Tertiary lymphoid structures in cancer: immune mechanisms and clinical implications

Siyu Wang1,2,3, Hua Wang1,2, Chenbei Li1,2, Binfeng Liu1,2, Shasha He4(), Chao Tu1,2,5,6()

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (3) : e489. DOI: 10.1002/mco2.489
REVIEW

Tertiary lymphoid structures in cancer: immune mechanisms and clinical implications

  • Siyu Wang1,2,3, Hua Wang1,2, Chenbei Li1,2, Binfeng Liu1,2, Shasha He4(), Chao Tu1,2,5,6()
Author information +
History +

Abstract

Cancer is a major cause of death globally, and traditional treatments often have limited efficacy and adverse effects. Immunotherapy has shown promise in various malignancies but is less effective in tumors with low immunogenicity or immunosuppressive microenvironment, especially sarcomas. Tertiary lymphoid structures (TLSs) have been associated with a favorable response to immunotherapy and improved survival in cancer patients. However, the immunological mechanisms and clinical significance of TLS in malignant tumors are not fully understood. In this review, we elucidate the composition, neogenesis, and immune characteristics of TLS in tumors, as well as the inflammatory response in cancer development. An in-depth discussion of the unique immune characteristics of TLSs in lung cancer, breast cancer, melanoma, and soft tissue sarcomas will be presented. Additionally, the therapeutic implications of TLS, including its role as a marker of therapeutic response and prognosis, and strategies to promote TLS formation and maturation will be explored. Overall, we aim to provide a comprehensive understanding of the role of TLS in the tumor immune microenvironment and suggest potential interventions for cancer treatment.

Keywords

clinical implications / immune mechanisms / soft tissue sarcoma / tertiary lymphoid structures (TLSs) / tumor immune microenvironment (TIME)

Cite this article

Download citation ▾
Siyu Wang, Hua Wang, Chenbei Li, Binfeng Liu, Shasha He, Chao Tu. Tertiary lymphoid structures in cancer: immune mechanisms and clinical implications. MedComm, 2024, 5(3): e489 https://doi.org/10.1002/mco2.489

