High Treg and PMN-MDSC densities are a hallmark of tertiary lymphoid structures in fatal cases of cervical cancer
Language English Country Great Britain, England Media electronic
Document type Journal Article
PubMed
40998518
PubMed Central
PMC12481325
DOI
10.1136/jitc-2025-012613
PII: jitc-2025-012613
Knihovny.cz E-resources
- Keywords
- Cervical Cancer, Head and Neck Cancer, Myeloid-derived suppressor cell - MDSC, T regulatory cell - Treg,
- MeSH
- Squamous Cell Carcinoma of Head and Neck immunology MeSH
- Tertiary Lymphoid Structures * immunology pathology MeSH
- Papillomavirus Infections * complications immunology MeSH
- Middle Aged MeSH
- Humans MeSH
- Myeloid-Derived Suppressor Cells * immunology metabolism MeSH
- Uterine Cervical Neoplasms * immunology pathology mortality MeSH
- Prognosis MeSH
- T-Lymphocytes, Regulatory * immunology metabolism MeSH
- Aged MeSH
- Check Tag
- Middle Aged MeSH
- Humans MeSH
- Aged MeSH
- Female MeSH
- Publication type
- Journal Article MeSH
BACKGROUND: High densities of tertiary lymphoid structures (TLSs) are associated with improved clinical outcomes in various malignancies, including human papillomavirus (HPV)-associated head and neck squamous cell carcinoma (HNSCC). However, the role of TLSs in shaping antitumor immunity in HPV-induced cervical cancer (CESC) remains unclear. Therefore, we analyzed the density, composition, and prognostic impact of TLSs in patients with CESC as well as patients with HNSCC. METHODS: Multiplex immunofluorescence, immunohistochemistry, and spatial transcriptomics were used to analyze TLS density and composition in HNSCC and CESC tissue sections with respect to patient prognosis. The spatial approach was supplemented by flow cytometry-based analysis of the polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC) phenotype in freshly resected primary tumor tissues. RESULTS: Although both indications were associated with HPV infection, we confirmed a positive correlation between TLS density and improved overall survival only in patients with HNSCC. The TLS composition differed markedly between HNSCC and CESC samples, with a shift toward high regulatory T cell (Treg) and PMN-MDSC abundance in CESC samples. The highest Treg and PMN-MDSC levels were observed in patients with CESC who died of the disease. CESC-infiltrating PMN-MDSCs showed high arginase 1 expression, which correlated with diminished T-cell receptor (TCR)ζ chain expression in CESC-infiltrating T cells. Additionally, the high number of PMN-MDSCs in TLSs was associated with the absence of HPV-specific T cells in CESC. CONCLUSIONS: Unlike in HNSCC, the composition of TLSs, rather than their quantity, was associated with the overall survival of patients with CESC. High numbers of Tregs and PMN-MDSCs infiltrating immature TLSs prevail in patients with CESC who succumbed to the disease and seem to affect tumor-specific immune responses.
