Tertiary Lymphoid Structures: A Potential Biomarker for Anti-Cancer Therapy
Abstract
:Simple Summary
Abstract
1. Introduction
2. The Function of TLSs as Complete Structures
3. The Role of TLS Components in Tumor-Specific Immune Response
3.1. T Cells
3.2. B Cells
3.3. Dendritic Cells
3.4. High Endothelial Venules
3.5. TLS-Associated Immune Cells
4. Predictive Roles of TLSs
4.1. Surgery
4.2. Immunotherapy
5. Possible Methods in Inducing TLS Formation
5.1. Cancer Therapy
5.2. Non-Cancer Therapy
6. Discussion
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Galluzzi, L.; Humeau, J.; Buqué, A.; Zitvogel, L.; Kroemer, G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat. Rev. Clin. Oncol. 2020, 17, 725–741. [Google Scholar] [CrossRef]
- Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
- O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef]
- Kim, T.K.; Vandsemb, E.N.; Herbst, R.S.; Chen, L. Adaptive immune resistance at the tumour site: Mechanisms and therapeutic opportunities. Nat. Rev. Drug Discov. 2022, 21, 529–540. [Google Scholar] [CrossRef] [PubMed]
- Bagchi, S.; Yuan, R.; Engleman, E.G. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu. Rev. Pathol. 2021, 16, 223–249. [Google Scholar] [CrossRef] [PubMed]
- Shen, X.; Zhao, B. Efficacy of PD-1 or PD-L1 inhibitors and PD-L1 expression status in cancer: Meta-analysis. BMJ 2018, 362, k3529. [Google Scholar] [CrossRef] [Green Version]
- Vanhersecke, L.; Brunet, M.; Guégan, J.P.; Rey, C.; Bougouin, A.; Cousin, S.; Moulec, S.L.; Besse, B.; Loriot, Y.; Larroquette, M.; et al. Mature tertiary lymphoid structures predict immune checkpoint inhibitor efficacy in solid tumors independently of PD-L1 expression. Nat. Cancer 2021, 2, 794–802. [Google Scholar] [CrossRef]
- Cohen, M.; Giladi, A.; Raposo, C.; Zada, M.; Li, B.; Ruckh, J.; Deczkowska, A.; Mohar, B.; Shechter, R.; Lichtenstein, R.G.; et al. Meningeal lymphoid structures are activated under acute and chronic spinal cord pathologies. Life Sci. Alliance 2021, 4, e202000907. [Google Scholar] [CrossRef]
- Corsiero, E.; Nerviani, A.; Bombardieri, M.; Pitzalis, C. Ectopic Lymphoid Structures: Powerhouse of Autoimmunity. Front. Immunol. 2016, 7, 430. [Google Scholar] [CrossRef] [Green Version]
- Pitzalis, C.; Jones, G.W.; Bombardieri, M.; Jones, S.A. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat. Rev. Immunol. 2014, 14, 447–462. [Google Scholar] [CrossRef]
- Li, K.; Guo, Q.; Zhang, X.; Dong, X.; Liu, W.; Zhang, A.; Li, Y.; Yan, J.; Jia, G.; Zheng, Z.; et al. Oral cancer-associated tertiary lymphoid structures: Gene expression profile and prognostic value. Clin. Exp. Immunol. 2020, 199, 172–181. [Google Scholar] [CrossRef] [PubMed]
- Federico, L.; McGrail, D.J.; Bentebibel, S.E.; Haymaker, C.; Ravelli, A.; Forget, M.A.; Karpinets, T.; Jiang, P.; Reuben, A.; Negrao, M.V.; et al. Distinct tumor-infiltrating lymphocyte landscapes are associated with clinical outcomes in localized non-small-cell lung cancer. Ann. Oncol. 2022, 33, 42–56. [Google Scholar] [CrossRef] [PubMed]
- Overacre-Delgoffe, A.E.; Bumgarner, H.J.; Cillo, A.R.; Burr, A.H.P.; Tometich, J.T.; Bhattacharjee, A.; Bruno, T.C.; Vignali, D.A.A.; Hand, T.W. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity 2021, 54, 2812–2824.e2814. [Google Scholar] [CrossRef] [PubMed]
- Ukita, M.; Hamanishi, J.; Yoshitomi, H.; Yamanoi, K.; Takamatsu, S.; Ueda, A.; Suzuki, H.; Hosoe, Y.; Furutake, Y.; Taki, M.; et al. CXCL13-producing CD4+ T cells accumulate in early phase of tertiary lymphoid structures in ovarian cancer. JCI Insight 2022, 7, e157215. [Google Scholar] [CrossRef]
- Cabrita, R.; Lauss, M.; Sanna, A.; Donia, M.; Skaarup Larsen, M.; Mitra, S.; Johansson, I.; Phung, B.; Harbst, K.; Vallon-Christersson, J.; et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020, 577, 561–565. [Google Scholar] [CrossRef]
- Meylan, M.; Petitprez, F.; Becht, E.; Bougouin, A.; Pupier, G.; Calvez, A.; Giglioli, I.; Verkarre, V.; Lacroix, G.; Verneau, J.; et al. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. Immunity 2022, 55, 527–541.e525. [Google Scholar] [CrossRef]
- Zhu, G.; Nemoto, S.; Mailloux, A.W.; Perez-Villarroel, P.; Nakagawa, R.; Falahat, R.; Berglund, A.E.; Mule, J.J. Induction of Tertiary Lymphoid Structures With Antitumor Function by a Lymph Node-Derived Stromal Cell Line. Front. Immunol. 2018, 9, 1609. [Google Scholar] [CrossRef] [Green Version]
- Sautes-Fridman, C.; Petitprez, F.; Calderaro, J.; Fridman, W.H. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat. Rev. Cancer 2019, 19, 307–325. [Google Scholar] [CrossRef]