References

1 J Qi, M Li, L Wang, et al. National and subnational trends in cancer burden in China, 2005–20: an analysis of national mortality surveillance data. Lancet Public Health. 2023;8(12):e943-e955.
2 YC Huang, MY Sung, TK Lin, CY Kuo, YC Hsu. Chinese herbal medicine compound of flavonoids adjunctive treatment for oral cancer. J Formos Med Assoc. 2023.
3 T Olivier, D Pop, A Chouiter Djebaili, et al. Treating metastatic sarcomas locally: a paradoxe, a rationale, an evidence? Crit Rev Oncol Hematol. 2015;95(1):62-77.
4 N Liu, X Xiao, Z Zhang, C Mao, M Wan, J Shen. Advances in cancer vaccine research. ACS Biomater Sci Eng. 2023;9(11):5999-6023.
5 V Matson, CS Chervin, TF Gajewski. Cancer and the microbiome-influence of the commensal microbiota on cancer, immune responses, and immunotherapy. Gastroenterology. 2021;160(2):600-613.
6 TN Schumacher, DS Thommen. Tertiary lymphoid structures in cancer. Science. 2022;375(6576):eabf9419.
7 Z Zhao, H Ding, ZB Lin, et al. Relationship between tertiary lymphoid structure and the prognosis and clinicopathologic characteristics in solid tumors. Int J Med Sci. 2021;18(11):2327-2338.
8 G Ma, H Jia, G Zhang, et al. Presence, subtypes, and prognostic significance of tertiary lymphoid structures in urothelial carcinoma of the bladder. Oncologist. 2023:oyad283.
9 GY Ding, JQ Ma, JP Yun, et al. Distribution and density of tertiary lymphoid structures predict clinical outcome in intrahepatic cholangiocarcinoma. J Hepatol. 2022;76(3):608-618.
10 S Nakamura, K Ohuchida, M Hayashi, et al. Tertiary lymphoid structures correlate with enhancement of antitumor immunity in esophageal squamous cell carcinoma. Br J Cancer. 2023;129(8):1314-1326.
11 W Feng, D Jiang, Y Xu, et al. CDK4/6i enhances the antitumor effect of PD1 antibody by promoting TLS formation in ovarian cancer. Heliyon. 2023;9(9):e19760.
12 R Kee, M Naughton, GV McDonnell, OW Howell, DC Fitzgerald. A review of compartmentalised inflammation and tertiary lymphoid structures in the pathophysiology of multiple sclerosis. Biomedicines. 2022;10(10):2604.
13 LL Munn. Cancer and inflammation. Wiley Interdiscip Rev Syst Biol Med. 2017;9(2).
14 NN Zhang, FJ Qu, H Liu, et al. Prognostic impact of tertiary lymphoid structures in breast cancer prognosis: a systematic review and meta-analysis. Cancer Cell Int. 2021;21(1):536.
15 R Cabrita, M Lauss, A Sanna, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577(7791):561-565.
16 A Italiano, A Bessede, M Pulido, et al. Pembrolizumab in soft-tissue sarcomas with tertiary lymphoid structures: a phase 2 PEMBROSARC trial cohort. Nat Med. 2022;28(6):1199-1206.
17 MJ Melake, HG Smith, D Mansfield, et al. OX40 and 4-1BB delineate distinct immune profiles in sarcoma. Oncoimmunology. 2022;11(1):2066050.
18 Y Hua, G Vella, F Rambow, et al. Cancer immunotherapies transition endothelial cells into HEVs that generate TCF1(+) T lymphocyte niches through a feed-forward loop. Cancer Cell. 2022;40(12):1600-1618. e10.
19 M Chelvanambi, RJ Fecek, JL Taylor, WJ Storkus. STING agonist-based treatment promotes vascular normalization and tertiary lymphoid structure formation in the therapeutic melanoma microenvironment. J Immunother Cancer. 2021;9(2):e001906.
20 W Liu, CH Tai, X Liu, I Pastan. Anti-mesothelin immunotoxin induces mesothelioma eradication, anti-tumor immunity, and the development of tertiary lymphoid structures. Proc Natl Acad Sci USA. 2022;119(48):e2214928119.
21 EM Pimenta, BJ Barnes. Role of tertiary lymphoid structures (TLS) in anti-tumor immunity: potential tumor-induced cytokines/chemokines that regulate TLS formation in epithelial-derived cancers. Cancers (Basel). 2014;6(2):969-997.
22 Y Sato, A Jain, S Ohtsuki, et al. Stem-like CD4(+) T cells in perivascular tertiary lymphoid structures sustain autoimmune vasculitis. Sci Transl Med. 2023;15(712):eadh0380.
23 Y Yamada, TT Nguyen, D Impellizzieri, et al. Biased IL-2 signals induce Foxp3-rich pulmonary lymphoid structures and facilitate long-term lung allograft acceptance in mice. Nat Commun. 2023;14(1):1383.
24 SJ Im, RC Obeng, TH Nasti, et al. Characteristics and anatomic location of PD-1(+)TCF1(+) stem-like CD8 T cells in chronic viral infection and cancer. Proc Natl Acad Sci USA. 2023;120(41):e2221985120.
25 M He, Q He, X Cai, et al. Intratumoral tertiary lymphoid structure (TLS) maturation is influenced by draining lymph nodes of lung cancer. J Immunother Cancer. 2023;11(4):e005539.
26 CG Mueller, S Nayar, J Campos, F Barone. Molecular and cellular requirements for the assembly of tertiary lymphoid structures. Adv Exp Med Biol. 2018;1060:55-72.
27 M Bombardieri, F Barone, D Lucchesi, et al. Inducible tertiary lymphoid structures, autoimmunity, and exocrine dysfunction in a novel model of salivary gland inflammation in C57BL/6 mice. J Immunol. 2012;189(7):3767-3776.
28 J Verneau, C Sautés-Fridman, CM Sun. Dendritic cells in the tumor microenvironment: prognostic and theranostic impact. Semin Immunol. 2020;48:101410.
29 C Gago da Gra?a, LGM van Baarsen, RE Mebius. Tertiary lymphoid structures: diversity in their development, composition, and role. J Immunol. 2021;206(2):273-281.
30 SA van de Pavert, BJ Olivier, G Goverse, et al. Chemokine CXCL13 is essential for lymph node initiation and is induced by retinoic acid and neuronal stimulation. Nat Immunol. 2009;10(11):1193-1199.
31 AT Krishnamurty, SJ Turley. Lymph node stromal cells: cartographers of the immune system. Nat Immunol. 2020;21(4):369-380.
32 JL Grogan, W Ouyang. A role for Th17 cells in the regulation of tertiary lymphoid follicles. Eur J Immunol. 2012;42(9):2255-2262.
33 S Sivori, D Pende, L Quatrini, et al. NK cells and ILCs in tumor immunotherapy. Mol Aspects Med. 2021;80:100870.
34 NH Ruddle. Basics of Inducible Lymphoid Organs. Curr Top Microbiol Immunol. 2020;426:1-19.
35 MY Kim. Roles of embryonic and adult lymphoid tissue inducer cells in primary and secondary lymphoid tissues. Yonsei Med J. 2008;49(3):352-356.
36 Y Lee, RK Chin, P Christiansen, et al. Recruitment and activation of naive T cells in the islets by lymphotoxin beta receptor-dependent tertiary lymphoid structure. Immunity. 2006;25(3):499-509.