1st Faculty of Medicine Charles University Prague Czech Republic
Department of Medical Oncology Leiden University Medical Center Leiden The Netherlands
Department of Pathology and Molecular Medicine Motol University Hospital Prague Czech Republic
Department of Pathology General University Hospital Prague Czech Republic
Fingerland Department of Pathology University Hospital Hradec Kralove Hradec Kralove Czech Republic
See more in PubMed
Cabrita R, Lauss M, Sanna A, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature New Biol. 2020;577:561–5. doi: 10.1038/s41586-019-1914-8. PubMed DOI
Helmink BA, Reddy SM, Gao J, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature New Biol. 2020;577:549–55. doi: 10.1038/s41586-019-1922-8. PubMed DOI PMC
Petitprez F, de Reyniès A, Keung EZ, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature New Biol. 2020;577:556–60. doi: 10.1038/s41586-019-1906-8. PubMed DOI
Hladíková K, Koucký V, Bouček J, et al. Tumor-infiltrating B cells affect the progression of oropharyngeal squamous cell carcinoma via cell-to-cell interactions with CD8+ T cells. J Immunother Cancer. 2019;7:261. doi: 10.1186/s40425-019-0726-6. PubMed DOI PMC
Näsman A, Romanitan M, Nordfors C, et al. Tumor infiltrating CD8+ and Foxp3+ lymphocytes correlate to clinical outcome and human papillomavirus (HPV) status in tonsillar cancer. PLoS One. 2012;7:e38711. doi: 10.1371/journal.pone.0038711. PubMed DOI PMC
Nordfors C, Grün N, Tertipis N, et al. CD8+ and CD4+ tumour infiltrating lymphocytes in relation to human papillomavirus status and clinical outcome in tonsillar and base of tongue squamous cell carcinoma. Eur J Cancer. 2013;49:2522–30. doi: 10.1016/j.ejca.2013.03.019. PubMed DOI
Ruffin AT, Cillo AR, Tabib T, et al. B cell signatures and tertiary lymphoid structures contribute to outcome in head and neck squamous cell carcinoma. Nat Commun. 2021;12:3349. doi: 10.1038/s41467-021-23355-x. PubMed DOI PMC
Garaud S, Dieu-Nosjean MC, Willard-Gallo K. T follicular helper and B cell crosstalk in tertiary lymphoid structures and cancer immunotherapy. Nat Commun. 2022;13:2259. doi: 10.1038/s41467-022-29753-z. PubMed DOI PMC
Zhu J, Lu H, Wang K, et al. Tertiary lymphoid structures in head and neck squamous cell carcinoma. Transl Oncol. 2024;44:101949. doi: 10.1016/j.tranon.2024.101949. PubMed DOI PMC
Pavelková L, Táborská E, Syding LA, et al. Tissue contexture determines the pattern and density of tumor-infiltrating immune cells in HPV-associated squamous cell carcinomas of oropharynx and uterine cervix. Transl Oncol. 2024;41:101884. doi: 10.1016/j.tranon.2024.101884. PubMed DOI PMC
Santegoets SJ, van Ham VJ, Ehsan I, et al. The Anatomical Location Shapes the Immune Infiltrate in Tumors of Same Etiology and Affects Survival. Clin Cancer Res. 2019;25:240–52. doi: 10.1158/1078-0432.CCR-18-1749. PubMed DOI
Posch F, Silina K, Leibl S, et al. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. Oncoimmunology. 2018;7:e1378844. doi: 10.1080/2162402X.2017.1378844. PubMed DOI PMC
Sautès-Fridman C, Petitprez F, Calderaro J, et al. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019;19:307–25. doi: 10.1038/s41568-019-0144-6. PubMed DOI
Linterman MA, Pierson W, Lee SK, et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat Med. 2011;17:975–82. doi: 10.1038/nm.2425. PubMed DOI PMC
Noël G, Fontsa ML, Garaud S, et al. Functional Th1-oriented T follicular helper cells that infiltrate human breast cancer promote effective adaptive immunity. J Clin Invest. 2021;131:e139905. doi: 10.1172/JCI139905. PubMed DOI PMC
Yamaguchi K, Ito M, Ohmura H, et al. Helper T cell-dominant tertiary lymphoid structures are associated with disease relapse of advanced colorectal cancer. Oncoimmunology. 2020;9:1724763. doi: 10.1080/2162402X.2020.1724763. PubMed DOI PMC