- Sofopoulos, M.; Fortis, S.P.; Vaxevanis, C.K.; Sotiriadou, N.N.; Arnogiannaki, N.; Ardavanis, A.; Vlachodimitropoulos, D.; Perez, S.A.; Baxevanis, C.N. The prognostic significance of peritumoral tertiary lymphoid structures in breast cancer. Cancer Immunol. Immunother. 2019, 68, 1733–1745. [Google Scholar] [CrossRef]
- Schumacher, T.N.; Thommen, D.S. Tertiary lymphoid structures in cancer. Science 2022, 375, eabf9419. [Google Scholar] [CrossRef]
- Jones, G.W.; Hill, D.G.; Jones, S.A. Understanding Immune Cells in Tertiary Lymphoid Organ Development: It Is All Starting to Come Together. Front. Immunol. 2016, 7, 401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mueller, C.G.; Nayar, S.; Campos, J.; Barone, F. Molecular and Cellular Requirements for the Assembly of Tertiary Lymphoid Structures. Adv. Exp. Med. Biol. 2018, 1060, 55–72. [Google Scholar] [CrossRef] [PubMed]
- Denton, A.E.; Innocentin, S.; Carr, E.J.; Bradford, B.M.; Lafouresse, F.; Mabbott, N.A.; Morbe, U.; Ludewig, B.; Groom, J.R.; Good-Jacobson, K.L.; et al. Type I interferon induces CXCL13 to support ectopic germinal center formation. J. Exp. Med. 2019, 216, 621–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van de Pavert, S.A.; Mebius, R.E. New insights into the development of lymphoid tissues. Nat. Rev. Immunol. 2010, 10, 664–674. [Google Scholar] [CrossRef] [PubMed]
- Chaurio, R.A.; Anadon, C.M.; Lee Costich, T.; Payne, K.K.; Biswas, S.; Harro, C.M.; Moran, C.; Ortiz, A.C.; Cortina, C.; Rigolizzo, K.E.; et al. TGF-beta-mediated silencing of genomic organizer SATB1 promotes Tfh cell differentiation and formation of intra-tumoral tertiary lymphoid structures. Immunity 2022, 55, 115–128.e119. [Google Scholar] [CrossRef]
- Hiraoka, N.; Ino, Y.; Yamazaki-Itoh, R.; Kanai, Y.; Kosuge, T.; Shimada, K. Intratumoral tertiary lymphoid organ is a favourable prognosticator in patients with pancreatic cancer. Br. J. Cancer 2015, 112, 1782–1790. [Google Scholar] [CrossRef] [Green Version]
- Silina, K.; Soltermann, A.; Attar, F.M.; Casanova, R.; Uckeley, Z.M.; Thut, H.; Wandres, M.; Isajevs, S.; Cheng, P.; Curioni-Fontecedro, A.; et al. Germinal Centers Determine the Prognostic Relevance of Tertiary Lymphoid Structures and Are Impaired by Corticosteroids in Lung Squamous Cell Carcinoma. Cancer Res. 2018, 78, 1308–1320. [Google Scholar] [CrossRef] [Green Version]
- Gunderson, A.J.; Rajamanickam, V.; Bui, C.; Bernard, B.; Pucilowska, J.; Ballesteros-Merino, C.; Schmidt, M.; McCarty, K.; Philips, M.; Piening, B.; 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, 1900635. [Google Scholar] [CrossRef]
- Posch, F.; Silina, K.; Leibl, S.; Mundlein, A.; Moch, H.; Siebenhuner, A.; Samaras, P.; Riedl, J.; Stotz, M.; Szkandera, J.; et al. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. Oncoimmunology 2018, 7, e1378844. [Google Scholar] [CrossRef] [Green Version]
- Li, Q.; Liu, X.; Wang, D.; Wang, Y.; Lu, H.; Wen, S.; Fang, J.; Cheng, B.; Wang, Z. Prognostic value of tertiary lymphoid structure and tumour infiltrating lymphocytes in oral squamous cell carcinoma. Int. J. Oral Sci. 2020, 12, 24. [Google Scholar] [CrossRef]
- Tang, J.; Ramis-Cabrer, D.; Curull, V.; Wang, X.; Mateu-Jiménez, M.; Pijuan, L.; Duran, X.; Qin, L.; Rodríguez-Fuster, A.; Aguiló, R.; et al. B Cells and Tertiary Lymphoid Structures Influence Survival in Lung Cancer Patients with Resectable Tumors. Cancers 2020, 12, 2644. [Google Scholar] [CrossRef] [PubMed]
- Kiessler, M.; Plesca, I.; Sommer, U.; Wehner, R.; Wilczkowski, F.; Muller, L.; Tunger, A.; Lai, X.; Rentsch, A.; Peuker, K.; et al. Tumor-infiltrating plasmacytoid dendritic cells are associated with survival in human colon cancer. J. Immunother. Cancer 2021, 9, e001813. [Google Scholar] [CrossRef] [PubMed]
- Castino, G.F.; Cortese, N.; Capretti, G.; Serio, S.; Di Caro, G.; Mineri, R.; Magrini, E.; Grizzi, F.; Cappello, P.; Novelli, F.; et al. Spatial distribution of B cells predicts prognosis in human pancreatic adenocarcinoma. Oncoimmunology 2016, 5, e1085147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, B.; Liu, J.; Han, Y.; Deng, Y.; Li, J.; Jiang, Y. The Presence of Tertiary Lymphoid Structures Provides New Insight Into the Clinicopathological Features and Prognosis of Patients With Breast Cancer. Front. Immunol. 2022, 13, 868155. [Google Scholar] [CrossRef]
- Zhang, W.-H.; Wang, W.-Q.; Han, X.; Gao, H.-L.; Xu, S.-S.; Li, S.; Li, T.-J.; Xu, H.-X.; Li, H.; Ye, L.-Y.; et al. Infiltrating pattern and prognostic value of tertiary lymphoid structures in resected non-functional pancreatic neuroendocrine tumors. J. Immunother. Cancer 2020, 8, e001188. [Google Scholar] [CrossRef]