37 C Bénézech, NT Luu, JA Walker, et al. Inflammation-induced formation of fat-associated lymphoid clusters. Nat Immunol. 2015;16(8):819-828.
38 AL Fletcher, SE Acton, K Knoblich. Lymph node fibroblastic reticular cells in health and disease. Nat Rev Immunol. 2015;15(6):350-361.
39 L de Chaisemartin, J Goc, D Damotte, 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(20):6391-6399.
40 M Kwak, G Erdag, KM Leick, S Bekiranov, VH Engelhard, CL Slingluff. Associations of immune cell homing gene signatures and infiltrates of lymphocyte subsets in human melanomas: discordance with CD163(+) myeloid cell infiltrates. J Transl Med. 2021;19(1):371.
41 CP Freier, C Kuhn, S Endres, et al. FOXP3+ cells recruited by CCL22 into breast cancer correlates with less tumor nodal infiltration. Anticancer Res. 2016;36(6):3139-3145.
42 HY Huang, A Rivas-Caicedo, F Renevey, et al. Identification of a new subset of lymph node stromal cells involved in regulating plasma cell homeostasis. Proc Natl Acad Sci USA. 2018;115(29):E6826-E6835.
43 F Ciccia, A Rizzo, R Maugeri, et al. Ectopic expression of CXCL13, BAFF, APRIL and LT-β is associated with artery tertiary lymphoid organs in giant cell arteritis. Ann Rheum Dis. 2017;76(1):235-243.
44 R Luo, Y Cheng, D Chang, et al. Tertiary lymphoid organs are associated with the progression of kidney damage and regulated by interleukin-17A. Theranostics. 2021;11(1):117-131.
45 ZZ Wang, J Song, H Wang, et al. Stromal cells and B cells orchestrate ectopic lymphoid tissue formation in nasal polyps. Allergy. 2021;76(5):1416-1431.
46 I Truxova, L Kasikova, M Hensler, et al. Mature dendritic cells correlate with favorable immune infiltrate and improved prognosis in ovarian carcinoma patients. J Immunother Cancer. 2018;6(1):139.
47 S Wakasu, T Tagawa, N Haratake, et al. Preventive effect of tertiary lymphoid structures on lymph node metastasis of lung adenocarcinoma. Cancer Immunol Immunother. 2023;72(6):1823-1834.
48 N Jacquelot, J Tellier, Sl Nutt, GT Belz. Tertiary lymphoid structures and B lymphocytes in cancer prognosis and response to immunotherapies. Oncoimmunology. 2021;10(1):1900508.
49 T Cupedo, W Jansen, G Kraal, RE Mebius. Induction of secondary and tertiary lymphoid structures in the skin. Immunity. 2004;21(5):655-667.
50 L Onder, R Danuser, E Scandella, et al. Endothelial cell-specific lymphotoxin-β receptor signaling is critical for lymph node and high endothelial venule formation. J Exp Med. 2013;210(3):465-473.
51 K Sili?a, A Soltermann, FM Attar, 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.
52 X Chao, L Liu, P Sun, et al. Immune parameters associated with survival in metaplastic breast cancer. Breast Cancer Res. 2020;22(1):92.
53 Y Xu, Z Li, Y Yang, et al. A CT-based radiomics approach to predict intra-tumoral tertiary lymphoid structures and recurrence of intrahepatic cholangiocarcinoma. Insights Imaging. 2023;14(1):173.
54 M Yang, Y Che, K Li, et al. Detection and quantitative analysis of tumor-associated tertiary lymphoid structures. J Zhejiang Univ Sci B. 2023;24(9):779-795.
55 G Sarti Kinker, T da Silva Medina. Tertiary lymphoid structures as hubs of antitumour immunity. Nat Rev Cancer. 2023;23(12):803.
56 J Rangel-Moreno, DM Carragher, M de la Luz Garcia-Hernandez, et al. The development of inducible bronchus-associated lymphoid tissue depends on IL-17. Nat Immunol. 2011;12(7):639-646.
57 L Berteloot, TJ Molina, J Bruneau, et al. Alternative pathways for the development of lymphoid structures in humans. Proc Natl Acad Sci USA. 2021;118(29):e2108082118.
58 F Balkwill, A Mantovani. Inflammation and cancer: back to Virchow? Lancet. 2001;357(9255):539-545.
59 J Zheng, J Mo, T Zhu, et al. Comprehensive elaboration of the cGAS-STING signaling axis in cancer development and immunotherapy. Mol Cancer. 2020;19(1):133.
60 H Lemos, E Mohamed, L Huang, et al. STING Promotes the growth of tumors characterized by low antigenicity via IDO activation. Cancer Res. 2016;76(8):2076-2081.
61 SS Du, GW Chen, P Yang, et al. Radiation therapy promotes hepatocellular carcinoma immune cloaking via PD-L1 upregulation induced by cGAS-STING activation. Int J Radiat Oncol Biol Phys. 2022;112(5):1243-1255.
62 H Liang, L Deng, Y Hou, et al. Host STING-dependent MDSC mobilization drives extrinsic radiation resistance. Nat Commun. 2017;8(1):1736.
63 B Larkin, V Ilyukha, M Sorokin, A Buzdin, E Vannier, A Poltorak. Cutting edge: activation of STING in T cells induces type I IFN responses and cell death. J Immunol. 2017;199(2):397-402.
64 A Crombé, M Roulleau-Dugage, A Italiano. The diagnosis, classification, and treatment of sarcoma in this era of artificial intelligence and immunotherapy. Cancer Commun (Lond). 2022;42(12):1288-1313.
65 F Petitprez, A de Reyniès, EZ Keung, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577(7791):556-560.
66 S Morand, M Devanaboyina, H Staats, L Stanbery, J Nemunaitis. Ovarian cancer immunotherapy and personalized medicine. Int J Mol Sci. 2021;22(12):6532.
67 A Marabelle, M Fakih, J Lopez, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020;21(10):1353-1365.
68 D Lu, Y Chen, M Jiang, et al. KRAS G12V neoantigen specific T cell receptor for adoptive T cell therapy against tumors. Nat Commun. 2023;14(1):6389.
69 R Wei, DC Dean, P Thanindratarn, FJ Hornicek, W Guo, Z Duan. Cancer testis antigens in sarcoma: expression, function and immunotherapeutic application. Cancer Lett. 2020;479:54-60.
70 M Vasella, E Gousopoulos, M Guidi, et al. Targeted therapies and checkpoint inhibitors in sarcoma. Qjm. 2022;115(12):793-805.
71 V Meftahpour, A Aghebati-Maleki, A Fotouhi, E Safarzadeh, L Aghebati-Maleki. Prognostic significance and therapeutic potentials of immune checkpoints in osteosarcoma. Excli J. 2022;21:250-268.
72 M Bugatti, M Bergamini, F Missale, et al. A population of TIM4+FOLR2+ macrophages localized in tertiary lymphoid structures correlates to an active immune infiltrate across several cancer types. Cancer Immunol Res. 2022;10(11):1340-1353.
73 SL Topalian, FS Hodi, JR Brahmer, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443-2454.
