Diniz AB, et al. Mature tertiary lymphoid structures are key niches of tumour-specific immune responses in pancreatic ductal adenocarcinomas. Gut. 2023; 72(10): 1927-1941.
|
| [43] |
Calderaro J, Petitprez F, Becht E, 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.
|
| [44] |
Xu W, Lu J, Tian X, et al. Unveiling the impact of tertiary lymphoid structures on immunotherapeutic responses of clear cell renal cell carcinoma. MedComm (2020). 2024; 5(1): e461.
|
| [45] |
Braun DA, Street K, Burke KP, et al. Progressive immune dysfunction with advancing disease stage in renal cell carcinoma. Cancer Cell. 2021; 39(5): 632-648.
|
| [46] |
Sharma M, Khong H, Fa'ak F, et al. Bempegaldesleukin selectively depletes intratumoral Tregs and potentiates T cell-mediated cancer therapy. Nat Commun. 2020; 11(1): 661.
|
| [47] |
Zhang C, Chen L, Liu Y, et al. Downregulated METTL14 accumulates BPTF that reinforces super-enhancers and distal lung metastasis via glycolytic reprogramming in renal cell carcinoma. Theranostics. 2021; 11(8): 3676-3693.
|
| [48] |
Bacigalupa ZA, Rathmell WK. Beyond glycolysis: hypoxia signaling as a master regulator of alternative metabolic pathways and the implications in clear cell renal cell carcinoma. Cancer Letters. 2020; 489: 19-28.
|
| [49] |
Messai Y, Gad S, Noman MZ, et al. Renal cell carcinoma programmed death-ligand 1, a new direct target of hypoxia-inducible factor-2 alpha, is regulated by von Hippel-Lindau gene mutation status. Euro Urol. 2016; 70(4): 623-632.
|
| [50] |
Zhu Z, Jin Y, Zhou J, et al. PD1/PD-L1 blockade in clear cell renal cell carcinoma: mechanistic insights, clinical efficacy, and future perspectives. Mol Cancer. 2024; 23(1): 146.
|
| [51] |
Ghoreifi A, Vaishampayan U, Yin M, Psutka SP, Djaladat H. Immune checkpoint inhibitor therapy before nephrectomy for locally advanced and metastatic renal cell carcinoma: a review. JAMA Oncol. 2024; 10(2): 240-248.
|
| [52] |
Hugaboom MB, Wirth LV, Street K, et al. Presence of tertiary lymphoid structures and exhausted tissue-resident T cells determines clinical response to PD-1 blockade in renal cell carcinoma. Cancer Discov. 2025; 15(5): 948-968.
|
| [53] |
Mella M, Kauppila JH, Karihtala P, et al. Tumor infiltrating CD8+ T lymphocyte count is independent of tumor TLR9 status in treatment naïve triple negative breast cancer and renal cell carcinoma. Oncoimmunology. 2015; 4(6): e1002726.
|
| [54] |
Tang Y, Chen J, Zhang M, et al. Tertiary lymphoid structures potentially promote immune checkpoint inhibitor response in SMARCB1-deficient medullary renal cell carcinoma. NPJ Precis Oncol. 2024; 8(1): 261.
|
| [55] |
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.
|
| [56] |
Hu C, You W, Kong D, et al. Tertiary lymphoid structure-associated B cells enhance CXCL13+CD103+CD8+ tissue-resident memory T-Cell response to programmed cell death protein 1 blockade in cancer immunotherapy. Gastroenterology. 2024; 166(6): 1069-1084.
|
| [57] |
Kasikova L, Rakova J, Hensler M, et al. Tertiary lymphoid structures and B cells determine clinically relevant T cell phenotypes in ovarian cancer. Nat Commun. 2024; 15(1): 2528.
|
| [58] |
Zhang Y, Xu M, Ren Y, et al. Tertiary lymphoid structural heterogeneity determines tumour immunity and prospects for clinical application. Mol Cancer. 2024; 23(1): 75.
|
| [59] |
Ishigami E, Sakakibara M, Sakakibara J, et al. Coexistence of regulatory B cells and regulatory T cells in tumor-infiltrating lymphocyte aggregates is a prognostic factor in patients with breast cancer. Breast Cancer. 2019; 26(2): 180-189.
|
| [60] |
Joshi NS, Akama-Garren EH, Lu Y, et al. Regulatory T cells in tumor-associated tertiary lymphoid structures suppress anti-tumor T cell responses. Immunity. 2015; 43(3): 579-590.
|
| [61] |
Devi-Marulkar P, Fastenackels S, Karapentiantz P, et al. Regulatory T cells infiltrate the tumor-induced tertiary lymphoïd structures and are associated with poor clinical outcome in NSCLC. Commun Biol. 2022; 5(1): 1416.
|
| [62] |
Germain C, Devi-Marulkar P, Knockaert S, et al. Tertiary lymphoid structure-B cells narrow regulatory T cells impact in lung cancer patients. Front Immunol. 2021; 12: 626776.
|
| [63] |
N J, J T, Sl N, Gt B. Tertiary lymphoid structures and B lymphocytes in cancer prognosis and response to immunotherapies. Oncoimmunology. 2021; 10(1): 1900508.