Hladíková K, Partlová S, Koucký V, et al. Dysfunction of HPV16-specific CD8+ T cells derived from oropharyngeal tumors is related to the expression of Tim-3 but not PD-1. Oral Oncol. 2018;82:75–82. doi: 10.1016/j.oraloncology.2018.05.010. PubMed DOI
de Vos van Steenwijk PJ, Heusinkveld M, Ramwadhdoebe TH, et al. An Unexpectedly Large Polyclonal Repertoire of HPV-Specific T Cells Is Poised for Action in Patients with Cervical Cancer. Cancer Res. 2010;70:2707–17. doi: 10.1158/0008-5472.CAN-09-4299. PubMed DOI
Piersma SJ, Welters MJP, van der Hulst JM, et al. Human papilloma virus specific T cells infiltrating cervical cancer and draining lymph nodes show remarkably frequent use of HLA-DQ and -DP as a restriction element. Int J Cancer. 2008;122:486–94. doi: 10.1002/ijc.23162. PubMed DOI
Condamine T, Dominguez GA, Youn J-I, et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci Immunol. 2016;1:aaf8943. doi: 10.1126/sciimmunol.aaf8943. PubMed DOI PMC
Ezernitchi AV, Vaknin I, Cohen-Daniel L, et al. TCR zeta down-regulation under chronic inflammation is mediated by myeloid suppressor cells differentially distributed between various lymphatic organs. J Immunol. 2006;177:4763–72. doi: 10.4049/jimmunol.177.7.4763. PubMed DOI
Fleming V, Hu X, Weber R, et al. Targeting Myeloid-Derived Suppressor Cells to Bypass Tumor-Induced Immunosuppression. Front Immunol. 2018;9:398. doi: 10.3389/fimmu.2018.00398. PubMed DOI PMC
Zhang Y, Li J, Yang F, et al. Relationship and prognostic significance of IL-33, PD-1/PD-L1, and tertiary lymphoid structures in cervical cancer. J Leukoc Biol. 2022;112:1591–603. doi: 10.1002/JLB.5MA0322-746R. PubMed DOI
Xiong G, Shan J, Chong Q, et al. Tertiary lymphoid structures associated with enhanced anti-tumor immunity and favorable prognosis in cervical squamous carcinoma. Aging (Milano) 2024;16:6898–920. doi: 10.18632/aging.205733. PubMed DOI PMC
Jiang C, Yuan F, Wang J, et al. Oral squamous cell carcinoma suppressed antitumor immunity through induction of PD-L1 expression on tumor-associated macrophages. Immunobiology. 2017;222:651–7. doi: 10.1016/j.imbio.2016.12.002. PubMed DOI
van Elsas MJ, Middelburg J, Labrie C, et al. Immunotherapy-activated T cells recruit and skew late-stage activated M1-like macrophages that are critical for therapeutic efficacy. Cancer Cell. 2024;42:1032–50. doi: 10.1016/j.ccell.2024.04.011. PubMed DOI
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;5:1416. doi: 10.1038/s42003-022-04356-y. PubMed DOI PMC
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;43:579–90. doi: 10.1016/j.immuni.2015.08.006. PubMed DOI PMC
Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162–74. doi: 10.1038/nri2506. PubMed DOI PMC
Si Y, Merz SF, Jansen P, et al. Multidimensional imaging provides evidence for down-regulation of T cell effector function by MDSC in human cancer tissue. Sci Immunol. 2019;4:eaaw9159. doi: 10.1126/sciimmunol.aaw9159. PubMed DOI
Nan J, Xing YF, Hu B, et al. Endoplasmic reticulum stress induced LOX-1(+) CD15(+) polymorphonuclear myeloid-derived suppressor cells in hepatocellular carcinoma. Immunology. 2018;154:144–55. doi: 10.1111/imm.12876. PubMed DOI PMC
Welters MJP, Ma W, Santegoets SJAM, et al. Intratumoral HPV16-Specific T Cells Constitute a Type I-Oriented Tumor Microenvironment to Improve Survival in HPV16-Driven Oropharyngeal Cancer. Clin Cancer Res. 2018;24:634–47. doi: 10.1158/1078-0432.CCR-17-2140. PubMed DOI
Fridman WH, Meylan M, Petitprez F, et al. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat Rev Clin Oncol. 2022;19:441–57. doi: 10.1038/s41571-022-00619-z. PubMed DOI
Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science. 2022;375:eabf9419. doi: 10.1126/science.abf9419. PubMed DOI
Eberhardt CS, Kissick HT, Patel MR, et al. Functional HPV-specific PD-1+ stem-like CD8 T cells in head and neck cancer. Nature New Biol. 2021;597:279–84. doi: 10.1038/s41586-021-03862-z. PubMed DOI PMC