74 FH Wu, Y Yuan, D Li, et al. Endothelial cell-expressed Tim-3 facilitates metastasis of melanoma cells by activating the NF-kappaB pathway. Oncol Rep. 2010;24(3):693-699.
75 SC Wei, CR Duffy, JP Allison. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8(9):1069-1086.
76 S Sánchez-Molina, E Figuerola-Bou, V Sánchez-Margalet, et al. Ewing sarcoma meets epigenetics, immunology and nanomedicine: moving forward into novel therapeutic strategies. Cancers (Basel). 2022;14(21):5473.
77 R Wu, W Guo, X Qiu, et al. Comprehensive analysis of spatial architecture in primary liver cancer. Sci Adv. 2021;7(51):eabg3750.
78 W Xu, J Yang, S Ye, et al. Insights into the maturation heterogeneity of tumor-associated tertiary lymphoid structures in cancer immunotherapy. Immunotherapy. 2024;16(3):131-134.
79 Y Ling, J Zhong, Z Weng, et al. The prognostic value and molecular properties of tertiary lymphoid structures in oesophageal squamous cell carcinoma. Clin Transl Med. 2022;12(10):e1074.
80 M Meylan, F Petitprez, L Lacroix, et al. Early hepatic lesions display immature tertiary lymphoid structures and show elevated expression of immune inhibitory and immunosuppressive molecules. Clin Cancer Res. 2020;26(16):4381-4389.
81 WH Fridman, S Sibéril, G Pupier, S Soussan, C Sautès-Fridman. Activation of B cells in tertiary lymphoid structures in cancer: anti-tumor or anti-self? Semin Immunol. 2023;65:101703.
82 S Tietscher, J Wagner, T Anzeneder, et al. A comprehensive single-cell map of T cell exhaustion-associated immune environments in human breast cancer. Nat Commun. 2023;14(1):98.
83 S Yin, C Zhang, F Gao. Immature tertiary lymphoid structure formation was increased in the melanoma tumor microenvironment of IKZF1 transgenic mice. Transl Cancer Res. 2022;11(7):2388-2397.
84 C Zhang, XY Wang, JL Zuo, et al. Localization and density of tertiary lymphoid structures associate with molecular subtype and clinical outcome in colorectal cancer liver metastases. J Immunother Cancer. 2023;11(2):e006425.
85 T Zhang, X Lei, W Jia, et al. Peritumor tertiary lymphoid structures are associated with infiltrating neutrophils and inferior prognosis in hepatocellular carcinoma. Cancer Med. 2023;12(3):3068-3078.
86 M Sofopoulos, SP Fortis, CK Vaxevanis, et al. The prognostic significance of peritumoral tertiary lymphoid structures in breast cancer. Cancer Immunol Immunother. 2019;68(11):1733-1745.
87 W Xu, J Lu, WR Liu, 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.
88 Y Hayashi, T Makino, E Sato, et al. Density and maturity of peritumoral tertiary lymphoid structures in oesophageal squamous cell carcinoma predicts patient survival and response to immune checkpoint inhibitors. Br J Cancer. 2023;128(12):2175-2185.
89 K Li, Q Guo, X Zhang, et al. Oral cancer-associated tertiary lymphoid structures: gene expression profile and prognostic value. Clin Exp Immunol. 2020;199(2):172-181.
90 N Hiraoka, Y Ino, R Yamazaki-Itoh, Y Kanai, T Kosuge, K Shimada. Intratumoral tertiary lymphoid organ is a favourable prognosticator in patients with pancreatic cancer. Br J Cancer. 2015;112(11):1782-1790.
91 H Sun, Y Shi, H Ran, et al. Prognostic value of tertiary lymphoid structures (TLS) in digestive system cancers: a systematic review and meta-analysis. BMC Cancer. 2023;23(1):1248.
92 S Finkin, D Yuan, I Stein, et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat Immunol. 2015;16(12):1235-1244.
93 J Li, Y Nie, W Jia, W Wu, W Song, Y Li. 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.
94 X Wang, A Juncker-Jensen, G Huang, et al. Spatial relationship of tertiary lymphoid structures and tumor-associated neutrophils in bladder cancer and prognostic potential for anti-PD-L1 immunotherapy. Cancer Commun (Lond). 2023.
95 WH Fridman, M Meylan, G Pupier, A Calvez, I Hernandez, C Sautès-Fridman. Tertiary lymphoid structures and B cells: An intratumoral immunity cycle. Immunity. 2023;56(10):2254-2269.
96 C Germain, S Gnjatic, F Tamzalit, 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(7):832-844.
97 SL Figenschau, E Knutsen, I Urbarova, et al. ICAM1 expression is induced by proinflammatory cytokines and associated with TLS formation in aggressive breast cancer subtypes. Sci Rep. 2018;8(1):11720.
98 MB Schabath, ML Cote. Cancer progress and priorities: lung cancer. Cancer Epidemiol Biomarkers Prev. 2019;28(10):1563-1579.
99 A Giatromanolaki, P Chatzipantelis, CA Contrafouris, MI Koukourakis. Tertiary lymphoid structures, immune response, and prognostic relevance in non-small cell lung cancer. Cancer Invest. 2023;41(1):48-57.
100 M Fukuhara, S Muto, S Inomata, et al. The clinical significance of tertiary lymphoid structure and its relationship with peripheral blood characteristics in patients with surgically resected non-small cell lung cancer: a single-center, retrospective study. Cancer Immunol Immunother. 2022;71(5):1129-1137.
101 P Devi-Marulkar, S Fastenackels, P Karapentiantz, et al. Regulatory T cells infiltrate the tumor-induced tertiary lympho?d structures and are associated with poor clinical outcome in NSCLC. Commun Biol. 2022;5(1):1416.
102 A Sandstr?m Gerdtsson, M Knulst, J Botling, A Mezheyeuski, P Micke, S Ek. Phenotypic characterization of spatial immune infiltration niches in non-small cell lung cancer. Oncoimmunology. 2023;12(1):2206725.
103 X Xu, Y Gao, S Duan, et al. Clinical implications and molecular features of tertiary lymphoid structures in stage I lung adenocarcinoma. Cancer Med. 2023;12(8):9547-9558.
104 Y Wang, H Lin, N Yao, et al. Computerized tertiary lymphoid structures density on H&E-images is a prognostic biomarker in resectable lung adenocarcinoma. iScience. 2023;26(9):107635.
105 T Cascone, G Kar, JD Spicer, et al. Neoadjuvant durvalumab alone or combined with novel immuno-oncology agents in resectable lung cancer: the phase 2 NeoCOAST platform trial. Cancer Discov. 2023;13(11):2394-2411.
106 X Sun, W Liu, L Sun, 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 Immunother Cancer. 2022;10(11):e005531.
107 T Chen, Z Cao, Y Sun, et al. Neoadjuvant chemoimmunotherapy increases tumor immune lymphocytes infiltration in resectable non-small cell lung cancer. Ann Surg Oncol. 2023;30(12):7549-7560.