|
| [64] |
Maalej KM, Merhi M, Inchakalody VP, et al. CAR-cell therapy in the era of solid tumor treatment: current challenges and emerging therapeutic advances. Mol Cancer. 2023; 22(1): 20.
|
| [65] |
Ma S, Li X, Wang X, et al. Current progress in CAR-T cell therapy for solid tumors. Int J Biol Sci. 2019; 15(12): 2548-2560.
|
| [66] |
Sabahi M, Salehipour A, Bazl MSY, Rezaei N, Mansouri A, Borghei-Razavi H. Local immunotherapy of glioblastoma: a comprehensive review of the concept. J Neuroimmunol. 2023; 381: 578146.
|
| [67] |
Wagner J, Wickman E, DeRenzo C, Gottschalk S. CAR T cell therapy for solid tumors: bright future or dark reality? Mol Ther. 2020; 28(11): 2320-2339.
|
| [68] |
Corti C, Venetis K, Sajjadi E, Zattoni L, Curigliano G, Fusco N. CAR-T cell therapy for triple-negative breast cancer and other solid tumors: preclinical and clinical progress. Expert Opin Investig Drugs. 2022; 31(6): 593-605.
|
| [69] |
Zhang N, Liu X, Qin J, et al. LIGHT/TNFSF14 promotes CAR-T cell trafficking and cytotoxicity through reversing immunosuppressive tumor microenvironment. Mol Ther. 2023; 31(9): 2575-2590.
|
| [70] |
Nakamura S, Ohuchida K, Hayashi M, et al. Tertiary lymphoid structures correlate with enhancement of antitumor immunity in esophageal squamous cell carcinoma. British Journal of Cancer. 2023; 129(8): 1314-1326.
|
| [71] |
Wang Q, Sun K, Liu R, et al. Single-cell transcriptome sequencing of B-cell heterogeneity and tertiary lymphoid structure predicts breast cancer prognosis and neoadjuvant therapy efficacy. Clin Transl Med. 2023; 13(8): e1346.
|
| [72] |
Lanickova T, Hensler M, Kasikova L, et al. Chemotherapy drives tertiary lymphoid structures that correlate with ICI-responsive TCF1+CD8+ T cells in metastatic ovarian cancer. Clin Cancer Res. 2024; 31(1): 164-180.
|
| [73] |
Lin J, Jiang S, Chen B, et al. Tertiary lymphoid structures are linked to enhanced antitumor immunity and better prognosis in muscle-invasive bladder cancer. Adv Sci (Weinh). 2025; 12(7): e2410998.
|
| [74] |
Wang J, Liang Y, Xue A, et al. Intratumoral CXCL13+ CD160+ CD8+ T cells promote the formation of tertiary lymphoid structures to enhance the efficacy of immunotherapy in advanced gastric cancer. J Immunotherap Cancer. 2024; 12(9): e009603.
|
| [75] |
Rouanne M, Arpaia N, Marabelle A. CXCL13 shapes tertiary lymphoid structures and promotes response to immunotherapy in bladder cancer. Eur J Cancer. 2021; 151: 245-248.
|
| [76] |
Dai S, Zeng H, Liu Z, et al. Intratumoral CXCL13+CD8+T cell infiltration determines poor clinical outcomes and immunoevasive contexture in patients with clear cell renal cell carcinoma. J Immunother Cancer. 2021; 9(2): e001823.
|
| [77] |
Chaurio RA, Anadon CM, Lee Costich T, 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): 115-128.
|
| [78] |
Li J-P, Wu C-Y, Chen M-Y, et al. PD-1+CXCR5-CD4+ Th-CXCL13 cell subset drives B cells into tertiary lymphoid structures of nasopharyngeal carcinoma. J Immunother Cancer. 2021; 9(7): e002101.
|
| [79] |
Fridman WH, Meylan M, Petitprez F, Sun C-M, Italiano A. Sautès-Fridman C. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat Rev Clin Oncol. 2022; 19(7): 441-457.
|
| [80] |
Ding L, Sun L, Bu MT, et al. Antigen presentation by clonally diverse CXCR5+ B cells to CD4 and CD8 T cells is associated with durable response to immune checkpoint inhibitors. Front Immunol. 2023; 14: 1176994.
|
| [81] |
Hsieh C-H, Jian C-Z, Lin L-I, et al. Potential role of CXCL13/CXCR5 signaling in immune checkpoint inhibitor treatment in cancer. Cancers (Basel). 2022; 14(2): 294.
|
| [82] |
He M, He Q, Cai X, et al. Intratumoral tertiary lymphoid structure (TLS) maturation is influenced by draining lymph nodes of lung cancer. J Immunotherap Cancer. 2023; 11(4): e005539.
|
| [83] |
Allen E, Jabouille A, Rivera LB, et al. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med. 2017; 9(385): eaak9679.
|
| [84] |
Park JA, Espinosa-Cotton M, Guo H-F, Monette S, Cheung N-KV. Targeting tumor vasculature to improve antitumor activity of T cells armed ex vivo with T cell engaging bispecific antibody. J Immunotherap Cancer. 2023; 11(3): e006680.