- Wen, S.; Chen, Y.; Hu, C.; Du, X.; Xia, J.; Wang, X.; Zhu, W.; Wang, Q.; Zhu, M.; Chen, Y.; et al. Combination of Tertiary Lymphoid Structure and Neutrophil-to-Lymphocyte Ratio Predicts Survival in Patients With Hepatocellular Carcinoma. Front. Immunol. 2021, 12, 788640. [Google Scholar] [CrossRef]
- Buisseret, L.; Garaud, S.; de Wind, A.; Van den Eynden, G.; Boisson, A.; Solinas, C.; Gu-Trantien, C.; Naveaux, C.; Lodewyckx, J.N.; Duvillier, H.; et al. Tumor-infiltrating lymphocyte composition, organization and PD-1/ PD-L1 expression are linked in breast cancer. Oncoimmunology 2017, 6, e1257452. [Google Scholar] [CrossRef]
- Zhao, Z.; Ding, H.; Lin, Z.B.; Qiu, S.H.; Zhang, Y.R.; Guo, Y.G.; Chu, X.D.; Sam, L.I.; Pan, J.H.; Pan, Y.L. Relationship between Tertiary Lymphoid Structure and the Prognosis and Clinicopathologic Characteristics in Solid Tumors. Int. J. Med. Sci. 2021, 18, 2327–2338. [Google Scholar] [CrossRef]
- Zhou, X.; Li, W.; Yang, J.; Qi, X.; Chen, Y.; Yang, H.; Chu, L. Tertiary lymphoid structure stratifies glioma into three distinct tumor subtypes. Aging 2021, 13, 26063–26094. [Google Scholar] [CrossRef]
- Ahmed, A.; Köhler, S.; Klotz, R.; Giese, N.; Hackert, T.; Springfeld, C.; Zörnig, I.; Jäger, D.; Halama, N. Tertiary lymphoid structures and their association to immune phenotypes and circulatory IL2 levels in pancreatic ductal adenocarcinoma. OncoImmunology 2022, 11, 2027148. [Google Scholar] [CrossRef]
- Maoz, A.; Dennis, M.; Greenson, J.K. The Crohn’s-Like Lymphoid Reaction to Colorectal Cancer-Tertiary Lymphoid Structures With Immunologic and Potentially Therapeutic Relevance in Colorectal Cancer. Front. Immunol. 2019, 10, 1884. [Google Scholar] [CrossRef] [Green Version]
- Wirsing, A.M.; Ervik, I.K.; Seppola, M.; Uhlin-Hansen, L.; Steigen, S.E.; Hadler-Olsen, E. Presence of high-endothelial venules correlates with a favorable immune microenvironment in oral squamous cell carcinoma. Mod. Pathol. 2018, 31, 910–922. [Google Scholar] [CrossRef] [Green Version]
- Messina, J.L.; Fenstermacher, D.A.; Eschrich, S.; Qu, X.; Berglund, A.E.; Lloyd, M.C.; Schell, M.J.; Sondak, V.K.; Weber, J.S.; Mulé, J.J. 12-Chemokine gene signature identifies lymph node-like structures in melanoma: Potential for patient selection for immunotherapy? Sci. Rep. 2012, 2, 765. [Google Scholar] [CrossRef] [Green Version]
- Truxova, I.; Kasikova, L.; Hensler, M.; Skapa, P.; Laco, J.; Pecen, L.; Belicova, L.; Praznovec, I.; Halaska, M.J.; Brtnicky, T.; et al. Mature dendritic cells correlate with favorable immune infiltrate and improved prognosis in ovarian carcinoma patients. J. Immunother. Cancer 2018, 6, 139. [Google Scholar] [CrossRef]
- Dieu-Nosjean, M.C.; Antoine, M.; Danel, C.; Heudes, D.; Wislez, M.; Poulot, V.; Rabbe, N.; Laurans, L.; Tartour, E.; de Chaisemartin, L.; et al. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J. Clin. Oncol. 2008, 26, 4410–4417. [Google Scholar] [CrossRef] [Green Version]
- Calderaro, J.; Petitprez, F.; Becht, E.; Laurent, A.; Hirsch, T.Z.; Rousseau, B.; Luciani, A.; Amaddeo, G.; Derman, J.; Charpy, C.; et al. Intra-tumoral tertiary lymphoid structures are associated with a low risk of early recurrence of hepatocellular carcinoma. J. Hepatol. 2019, 70, 58–65. [Google Scholar] [CrossRef]
- Ding, G.Y.; Ma, J.Q.; Yun, J.P.; Chen, X.; Ling, Y.; Zhang, S.; Shi, J.Y.; Chang, Y.Q.; Ji, Y.; Wang, X.Y.; et al. Distribution and density of tertiary lymphoid structures predict clinical outcome in intrahepatic cholangiocarcinoma. J. Hepatol. 2022, 76, 608–618. [Google Scholar] [CrossRef]
- Fridman, W.H.; Meylan, M.; Petitprez, F.; Sun, C.M.; Italiano, A.; Sautes-Fridman, C. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat. Rev. Clin. Oncol. 2022, 19, 441–457. [Google Scholar] [CrossRef]
- Matsubara, S.; Seki, M.; Suzuki, S.; Komori, T.; Takamori, M. Tertiary lymphoid organs in the inflammatory myopathy associated with PD-1 inhibitors. J. Immunother. Cancer 2019, 7, 256. [Google Scholar] [CrossRef]
- Yamaguchi, K.; Ito, M.; Ohmura, H.; Hanamura, F.; Nakano, M.; Tsuchihashi, K.; Nagai, S.; Ariyama, H.; Kusaba, H.; Yamamoto, H.; et al. Helper T cell-dominant tertiary lymphoid structures are associated with disease relapse of advanced colorectal cancer. Oncoimmunology 2020, 9, 1724763. [Google Scholar] [CrossRef]