108 A Bessede, F Peyraud, S Le Moulec, et al. Upregulation of Indoleamine 2,3-dioxygenase 1 in tumor cells and tertiary lymphoid structures is a hallmark of inflamed non-small cell lung cancer. Clin Cancer Res. 2023;29(23):4883-4893.
109 RH Vonderheide, SM Domchek, AS Clark. Immunotherapy for breast cancer: what are we missing? Clin Cancer Res. 2017;23(11):2640-2646.
110 K Li, J Ji, S Li, et al. Analysis of the correlation and prognostic significance of tertiary lymphoid structures in breast cancer: a radiomics-clinical integration approach. J Magn Reson Imaging. 2023.
111 L Wang, S Gong, L Pang, S Zhang, X Zhang, W He. Genomic properties and clinical outcomes associated with tertiary lymphoid structures in patients with breast cancer. Sci Rep. 2023;13(1):13542.
112 AC Barb, M Pasca Fenesan, M Pirtea, et al. Tertiary lymphoid structures (TLSs) and stromal blood vessels have significant and heterogeneous impact on recurrence, lymphovascular and perineural invasion amongst breast cancer molecular subtypes. Cells. 2023;12(8):1176.
113 E Acar, G Esenda?l?, O Yaz?c?, A Dursun. Tumor-infiltrating lymphocytes (TIL), tertiary lymphoid structures (TLS), and expression of PD-1, TIM-3, LAG-3 on TIL in invasive and in situ ductal breast carcinomas and their relationship with prognostic factors. Clin Breast Cancer. 2022;22(8):e901-e915.
114 F Bertucci, A De Nonneville, P Finetti, E Mamessier. Predictive power of tertiary lymphoid structure signature for neoadjuvant chemotherapy response and immunotherapy benefit in HER2-negative breast cancer. Cancer Commun (Lond). 2023;43(8):943-946.
115 S Jiao, Q Xiong, M Yan, et al. Intratumor expanded T cell clones can be non-sentinel lymph node derived in breast cancer revealed by single-cell immune profiling. J Immunother Cancer. 2022;10(1):e003325.
116 M Gonzàlez-Farré, J Gibert, P Santiago-Díaz, et al. Characterization and spatial distribution of the immune cell infiltrate in triple-negative breast cancer: a novel classification based on plasma cells and CD8+ T cells. Hum Pathol. 2023;139:91-105.
117 Y Hou, Y Cao, L Dong, et al. Regulation of follicular T helper cell differentiation in antitumor immunity. Int J Cancer. 2023;153(2):265-277.
118 G No?l, ML Fontsa, S Garaud, et al. Functional Th1-oriented T follicular helper cells that infiltrate human breast cancer promote effective adaptive immunity. J Clin Invest. 2021;131(19):e139905.
119 O Briem, E K?llberg, S Kimbung, et al. CD169(+) macrophages in primary breast tumors associate with tertiary lymphoid structures, T(regs) and a worse prognosis for patients with advanced breast cancer. Cancers (Basel). 2023;15(4):1262.
120 Q Wang, K Sun, R Liu, 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.
121 SY Wu, SW Zhang, D Ma, et al. CCL19(+) dendritic cells potentiate clinical benefit of anti-PD-(L)1 immunotherapy in triple-negative breast cancer. Med. 2023;4(6):373-393. e8.
122 L Zeng, VCY Koh, XY Chen, PH Tan. Tertiary lymphoid structures in breast ductal carcinoma in situ correlate with adverse pathological parameters. Histopathology. 2023;82(5):779-788.
123 AM Stowman, AW Hickman, IS Mauldin, A Mahmutovic, AA Gru, CL, Slingluff Lymphoid aggregates in desmoplastic melanoma have features of tertiary lymphoid structures. Melanoma Res. 2018;28(3):237-245.
124 X Li, Z Wan, X Liu, K Ou, L Yang. A 12-chemokine gene signature is associated with the enhanced immunogram scores and is relevant for precision immunotherapy. Med Oncol. 2022;39(4):43.
125 NL Edmonds, SE Gradecki, P Katyal, et al. Tertiary lymphoid structures in desmoplastic melanoma have increased lymphocyte density, lymphocyte proliferation, and immune cross talk with tumor when compared to non-desmoplastic melanomas. Oncoimmunology. 2023;12(1):2164476.
126 T Sebestyén, A Mohos, G Liszkay, B Somlai, I Gaudi, A Ladányi. Correlation with lymphocyte infiltration, but lack of prognostic significance of MECA-79-positive high endothelial venules in primary malignant melanoma. Melanoma Res. 2018;28(4):304-310.
127 S Eschweiler, J Clarke, C Ramírez-Suástegui, et al. Intratumoral follicular regulatory T cells curtail anti-PD-1 treatment efficacy. Nat Immunol. 2021;22(8):1052-1063.
128 KT Lynch, SJ Young, MO Meneveau, et al. Heterogeneity in tertiary lymphoid structure B-cells correlates with patient survival in metastatic melanoma. J Immunother Cancer. 2021;9(6):e002273.
129 AB Rodriguez, JD Peske, AN Woods, et al. Immune mechanisms orchestrate tertiary lymphoid structures in tumors via cancer-associated fibroblasts. Cell Rep. 2021;36(3):109422.
130 EK Rainey-Barger, JM Rumble, SJ Lalor, N Esen, BM Segal, DN Irani. The lymphoid chemokine, CXCL13, is dispensable for the initial recruitment of B cells to the acutely inflamed central nervous system. Brain Behav Immun. 2011;25(5):922-931.
131 T Hoch, D Schulz, N Eling, JM Gómez, MP Levesque, B Bodenmiller. Multiplexed imaging mass cytometry of the chemokine milieus in melanoma characterizes features of the response to immunotherapy. Sci Immunol. 2022;7(70):eabk1692.
132 JY Hui. Epidemiology and Etiology of Sarcomas. Surg Clin North Am. 2016;96(5):901-914.
133 L Pekarek, B De la Torre-Escuredo, O Fraile-Martinez, et al. Towards the search for potential biomarkers in osteosarcoma: state-of-the-art and translational expectations. Int J Mol Sci. 2022;23(23):14939.
134 G Rusu-Zota, OM Manole, C Gale?, E Porumb-Andrese, O Obad?, CV Mocanu. Kaposi sarcoma, a trifecta of pathogenic mechanisms. Diagnostics (Basel). 2022;12(5):1242.
135 E Cesarman, B Damania, SE Krown, J Martin, M Bower, D Whitby. Kaposi sarcoma. Nat Rev Dis Primers. 2019;5(1):9.
136 B Gangadharan, MA Hoeve, JE Allen, et al. Murine gammaherpesvirus-induced fibrosis is associated with the development of alternatively activated macrophages. J Leukoc Biol. 2008;84(1):50-58.
137 T Horino, S Inotani, K Takamatsu, H Ohnishi, M Komori, Y Terada. Bronchus-associated lymphoid tissue (BALT) lymphoma and autoimmune disease. Qjm. 2022;115(6):405-407.
138 JR Kocks, H Adler, H Danzer, et al. Chemokine receptor CCR7 contributes to a rapid and efficient clearance of lytic murine gamma-herpes virus 68 from the lung, whereas bronchus-associated lymphoid tissue harbors virus during latency. J Immunol. 2009;182(11):6861-6869.