|
| [85] |
Zhang Y, Liu G, Zeng Q, et al. CCL19-producing fibroblasts promote tertiary lymphoid structure formation enhancing anti-tumor IgG response in colorectal cancer liver metastasis. Cancer Cell. 2024; 42(8): 1370-1385.
|
| [86] |
Melssen MM, Sheybani ND, Leick KM, Slingluff CL. Barriers to immune cell infiltration in tumors. J Immunotherap Cancer. 2023; 11(4): e006401.
|
| [87] |
Guo S, Yuan J, Meng X, et al. Cancer-associated fibroblasts: just on the opposite side of antitumour immunity? Int Immunopharmacol. 2023; 122: 110601.
|
| [88] |
Omotesho QA, Escamilla A, Pérez-Ruiz E, Frecha CA, Rueda-Domínguez A, Barragán I. Epigenetic targets to enhance antitumor immune response through the induction of tertiary lymphoid structures. Front Immunol. 2024; 15: 1348156.
|
| [89] |
Weinstein AM, Giraldo NA, Petitprez F, et al. Association of IL-36γ with tertiary lymphoid structures and inflammatory immune infiltrates in human colorectal cancer. Cancer Immunol Immunother. 2019; 68(1): 109-120.
|
| [90] |
Garcia J, Hurwitz HI, Sandler AB, et al. Bevacizumab (Avastin®) in cancer treatment: a review of 15 years of clinical experience and future outlook. Cancer Treat Rev. 2020; 86: 102017.
|
| [91] |
Powles T, Atkins MB, Escudier B, et al. Efficacy and safety of atezolizumab plus bevacizumab following disease progression on atezolizumab or sunitinib monotherapy in patients with metastatic renal cell carcinoma in IMmotion150: a randomized phase 2 clinical trial. Europ Urol. 2021; 79(5): 665-673.
|
| [92] |
Amisaki M, Zebboudj A, Yano H, et al. IL-33-activated ILC2s induce tertiary lymphoid structures in pancreatic cancer. Nature. 2025; 638(8052): 1076-1084.
|
| [93] |
Yang M, Lu J, Zhang G, et al. CXCL13 shapes immunoactive tumor microenvironment and enhances the efficacy of PD-1 checkpoint blockade in high-grade serous ovarian cancer. J Immunotherap Cancer. 2021; 9(1): e001136.
|
| [94] |
Yuan H, Mao X, Yan Y, et al. Single-cell sequencing reveals the heterogeneity of B cells and tertiary lymphoid structures in muscle-invasive bladder cancer. J Transl Med. 2024; 22(1): 48.
|
| [95] |
An D, Chen G, Cheng W-Y, et al. LTβR agonism promotes antitumor immune responses via modulation of the tumor microenvironment. Cancer Res. 2024; 84(23): 3984-4001.
|
| [96] |
Calvanese AL, Cecconi V, Stäheli S, et al. Sustained innate interferon is an essential inducer of tertiary lymphoid structures. Eur J Immunol. 2024; 54(10): e2451207.
|
| [97] |
Dai Z, Wang Q, Tang J, et al. A Sub-6 nm MnFe2O4-dichloroacetic acid nanocomposite modulates tumor metabolism and catabolism for reversing tumor immunosuppressive microenvironment and boosting immunotherapy. Biomaterials. 2022; 284: 121533.
|
| [98] |
Rodriguez AB, Peske JD, Woods AN, et al. Immune mechanisms orchestrate tertiary lymphoid structures in tumors via cancer-associated fibroblasts. Cell Rep. 2021; 36(3): 109422.
|
| [99] |
Mao Y, Wang X, Xi L, et al. Prediction values of tertiary lymphoid structures in the prognosis of patients with left- and right-sided colon cancer: a multicenter propensity score-matched study. Int J Surg. 2023; 109(8): 2344-2358.
|
| [100] |
Lei J-X, Wang R, Hu C, et al. Deciphering tertiary lymphoid structure heterogeneity reveals prognostic signature and therapeutic potentials for colorectal cancer: a multicenter retrospective cohort study. Int J Surg. 2024; 110(9): 5627-5640.
|
| [101] |
Vanhersecke L, Bougouin A, Crombé A, et al. Standardized pathology screening of mature tertiary lymphoid structures in cancers. Lab Invest. 2023; 103(5): 100063.
|
| [102] |
Li S, Zhang N, Zhang H, et al. Deciphering the role of LGALS2: insights into tertiary lymphoid structure-associated dendritic cell activation and immunotherapeutic potential in breast cancer patients. Mol Cancer. 2024; 23(1): 216.
|
| [103] |
Fischer K, Hoffmann P, Voelkl S, et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood. 2007; 109(9): 3812-3819.
|
| [104] |
Xu W, Ma C, Liu W, et al. Prognostic value, DNA variation and immunologic features of a tertiary lymphoid structure-related chemokine signature in clear cell renal cell carcinoma. Cancer Immunol Immunother. 2022; 71(8): 1923-1935.
|
RIGHTS & PERMISSIONS
2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.