- Hennequin, A.; Derangere, V.; Boidot, R.; Apetoh, L.; Vincent, J.; Orry, D.; Fraisse, J.; Causeret, S.; Martin, F.; Arnould, L.; et al. Tumor infiltration by Tbet+ effector T cells and CD20+ B cells is associated with survival in gastric cancer patients. Oncoimmunology 2016, 5, e1054598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.P.; Wu, C.Y.; Chen, M.Y.; Liu, S.X.; Yan, S.M.; Kang, Y.F.; Sun, C.; Grandis, J.R.; Zeng, M.S.; Zhong, Q. PD-1(+)CXCR5(-)CD4(+) Th-CXCL13 cell subset drives B cells into tertiary lymphoid structures of nasopharyngeal carcinoma. J. Immunother. Cancer 2021, 9, e002101. [Google Scholar] [CrossRef] [PubMed]
- Johnston, R.J.; Poholek, A.C.; DiToro, D.; Yusuf, I.; Eto, D.; Barnett, B.; Dent, A.L.; Craft, J.; Crotty, S. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science 2009, 325, 1006–1010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ruffell, B.; DeNardo, D.G.; Affara, N.I.; Coussens, L.M. Lymphocytes in cancer development: Polarization towards pro-tumor immunity. Cytokine Growth Factor Rev. 2010, 21, 3–10. [Google Scholar] [CrossRef] [Green Version]
- Schreiber, S.; Hammers, C.M.; Kaasch, A.J.; Schraven, B.; Dudeck, A.; Kahlfuss, S. Metabolic Interdependency of Th2 Cell-Mediated Type 2 Immunity and the Tumor Microenvironment. Front. Immunol. 2021, 12, 632581. [Google Scholar] [CrossRef]
- Gu-Trantien, C.; Loi, S.; Garaud, S.; Equeter, C.; Libin, M.; de Wind, A.; Ravoet, M.; Le Buanec, H.; Sibille, C.; Manfouo-Foutsop, G.; et al. CD4⁺ follicular helper T cell infiltration predicts breast cancer survival. J. Clin. Investig. 2013, 123, 2873–2892. [Google Scholar] [CrossRef] [Green Version]
- Noel, G.; Fontsa, M.L.; Garaud, S.; De Silva, P.; de Wind, A.; Van den Eynden, G.G.; Salgado, R.; Boisson, A.; Locy, H.; Thomas, N.; et al. Functional Th1-oriented T follicular helper cells that infiltrate human breast cancer promote effective adaptive immunity. J. Clin. Investig. 2021, 131, e139905. [Google Scholar] [CrossRef]
- Jia, Y.; Zeng, Z.; Li, Y.; Li, Z.; Jin, L.; Zhang, Z.; Wang, L.; Wang, F.S. Impaired function of CD4+ T follicular helper (Tfh) cells associated with hepatocellular carcinoma progression. PLoS ONE 2015, 10, e0117458. [Google Scholar] [CrossRef]
- Ng, K.W.; Marshall, E.A.; Enfield, K.S.; Martin, S.D.; Milne, K.; Pewarchuk, M.E.; Abraham, N.; Lam, W.L. Somatic mutation-associated T follicular helper cell elevation in lung adenocarcinoma. Oncoimmunology 2018, 7, e1504728. [Google Scholar] [CrossRef]
- Tian, C.; Li, C.; Zeng, Y.; Liang, J.; Yang, Q.; Gu, F.; Hu, Y.; Liu, L. Identification of CXCL13/CXCR5 axis’s crucial and complex effect in human lung adenocarcinoma. Int. Immunopharmacol. 2021, 94, 107416. [Google Scholar] [CrossRef]
- Groeneveld, C.S.; Fontugne, J.; Cabel, L.; Bernard-Pierrot, I.; Radvanyi, F.; Allory, Y.; de Reynies, A. 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. [Google Scholar] [CrossRef] [PubMed]
- Qian, G.; Wu, M.; Zhao, Y.; Li, Q.; Zhang, M.; Cai, C.; Tong, D. Thyroid cancer metastasis is associated with an overabundance of defective follicular helper T cells. APMIS 2020, 128, 487–496. [Google Scholar] [CrossRef] [PubMed]
- Rubio, A.J.; Porter, T.; Zhong, X. Duality of B Cell-CXCL13 Axis in Tumor Immunology. Front. Immunol. 2020, 11, 521110. [Google Scholar] [CrossRef] [PubMed]
- Fonseca, V.R.; Ribeiro, F.; Graca, L. T follicular regulatory (Tfr) cells: Dissecting the complexity of Tfr-cell compartments. Immunol. Rev. 2019, 288, 112–127. [Google Scholar] [CrossRef] [PubMed]
- Baumjohann, D.; Brossart, P. T follicular helper cells: Linking cancer immunotherapy and immune-related adverse events. J. Immunother. Cancer 2021, 9, e002588. [Google Scholar] [CrossRef]
- Joshi, N.S.; Akama-Garren, E.H.; Lu, Y.; Lee, D.Y.; Chang, G.P.; Li, A.; DuPage, M.; Tammela, T.; Kerper, N.R.; Farago, A.F.; et al. Regulatory T Cells in Tumor-Associated Tertiary Lymphoid Structures Suppress Anti-tumor T Cell Responses. Immunity 2015, 43, 579–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Freier, C.P.; Kuhn, C.; Endres, S.; Mayr, D.; Friese, K.; Jeschke, U.; Anz, D. FOXP3+ Cells Recruited by CCL22 into Breast Cancer Correlates with Less Tumor Nodal Infiltration. Anticancer Res. 2016, 36, 3139–3145. [Google Scholar]
- Whiteside, T.L. FOXP3+ Treg as a therapeutic target for promoting anti-tumor immunity. Expert Opin. Ther. Targets 2018, 22, 353–363. [Google Scholar] [CrossRef]
- Wang, J.; Jiang, D.; Zheng, X.; Li, W.; Zhao, T.; Wang, D.; Yu, H.; Sun, D.; Li, Z.; Zhang, J.; et al. Tertiary lymphoid structure and decreased CD8+ T cell infiltration in minimally invasive adenocarcinoma. iScience 2022, 25, 103883. [Google Scholar] [CrossRef]