139 C Dahlke, K Maul, T Christalla, et al. A microRNA encoded by Kaposi sarcoma-associated herpesvirus promotes B-cell expansion in vivo. PLoS One. 2012;7(11):e49435.
140 P Pekkonen, A J?rviluoma, N Zinovkina, et al. KSHV viral cyclin interferes with T-cell development and induces lymphoma through Cdk6 and Notch activation in vivo. Cell Cycle. 2014;13(23):3670-3684.
141 B Cieniewicz, V Kirillov, I Daher, et al. IKKα-mediated noncanonical NF-κB signaling is required to support murine gammaherpesvirus 68 latency in vivo. J Virol. 2022;96(10):e0002722.
142 A Robles-Tenorio, G Solis-Ledesma. Undifferentiated pleomorphic sarcoma. StatPearls. StatPearls Publishing Copyright ? 2023, StatPearls Pusblishing LLC. 2023.
143 SP D'Angelo, MR Mahoney, BA Van Tine, et al. Nivolumab with or without ipilimumab treatment for metastatic sarcoma (Alliance A091401): two open-label, non-comparative, randomised, phase 2 trials. Lancet Oncol. 2018;19(3):416-426.
144 L Chen, T Oke, N Siegel, 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(15):4018-4030.
145 MA Cannarile, M Weisser, W Jacob, AM Jegg, CH Ries, D Rüttinger. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J Immunother Cancer. 2017;5(1):53.
146 P Tsagozis, M Augsten, Y Zhang, et al. An immunosuppressive macrophage profile attenuates the prognostic impact of CD20-positive B cells in human soft tissue sarcoma. Cancer Immunol Immunother. 2019;68(6):927-936.
147 M Li, N Che, X Liu, Y Xuan, Y Jin. Dauricine regulates prostate cancer progression by inhibiting PI3K/AKT-dependent M2 polarization of macrophages. Biochem Pharmacol. 2023;217:115838.
148 S Gordon, FO Martinez. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32(5):593-604.
149 Y Lu, G Han, Y Zhang, et al. M2 macrophage-secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinoma. Cell Commun Signal. 2023;21(1):299.
150 C Ngambenjawong, HH Gustafson, SH Pun. Progress in tumor-associated macrophage (TAM)-targeted therapeutics. Adv Drug Deliv Rev. 2017;114:206-221.
151 X Wang, J Wang, J Zhao, H Wang, J Chen, J Wu. HMGA2 facilitates colorectal cancer progression via STAT3-mediated tumor-associated macrophage recruitment. Theranostics. 2022;12(2):963-975.
152 R Duhen, C Ballesteros-Merino, AK Frye, et al. Neoadjuvant anti-OX40 (MEDI6469) therapy in patients with head and neck squamous cell carcinoma activates and expands antigen-specific tumor-infiltrating T cells. Nat Commun. 2021;12(1):1047.
153 NS Joshi, EH Akama-Garren, Y Lu, et al. Regulatory T cells in tumor-associated tertiary lymphoid structures suppress anti-tumor T cell responses. Immunity. 2015;43(3):579-590.
154 A Gronchi, P Collini, R Miceli, et al. Myogenic differentiation and histologic grading are major prognostic determinants in retroperitoneal liposarcoma. Am J Surg Pathol. 2015;39(3):383-393.
155 J Barretina, BS Taylor, S Banerji, et al. Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nat Genet. 2010;42(8):715-721.
156 WW Tseng, S Malu, M Zhang, et al. Analysis of the intratumoral adaptive immune response in well differentiated and dedifferentiated retroperitoneal liposarcoma. Sarcoma. 2015;2015:547460.
157 L Yan, Z Wang, C Cui, et al. Comprehensive immune characterization and T-cell receptor repertoire heterogeneity of retroperitoneal liposarcoma. Cancer Sci. 2019;110(10):3038-3048.
158 E Rosenbaum, CR Antonescu, S Smith, et al. Clinical, genomic, and transcriptomic correlates of response to immune checkpoint blockade-based therapy in a cohort of patients with angiosarcoma treated at a single center. J Immunother Cancer. 2022;10(4):e004149.
159 T Magara, M Nakamura, Y Nojiri, et al. Tertiary lymphoid structures correlate with better prognosis in cutaneous angiosarcoma. J Dermatol Sci. 2021;103(1):57-59.
160 MA Pantaleo, G Tarantino, C Agostinelli, et al. Immune microenvironment profiling of gastrointestinal stromal tumors (GIST) shows gene expression patterns associated to immune checkpoint inhibitors response. Oncoimmunology. 2019;8(9):e1617588.
161 M van Dongen, ND Savage, ES Jordanova, et al. Anti-inflammatory M2 type macrophages characterize metastasized and tyrosine kinase inhibitor-treated gastrointestinal stromal tumors. Int J Cancer. 2010;127(4):899-909.
162 Q Lin, P Tao, J Wang, et al. Tumor-associated tertiary lymphoid structure predicts postoperative outcomes in patients with primary gastrointestinal stromal tumors. Oncoimmunology. 2020;9(1):1747339.
163 CC Schafer, Y Wang, KP Hough, et al. Indoleamine 2,3-dioxygenase regulates anti-tumor immunity in lung cancer by metabolic reprogramming of immune cells in the tumor microenvironment. Oncotarget. 2016;7(46):75407-75424.
164 M Toulmonde, N Penel, J Adam, et al. Use of PD-1 targeting, macrophage infiltration, and IDO pathway activation in sarcomas: a phase 2 clinical trial. JAMA Oncol. 2018;4(1):93-97.
165 E Cojocaru, A Napolitano, C Fisher, P Huang, RL Jones, K. Thway What's the latest with investigational drugs for soft tissue sarcoma? Expert Opin Investig Drugs. 2022;31(11):1239-1253.
166 HA Tawbi, M Burgess, V Bolejack, et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol. 2017;18(11):1493-1501.
167 Y Tamiya, T Nakai, A Suzuki, et al. The impact of tertiary lymphoid structures on clinicopathological, genetic and gene expression characteristics in lung adenocarcinoma. Lung Cancer. 2022;174:125-132.
168 N Gavrielatou, E Fortis, A Spathis, et al. B-cell infiltration is associated with survival outcomes following programmed cell death protein 1 inhibition in head and neck squamous cell carcinoma. Ann Oncol. 2023.
169 F Schettini, M Palleschi, F Mannozzi, et al. CDK4/6-inhibitors versus chemotherapy in advanced HR+/HER2-negative breast cancer: results and correlative biomarker analyses of the KENDO randomized phase II trial. Oncologist. 2024;oyad337.
170 DT Weed, S Zilio, C McGee, et al. The tumor immune microenvironment architecture correlates with risk of recurrence in head and neck squamous cell carcinoma. Cancer Res. 2023;83(23):3886-3900.