- Goc, J.; Germain, C.; Vo-Bourgais, T.K.; Lupo, A.; Klein, C.; Knockaert, S.; de Chaisemartin, L.; Ouakrim, H.; Becht, E.; Alifano, M.; et al. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res. 2014, 74, 705–715. [Google Scholar] [CrossRef] [Green Version]
- Dai, S.; Zeng, H.; Liu, Z.; Jin, K.; Jiang, W.; Wang, Z.; Lin, Z.; Xiong, Y.; Wang, J.; Chang, Y.; 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, e001823. [Google Scholar] [CrossRef]
- Fridman, W.H.; Petitprez, F.; Meylan, M.; Chen, T.W.; Sun, C.M.; Roumenina, L.T.; Sautes-Fridman, C. B cells and cancer: To B or not to B? J. Exp. Med. 2021, 218, e20200851. [Google Scholar] [CrossRef]
- Roumenina, L.T.; Daugan, M.V.; Noé, R.; Petitprez, F.; Vano, Y.A.; Sanchez-Salas, R.; Becht, E.; Meilleroux, J.; Clec’h, B.L.; Giraldo, N.A.; et al. Tumor Cells Hijack Macrophage-Produced Complement C1q to Promote Tumor Growth. Cancer Immunol. Res. 2019, 7, 1091–1105. [Google Scholar] [CrossRef]
- Tan, T.T.; Coussens, L.M. Humoral immunity, inflammation and cancer. Curr. Opin. Immunol. 2007, 19, 209–216. [Google Scholar] [CrossRef] [PubMed]
- Shen, P.; Fillatreau, S. Antibody-independent functions of B cells: A focus on cytokines. Nat. Rev. Immunol. 2015, 15, 441–451. [Google Scholar] [CrossRef]
- Kim, S.S.; Shen, S.; Miyauchi, S.; Sanders, P.D.; Franiak-Pietryga, I.; Mell, L.; Gutkind, J.S.; Cohen, E.E.W.; Califano, J.A.; Sharabi, A.B. B Cells Improve Overall Survival in HPV-Associated Squamous Cell Carcinomas and Are Activated by Radiation and PD-1 Blockade. Clin. Cancer Res. 2020, 26, 3345–3359. [Google Scholar] [CrossRef] [Green Version]
- Edin, S.; Kaprio, T.; Hagström, J.; Larsson, P.; Mustonen, H.; Böckelman, C.; Strigård, K.; Gunnarsson, U.; Haglund, C.; Palmqvist, R. The Prognostic Importance of CD20(+) B lymphocytes in Colorectal Cancer and the Relation to Other Immune Cell subsets. Sci. Rep. 2019, 9, 19997. [Google Scholar] [CrossRef] [Green Version]
- Phanthunane, C.; Wijers, R.; de Herdt, M.; Langeveld, T.P.M.; Koljenovic, S.; Dasgupta, S.; Sleijfer, S.; Baatenburg de Jong, R.J.; Hardillo, J.; Balcioglu, H.E.; et al. B-cell clusters at the invasive margin associate with longer survival in early-stage oral-tongue cancer patients. Oncoimmunology 2021, 10, 1882743. [Google Scholar] [CrossRef] [PubMed]
- Petitprez, F.; de Reyniès, A.; Keung, E.Z.; Chen, T.W.; Sun, C.M.; Calderaro, J.; Jeng, Y.M.; Hsiao, L.P.; Lacroix, L.; Bougoüin, A.; et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 2020, 577, 556–560. [Google Scholar] [CrossRef] [PubMed]
- Cillo, A.R.; Kurten, C.H.L.; Tabib, T.; Qi, Z.; Onkar, S.; Wang, T.; Liu, A.; Duvvuri, U.; Kim, S.; Soose, R.J.; et al. Immune Landscape of Viral- and Carcinogen-Driven Head and Neck Cancer. Immunity 2020, 52, 183–199.e189. [Google Scholar] [CrossRef] [PubMed]
- Bruno, T.C.; Ebner, P.J.; Moore, B.L.; Squalls, O.G.; Waugh, K.A.; Eruslanov, E.B.; Singhal, S.; Mitchell, J.D.; Franklin, W.A.; Merrick, D.T.; et al. Antigen-Presenting Intratumoral B Cells Affect CD4(+) TIL Phenotypes in Non-Small Cell Lung Cancer Patients. Cancer Immunol. Res. 2017, 5, 898–907. [Google Scholar] [CrossRef]
- Garaud, S.; Buisseret, L.; Solinas, C.; Gu-Trantien, C.; de Wind, A.; Van den Eynden, G.; Naveaux, C.; Lodewyckx, J.N.; Boisson, A.; Duvillier, H.; et al. Tumor infiltrating B-cells signal functional humoral immune responses in breast cancer. JCI Insight 2019, 5, e129641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harvey, B.P.; Raycroft, M.T.; Quan, T.E.; Rudenga, B.J.; Roman, R.M.; Craft, J.; Mamula, M.J. Transfer of antigen from human B cells to dendritic cells. Mol. Immunol. 2014, 58, 56–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heesters, B.A.; van der Poel, C.E.; Das, A.; Carroll, M.C. Antigen Presentation to B Cells. Trends Immunol. 2016, 37, 844–854. [Google Scholar] [CrossRef] [PubMed]
- Lynch, K.T.; Young, S.J.; Meneveau, M.O.; Wages, N.A.; Engelhard, V.H.; Slingluff, C.L., Jr.; Mauldin, I.S. Heterogeneity in tertiary lymphoid structure B-cells correlates with patient survival in metastatic melanoma. J. Immunother. Cancer 2021, 9, e002273. [Google Scholar] [CrossRef]
- Cogne, M. Activation-induced deaminase in B lymphocyte maturation and beyond. Biomed. J. 2013, 36, 259–268. [Google Scholar] [CrossRef]
- Finkin, S.; Yuan, D.; Stein, I.; Taniguchi, K.; Weber, A.; Unger, K.; Browning, J.L.; Goossens, N.; Nakagawa, S.; Gunasekaran, G.; et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat. Immunol. 2015, 16, 1235–1244. [Google Scholar] [CrossRef]
- Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef]
- Segura, E.; Touzot, M.; Bohineust, A.; Cappuccio, A.; Chiocchia, G.; Hosmalin, A.; Dalod, M.; Soumelis, V.; Amigorena, S. Human inflammatory dendritic cells induce Th17 cell differentiation. Immunity 2013, 38, 336–348. [Google Scholar] [CrossRef] [Green Version]
- KleinJan, A.; van Nimwegen, M.; Leman, K.; Wen, K.X.; Boon, L.; Hendriks, R.W. Involvement of Dendritic Cells and Th17 Cells in Induced Tertiary Lymphoid Structures in a Chronic Beryllium Disease Mouse Model. Mediat. Inflamm. 2021, 2021, 8845966. [Google Scholar] [CrossRef]
- Ji, A.L.; Rubin, A.J.; Thrane, K.; Jiang, S.; Reynolds, D.L.; Meyers, R.M.; Guo, M.G.; George, B.M.; Mollbrink, A.; Bergenstråhle, J.; et al. Multimodal Analysis of Composition and Spatial Architecture in Human Squamous Cell Carcinoma. Cell 2020, 182, 497–514.e422. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Li, Z.; Skrzypczynska, K.M.; Fang, Q.; Zhang, W.; O’Brien, S.A.; He, Y.; Wang, L.; Zhang, Q.; Kim, A.; et al. Single-Cell Analyses Inform Mechanisms of Myeloid-Targeted Therapies in Colon Cancer. Cell 2020, 181, 442–459.e429. [Google Scholar] [CrossRef] [PubMed]
- Blanchard, L.; Girard, J.P. High endothelial venules (HEVs) in immunity, inflammation and cancer. Angiogenesis 2021, 24, 719–753. [Google Scholar] [CrossRef] [PubMed]
- Weinstein, A.M.; Giraldo, N.A.; Petitprez, F.; Julie, C.; Lacroix, L.; Peschaud, F.; Emile, J.F.; Marisa, L.; Fridman, W.H.; Storkus, W.J.; et al. Association of IL-36gamma with tertiary lymphoid structures and inflammatory immune infiltrates in human colorectal cancer. Cancer Immunol. Immunother. 2019, 68, 109–120. [Google Scholar] [CrossRef]
- Song, I.H.; Heo, S.H.; Bang, W.S.; Park, H.S.; Park, I.A.; Kim, Y.A.; Park, S.Y.; Roh, J.; Gong, G.; Lee, H.J. Predictive Value of Tertiary Lymphoid Structures Assessed by High Endothelial Venule Counts in the Neoadjuvant Setting of Triple-Negative Breast Cancer. Cancer Res. Treat. 2017, 49, 399–407. [Google Scholar] [CrossRef] [Green Version]
- Garcia-Hernandez, M.L.; Uribe-Uribe, N.O.; Espinosa-Gonzalez, R.; Kast, W.M.; Khader, S.A.; Rangel-Moreno, J. A Unique Cellular and Molecular Microenvironment Is Present in Tertiary Lymphoid Organs of Patients with Spontaneous Prostate Cancer Regression. Front. Immunol. 2017, 8, 563. [Google Scholar] [CrossRef] [Green Version]
- Nayar, S.; Campos, J.; Smith, C.G.; Iannizzotto, V.; Gardner, D.H.; Mourcin, F.; Roulois, D.; Turner, J.; Sylvestre, M.; Asam, S.; et al. Immunofibroblasts are pivotal drivers of tertiary lymphoid structure formation and local pathology. Proc. Natl. Acad. Sci. USA 2019, 116, 13490–13497. [Google Scholar] [CrossRef] [Green Version]
- Rodriguez, A.B.; Peske, J.D.; Woods, A.N.; Leick, K.M.; Mauldin, I.S.; Meneveau, M.O.; Young, S.J.; Lindsay, R.S.; Melssen, M.M.; Cyranowski, S.; et al. Immune mechanisms orchestrate tertiary lymphoid structures in tumors via cancer-associated fibroblasts. Cell Rep. 2021, 36, 109422. [Google Scholar] [CrossRef]
- Asam, S.; Neag, G.; Berardicurti, O.; Gardner, D.; Barone, F. The role of stroma and epithelial cells in primary Sjögren’s syndrome. Rheumatology 2019, 60, 3503–3512. [Google Scholar] [CrossRef]
- Asam, S.; Nayar, S.; Gardner, D.; Barone, F. Stromal cells in tertiary lymphoid structures: Architects of autoimmunity. Immunol. Rev. 2021, 302, 184–195. [Google Scholar] [CrossRef]
- Kroeger, D.R.; Milne, K.; Nelson, B.H. Tumor-Infiltrating Plasma Cells Are Associated with Tertiary Lymphoid Structures, Cytolytic T-Cell Responses, and Superior Prognosis in Ovarian Cancer. Clin Cancer Res 2016, 22, 3005–3015. [Google Scholar] [CrossRef] [PubMed]
- Duan, L.; Liu, D.; Chen, H.; Mintz, M.A.; Chou, M.Y.; Kotov, D.I.; Xu, Y.; An, J.; Laidlaw, B.J.; Cyster, J.G. Follicular dendritic cells restrict interleukin-4 availability in germinal centers and foster memory B cell generation. Immunity 2021, 54, 2256–2272.e2256. [Google Scholar] [CrossRef] [PubMed]
- van der Poel, C.E.; Bajic, G.; Macaulay, C.W.; van den Broek, T.; Ellson, C.D.; Bouma, G.; Victora, G.D.; Degn, S.E.; Carroll, M.C. Follicular Dendritic Cells Modulate Germinal Center B Cell Diversity through FcγRIIB. Cell Rep. 2019, 29, 2745–2755.e1744. [Google Scholar] [CrossRef] [PubMed]
- Neagu, M.; Albulescu, R.; Tanase, C. Distinct soluble EGFR isoforms expressed in non-small-cell lung cancer cells compared with normal tissue. Biomark. Med. 2012, 6, 198–199. [Google Scholar] [CrossRef] [PubMed]