171 X Gan, W Dong, W You, et al. Spatial multimodal analysis revealed tertiary lymphoid structures as a risk stratification indicator in combined hepatocellular-cholangiocarcinoma. Cancer Lett. 2024;581:216513.
172 Moreno Tellez C, Y Leyfman, SP D'Angelo, BA Wilky, A Dufresne. Immunotherapy in sarcoma: where do things stand? Surg Oncol Clin N Am. 2022;31(3):381-397.
173 P Darvin, SM Toor, V Sasidharan Nair, E Elkord. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50(12):1-11.
174 KJ Sweeney, MT Tetzlaff, F Vega, et al. Tertiary lymphoid structures with overlapping histopathologic features of cutaneous marginal zone lymphoma during neoadjuvant cemiplimab therapy are associated with antitumor response. J Cutan Pathol. 2021;48(5):674-679.
175 Q Zhou, L Lan, W Wang, X Xu, W Wang. Comprehensive analysis of tertiary lymphoid structures-related genes for prognostic prediction, molecular subtypes and immune infiltration in gastric cancer. Aging (Albany NY). 2023;15(22):13368-13383.
176 Q Zhang, J Bi, X Zheng, et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat Immunol. 2018;19(7):723-732.
177 D Ozmadenci, JS Shankara Narayanan, J Andrew, et al. Tumor FAK orchestrates immunosuppression in ovarian cancer via the CD155/TIGIT axis. Proc Natl Acad Sci USA. 2022;119(17):e2117065119.
178 Y Wen, F Tang, C Tu, F Hornicek, Z Duan, L Min. Immune checkpoints in osteosarcoma: recent advances and therapeutic potential. Cancer Lett. 2022;547:215887.
179 MA Shetab Boushehri, A Lamprecht. TLR4-based immunotherapeutics in cancer: a review of the achievements and shortcomings. Mol Pharm. 2018;15(11):4777-4800.
180 M Spalato-Ceruso, F Bouteiller, JP Guegan, et al. Pembrolizumab combined with low-dose cyclophosphamide and intra-tumoral injection of the toll-like receptor 4 agonist G100 in patients with advanced pretreated soft tissue sarcoma: results from the PEMBROSARC basket study. J Hematol Oncol. 2022;15(1):157.
181 Z Qi, Z Xu, L Zhang, et al. Overcoming resistance to immune checkpoint therapy in PTEN-null prostate cancer by intermittent anti-PI3Kα/β/δ treatment. Nat Commun. 2022;13(1):182.
182 KW Ng, J Boumelha, KSS Enfield, et al. Antibodies against endogenous retroviruses promote lung cancer immunotherapy. Nature. 2023;616(7957):563-573.
183 AR Elia, M Grioni, V Basso, et al. Targeting tumor vasculature with TNF leads effector t cells to the tumor and enhances therapeutic efficacy of immune checkpoint blockers in combination with adoptive cell therapy. Clin Cancer Res. 2018;24(9):2171-2181.
184 MW Rohaan, S Wilgenhof, J Haanen. Adoptive cellular therapies: the current landscape. Virchows Arch. 2019;474(4):449-461.
185 HJ Lee, YA Kim, CK Sim, et al. Expansion of tumor-infiltrating lymphocytes and their potential for application as adoptive cell transfer therapy in human breast cancer. Oncotarget. 2017;8(69):113345-113359.
186 I Poschke, M Faryna, F Bergmann, et al. Identification of a tumor-reactive T-cell repertoire in the immune infiltrate of patients with resectable pancreatic ductal adenocarcinoma. Oncoimmunology. 2016;5(12):e1240859.
187 S Takai, J Schlom, J Tucker, KY Tsang, JW Greiner. Inhibition of TGF-β1 signaling promotes central memory T cell differentiation. J Immunol. 2013;191(5):2299-2307.
188 S Feins, W Kong, EF Williams, MC Milone, JA Fraietta. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol. 2019;94(S1):S3-S9.
189 S Grote, G Ure?a-Bailén, KC Chan, et al. In vitro evaluation of CD276-CAR NK-92 functionality, migration and invasion potential in the presence of immune inhibitory factors of the tumor microenvironment. Cells. 2021;10(5):1020.
190 B Yang, Z Zhang, X Chen, et al. An Asia-specific variant of human IgG1 represses colorectal tumorigenesis by shaping the tumor microenvironment. J Clin Invest. 2022;132(6):e153454.
191 J Ye, PP Lee. B cell receptor signaling strength modulates cancer immunity. J Clin Invest. 2022;132(6):e157665.
192 A Ozaniak, J Smetanova, R Bartolini, et al. A novel anti-CD47-targeted blockade promotes immune activation in human soft tissue sarcoma but does not potentiate anti-PD-1 blockade. J Cancer Res Clin Oncol. 2022.
193 Z Strizova, J, Vachtenheim, M Snajdauf, R Lischke, J Bartunkova, D Smrz. Tumoral and paratumoral NK cells and CD8(+) T cells of esophageal carcinoma patients express high levels of CD47. Sci Rep. 2020;10(1):13936.
194 AJ Boutilier, SF Elsawa. Macrophage polarization states in the tumor microenvironment. Int J Mol Sci. 2021;22(13):6995.
195 C Thepmalee, A Panya, M Junking, T Chieochansin, PT Yenchitsomanus. Inhibition of IL-10 and TGF-β receptors on dendritic cells enhances activation of effector T-cells to kill cholangiocarcinoma cells. Hum Vaccin Immunother. 2018;14(6):1423-1431.
196 MM Melssen, KE Pollack, MO Meneveau, et al. Characterization and comparison of innate and adaptive immune responses at vaccine sites in melanoma vaccine clinical trials. Cancer Immunol Immunother. 2021;70(8):2151-2164.
197 WJ Storkus, D Maurer, Y Lin, et al. Dendritic cell vaccines targeting tumor blood vessel antigens in combination with dasatinib induce therapeutic immune responses in patients with checkpoint-refractory advanced melanoma. J Immunother Cancer. 2021;9(11):e003675.
198 MO Meneveau, P Kumar, KT Lynch, SP Patel, CL Slingluff. The vaccine-site microenvironment: impacts of antigen, adjuvant, and same-site vaccination on antigen presentation and immune signaling. J Immunother Cancer. 2022;10(3):e003533.
199 JP Girard, C Moussion, R F?rster. HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes. Nat Rev Immunol. 2012;12(11):762-773.
200 JD Peske, AB Woods, VH Engelhard. Control of CD8 T-cell infiltration into tumors by vasculature and microenvironment. Adv Cancer Res. 2015;128:263-307.
201 Y Sakai, H Hoshino, R Kitazawa, M Kobayashi. High endothelial venule-like vessels and lymphocyte recruitment in testicular seminoma. Andrology. 2014;2(2):282-289.
202 SA van de Pavert, RE Mebius. New insights into the development of lymphoid tissues. Nat Rev Immunol. 2010;10(9):664-674.
203 JD Peske, ED Thompson, L Gemta, RA Baylis, YX Fu, VH Engelhard. Effector lymphocyte-induced lymph node-like vasculature enables naive T-cell entry into tumours and enhanced anti-tumour immunity. Nat Commun. 2015;6:7114.