- Su, S.; Zhao, J.; Xing, Y.; Zhang, X.; Liu, J.; Ouyang, Q.; Chen, J.; Su, F.; Liu, Q.; Song, E. Immune Checkpoint Inhibition Overcomes ADCP-Induced Immunosuppression by Macrophages. Cell 2018, 175, 442–457.e423. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.; Oke, T.; Siegel, N.; Cojocaru, G.; Tam, A.J.; Blosser, R.L.; Swailes, J.; Ligon, J.A.; Lebid, A.; Morris, C.; et al. The Immunosuppressive Niche of Soft-Tissue Sarcomas is Sustained by Tumor-Associated Macrophages and Characterized by Intratumoral Tertiary Lymphoid Structures. Clin. Cancer Res. 2020, 26, 4018–4030. [Google Scholar] [CrossRef] [Green Version]
- Morcrette, G.; Hirsch, T.Z.; Badour, E.; Pilet, J.; Caruso, S.; Calderaro, J.; Martin, Y.; Imbeaud, S.; Letouzé, E.; Rebouissou, S.; et al. APC germline hepatoblastomas demonstrate cisplatin-induced intratumor tertiary lymphoid structures. Oncoimmunology 2019, 8, e1583547. [Google Scholar] [CrossRef] [Green Version]
- Remark, R.; Lupo, A.; Alifano, M.; Biton, J.; Ouakrim, H.; Stefani, A.; Cremer, I.; Goc, J.; Régnard, J.; Dieu-Nosjean, M.; et al. Immune contexture and histological response after neoadjuvant chemotherapy predict clinical outcome of lung cancer patients. Oncoimmunology 2016, 5, e1255394. [Google Scholar] [CrossRef] [Green Version]
- van Dijk, N.; Gil-Jimenez, A.; Silina, K.; Hendricksen, K.; Smit, L.A.; de Feijter, J.M.; van Montfoort, M.L.; van Rooijen, C.; Peters, D.; Broeks, A.; et al. Preoperative ipilimumab plus nivolumab in locoregionally advanced urothelial cancer: The NABUCCO trial. Nat. Med. 2020, 26, 1839–1844. [Google Scholar] [CrossRef]
- Helmink, B.; Reddy, S.; Gao, J.; Zhang, S.; Basar, R.; Thakur, R.; Yizhak, K.; Sade-Feldman, M.; Blando, J.; Han, G.; et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020, 577, 549–555. [Google Scholar] [CrossRef]
- Maldonado, L.; Teague, J.E.; Morrow, M.P.; Jotova, I.; Wu, T.C.; Wang, C.; Desmarais, C.; Boyer, J.D.; Tycko, B.; Robins, H.S.; et al. Intramuscular therapeutic vaccination targeting HPV16 induces T cell responses that localize in mucosal lesions. Sci. Transl. Med. 2014, 6, 221ra213. [Google Scholar] [CrossRef] [PubMed]
- Lutz, E.; Wu, A.; Bigelow, E.; Sharma, R.; Mo, G.; Soares, K.; Solt, S.; Dorman, A.; Wamwea, A.; Yager, A.; et al. Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol. Res. 2014, 2, 616–631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cottrell, T.R.; Thompson, E.D.; Forde, P.M.; Stein, J.E.; Duffield, A.S.; Anagnostou, V.; Rekhtman, N.; Anders, R.A.; Cuda, J.D.; Illei, P.B.; et al. Pathologic features of response to neoadjuvant anti-PD-1 in resected non-small-cell lung carcinoma: A proposal for quantitative immune-related pathologic response criteria (irPRC). Ann. Oncol. 2018, 29, 1853–1860. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.; Kim, J.; Park, I.; Song, I.; Yu, J.; Ahn, J.; Gong, G. Prognostic Significance of Tumor-Infiltrating Lymphocytes and the Tertiary Lymphoid Structures in HER2-Positive Breast Cancer Treated With Adjuvant Trastuzumab. Am. J. Clin. Pathol. 2015, 144, 278–288. [Google Scholar] [CrossRef] [Green Version]
- Prabhakaran, S.; Rizk, V.; Ma, Z.; Cheng, C.; Berglund, A.; Coppola, D.; Khalil, F.; Mulé, J.; Soliman, H. Evaluation of invasive breast cancer samples using a 12-chemokine gene expression score: Correlation with clinical outcomes. Breast Cancer Res. 2017, 19, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Johansson-Percival, A.; He, B.; Li, Z.; Kjellén, A.; Russell, K.; Li, J.; Larma, I.; Ganss, R. De novo induction of intratumoral lymphoid structures and vessel normalization enhances immunotherapy in resistant tumors. Nat. Immunol. 2017, 18, 1207–1217. [Google Scholar] [CrossRef]
- Zhang, Y.; Xu, J.; Fei, Z.; Dai, H.; Fan, Q.; Yang, Q.; Chen, Y.; Wang, B.; Wang, C. 3D Printing Scaffold Vaccine for Antitumor Immunity. Adv. Mater. 2021, 33, e2106768. [Google Scholar] [CrossRef]
- van Hooren, L.; Vaccaro, A.; Ramachandran, M.; Vazaios, K.; Libard, S.; van de Walle, T.; Georganaki, M.; Huang, H.; Pietila, I.; Lau, J.; et al. Agonistic CD40 therapy induces tertiary lymphoid structures but impairs responses to checkpoint blockade in glioma. Nat. Commun. 2021, 12, 4127. [Google Scholar] [CrossRef]
- Munoz-Erazo, L.; Rhodes, J.; Marion, V.; Kemp, R. Tertiary lymphoid structures in cancer-considerations for patient prognosis. Cell. Mol. Immunol. 2020, 17, 570–575. [Google Scholar] [CrossRef]
- Schweiger, T.; Berghoff, A.; Glogner, C.; Glueck, O.; Rajky, O.; Traxler, D.; Birner, P.; Preusser, M.; Klepetko, W.; Hoetzenecker, K. Tumor-infiltrating lymphocyte subsets and tertiary lymphoid structures in pulmonary metastases from colorectal cancer. Clin. Exp. Metastasis 2016, 33, 727–739. [Google Scholar] [CrossRef] [Green Version]
- Kang, J.; Zhang, C.; Zhong, W. Neoadjuvant immunotherapy for non-small cell lung cancer: State of the art. Cancer Commun. 2021, 41, 287–302. [Google Scholar] [CrossRef] [PubMed]
Tumor | Number of Patients | Identification Method | Predictive Roles |
---|---|---|---|
lung cancer [45] | 74 | H&E staining, immunohistochemistry | DFS |
hepatocellular carcinoma [46] | 273 | H&E staining, immunohistochemistry | early relapse |
pancreatic neuroendocrine tumor [35] | 182 | H&E staining, immunohistochemistry, immunofluorescence | DFS OS |
ovarian carcinoma [44] | 167 | H&E staining, immunohistochemistry, RNA-seq, flow cytometry | OS |
Tumor | Number of Patients | Identification Method | Predictive Roles |
---|---|---|---|
solid tumor [7] | 328 | H&E staining, immunohistochemistry, immunofluorescence | DFS OS |
melanoma renal cell carcinoma [110] | melanoma 20 | H&E staining, immunohistochemistry, immunofluorescence, RNA-seq, flow cytometry | ORR |
renal cell carcinoma 14 | |||
lung cancer [113] | 20 | H&E staining | ORR |
lung cancer [31] | 133 | H&E staining, immunohistochemistry | OS |
melanoma [15] | 177 | H&E staining, immunohistochemistry, RNA-seq, immunofluorescence | OS |
melanoma [43] | 120 | H&E staining, immunohistochemistry, RNA-seq | ORR |
breast cancer [114] | 447 | H&E staining, immunohistochemistry | DFS (HR-) |
breast cancer [19] | 167 | H&E staining, immunohistochemistry | DFS |
pancreatic cancer [112] | 39 | H&E staining, immunohistochemistry, immunofluorescence | OS |
colorectal cancer [32] | 149 | H&E staining, Immunohistochemistry, RNA-seq | DFS OS |
oral squamous cell carcinoma [42] | 75 | H&E staining, immunohistochemistry, immunofluorescence | DFS |
oral squamous cell carcinoma [30] | 168 | H&E staining, immunohistochemistry, RNA-seq, immunofluorescence, flow cytometry | DFS OS |
pancreatic cancer [28] | 63 | H&E staining, immunohistochemistry, RNA-seq, flow cytometry | OS |
pancreatic cancer [33] | 104 | H&E staining, immunohistochemistry, RNA-seq | OS |
pancreatic ductal adenocarcinoma [40] | 51 | H&E staining, immunohistochemistry | OS |
Tumor | Experimental Subject | Treatment | Identification Method | TLS Change |
---|---|---|---|---|
resistant cancer [116] | mice | LIGHT-VTP immunotherapy | H&E staining, immunohistochemistry, immunofluorescence, flow cytometry | increase |
cancer [117] | mice | 3D printing scaffold vaccine | H&E staining, flow cytometry | increase |
glioma [118] | mice | agonistic CD40 therapy | H&E staining, immunohistochemistry, immunofluorescence, flow cytometry, RNA-seq | increase |
hepatoblastoma [107] | 12 patients | neoadjuvant chemotherapy | H&E staining, Immunohistochemistry, RNA-seq | increase |
lung cancer [108] | 122 patients | neoadjuvant chemotherapy | H&E staining, immunohistochemistry | not significant |
urothelial cancer [109] | 24 patients | neoadjuvant immunotherapy | H&E staining, immunohistochemistry, immunofluorescence, RNA-seq | increase |
melanoma renal cell carcinoma [110] | 34 patients | maintenance immunotherapy | H&E staining, immunohistochemistry, immunofluorescence, RNA-seq, flow cytometry | not significant |
cervical [111] | 12 patients | HPV 16 vaccine | H&E staining, Immunohistochemistry, RNA-seq, flow cytometry | increase |
pancreatic cancer [112] | 39 patients | pancreatic tumor vaccine | H&E staining, immunohistochemistry, RNA-seq | increase |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Zou, J.; Zhang, Y.; Zeng, Y.; Peng, Y.; Liu, J.; Xiao, C.; Wu, F. Tertiary Lymphoid Structures: A Potential Biomarker for Anti-Cancer Therapy. Cancers 2022, 14, 5968. https://doi.org/10.3390/cancers14235968
Zou J, Zhang Y, Zeng Y, Peng Y, Liu J, Xiao C, Wu F. Tertiary Lymphoid Structures: A Potential Biomarker for Anti-Cancer Therapy. Cancers. 2022; 14(23):5968. https://doi.org/10.3390/cancers14235968
Chicago/Turabian StyleZou, Ji’an, Yingzhe Zhang, Yue Zeng, Yurong Peng, Junqi Liu, Chaoyue Xiao, and Fang Wu. 2022. "Tertiary Lymphoid Structures: A Potential Biomarker for Anti-Cancer Therapy" Cancers 14, no. 23: 5968. https://doi.org/10.3390/cancers14235968
APA StyleZou, J., Zhang, Y., Zeng, Y., Peng, Y., Liu, J., Xiao, C., & Wu, F. (2022). Tertiary Lymphoid Structures: A Potential Biomarker for Anti-Cancer Therapy. Cancers, 14(23), 5968. https://doi.org/10.3390/cancers14235968