204 Z Zhan, L Shi-Jin, Z Yi-Ran, 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):114-126.
205 A Asrir, C Tardiveau, J Coudert, et al. Tumor-associated high endothelial venules mediate lymphocyte entry into tumors and predict response to PD-1 plus CTLA-4 combination immunotherapy. Cancer Cell. 2022;40(3):318-334. e9.
206 C Zhu, JM Kros, C Cheng, D Mustafa. The contribution of tumor-associated macrophages in glioma neo-angiogenesis and implications for anti-angiogenic strategies. Neuro Oncol. 2017;19(11):1435-1446.
207 E Allen, A Jabouille, LB Rivera, et al. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med. 2017;9(385):eaak9679.
208 Tertiary lymphoid structures validated as biomarker. Cancer Discov. 2024;14(1):OF2.
209 G Karpathiou, V Sramek, S Dagher, et al. Peripheral node addressin, a ligand for L-selectin is found in tumor cells and in high endothelial venules in endometrial cancer. Pathol Res Pract. 2022;233:153888.
210 M Dridi, L Krebs-Drouot, D Meyronet, et al. The immune microenvironment of chordomas: an immunohistochemical analysis. Cancers (Basel). 2021;13(13):3335.
211 S Fleig, T Kapanadze, J Bernier-Latmani, et al. Loss of vascular endothelial notch signaling promotes spontaneous formation of tertiary lymphoid structures. Nat Commun. 2022;13(1):2022.
212 F Jing, EY Choi. Potential of cells and cytokines/chemokines to regulate tertiary lymphoid structures in human diseases. Immune Netw. 2016;16(5):271-280.
213 CS Groeneveld, J Fontugne, L Cabel, et al. Tertiary lymphoid structures marker CXCL13 is associated with better survival for patients with advanced-stage bladder cancer treated with immunotherapy. Eur J Cancer. 2021;148:181-189.
214 AG Goubet, L Lordello, C Alves Costa Silva, et al. Escherichia coli-specific CXCL13-producing TFH are associated with clinical efficacy of neoadjuvant PD-1 Blockade against muscle-invasive bladder cancer. Cancer Discov. 2022;12(10):2280-2307.
215 S Park, H Cha, HS Kim, et al. Transcriptional upregulation of CXCL13 is correlated with a favorable response to immune checkpoint inhibitors in lung adenocarcinoma. Cancer Med. 2023;12(6):7639-7650.
216 M Ukita, J Hamanishi, H Yoshitomi, et al. CXCL13-producing CD4+ T cells accumulate in the early phase of tertiary lymphoid structures in ovarian cancer. JCI Insight. 2022;7(12):e157215.
217 X Bai, Y Zhou, Y Yokota, et al. Adaptive antitumor immune response stimulated by bio-nanoparticle based vaccine and checkpoint blockade. J Exp Clin Cancer Res. 2022;41(1):132.
218 CM Della Corte, T Sen, CM Gay, et al. STING pathway expression identifies NSCLC with an immune-responsive phenotype. J Thorac Oncol. 2020;15(5):777-791.
219 M Roulleaux Dugage, EF Nassif, A Italiano, R Bahleda. Improving immunotherapy efficacy in soft-tissue sarcomas: a biomarker driven and histotype tailored review. Front Immunol. 2021;12:775761.
220 MK Thomsen, MK Skouboe, C Boularan, et al. The cGAS-STING pathway is a therapeutic target in a preclinical model of hepatocellular carcinoma. Oncogene. 2020;39(8):1652-1664.
221 T He, Z Hao, M Lin, et al. Oncolytic adenovirus promotes vascular normalization and nonclassical tertiary lymphoid structure formation through STING-mediated DC activation. Oncoimmunology. 2022;11(1):2093054.
222 DG Hill, L Yu, H Gao, et al. Hyperactive gp130/STAT3-driven gastric tumourigenesis promotes submucosal tertiary lymphoid structure development. Int J Cancer. 2018;143(1):167-178.
223 X Yin, H Wang, R Li, et al. Tobacco exposure primes the secretion of CCL21 positively associated with tertiary lymphoid structure and response to immunotherapy. J Immunother Cancer. 2023;11(6):e006939.
224 S Nayar, E Pontarini, J Campos, et al. Immunofibroblasts regulate LTα3 expression in tertiary lymphoid structures in a pathway dependent on ICOS/ICOSL interaction. Commun Biol. 2022;5(1):413.
225 S Nayar, J Campos, MM Chung, et al. Bimodal Expansion of the Lymphatic Vessels Is Regulated by the Sequential Expression of IL-7 and Lymphotoxin α1β2 in Newly Formed Tertiary Lymphoid Structures. J Immunol. 2016;197(5):1957-1967.
226 FR Delvecchio, REA Fincham, S Spear, et al. Pancreatic cancer chemotherapy is potentiated by induction of tertiary lymphoid structures in mice. Cell Mol Gastroenterol Hepatol. 2021;12(5):1543-1565.
227 RA Chaurio, CM Anadon, T Lee Costich, 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. e9.
228 M Sylvestre, N Barbier, V Sibut, et al. KDM6B drives epigenetic reprogramming associated with lymphoid stromal cell early commitment and immune properties. Sci Adv. 2023;9(48):eadh2708.
229 K Vats, O Kruglov, B Sahoo, et al. Sensory nerves impede the formation of tertiary lymphoid structures and development of protective antimelanoma immune responses. Cancer Immunol Res. 2022;10(9):1141-1154.
230 N Doi, Y Ino, M Fuse, M Esaki, K Shimada, N Hiraoka. Correlation of vein-rich tumor microenvironment of intrahepatic cholangiocarcinoma with tertiary lymphoid structures and patient outcome. Mod Pathol. 2023;37(2):100401.
231 H Tsukamoto, Y Komohara, Y Tomita, et al. Aging-associated and CD4 T-cell-dependent ectopic CXCL13 activation predisposes to anti-PD-1 therapy-induced adverse events. Proc Natl Acad Sci USA. 2022;119(29):e2205378119.
232 L Vanhersecke, A Bougouin, A Crombé, et al. Standardized pathology screening of mature tertiary lymphoid structures in cancers. Lab Invest. 2023;103(5):100063.
233 Z Lin, L Huang, S Li, J Gu, X Cui, Y Zhou. Pan-cancer analysis of genomic properties and clinical outcome associated with tumor tertiary lymphoid structure. Sci Rep. 2020;10(1):21530.
234 P Barmpoutis, M Di Capite, H Kayhanian, et al. Tertiary lymphoid structures (TLS) identification and density assessment on H&E-stained digital slides of lung cancer. PLoS One. 2021;16(9):e0256907.
235 B Wang, L Zou, J Chen, et al. A weakly supervised segmentation network embedding cross-scale attention guidance and noise-sensitive constraint for detecting tertiary lymphoid structures of pancreatic tumors. IEEE J Biomed Health Inform. 2023.
PDF

Accesses

Citations

Detail

Sections
Recommended

/