Oncolytic herpes simplex virus propagates tertiary lymphoid structure formation via CXCL10/CXCR3 to boost antitumor immunity

Meng-Jie Zhang , Wen-Ping Lin , Qing Wang , Shuo Wang , An Song , Yuan-Yuan Wang , Hao Li , Zhi-Jun Sun

Cell Proliferation ›› 2025, Vol. 58 ›› Issue (1) : e13740

PDF
Cell Proliferation ›› 2025, Vol. 58 ›› Issue (1) : e13740 DOI: 10.1111/cpr.13740
ORIGINAL ARTICLE

Oncolytic herpes simplex virus propagates tertiary lymphoid structure formation via CXCL10/CXCR3 to boost antitumor immunity

Author information +
History +
PDF

Abstract

Inducing tertiary lymphoid structure (TLS) formation can fuel antitumor immunity. It is necessary to create mouse models containing TLS to explore strategies of TLS formation. Oncolytic herpes simplex virus-1 (oHSV) exhibited intense effects in preclinical and clinical trials. However, the role of oHSV in TLS formation remains to be elucidated. Here, we observed the presence of TLS in 4MOSC1 and MC38 subcutaneous tumour models. Interestingly, oHSV evoked TLS formation, and increased infiltration of B cells and stem-like TCF1+CD8+ T cells proliferation. Mechanistically, oHSV increased the expression of TLS-related chemokines, along with upregulated CXCL10/CXCR3 to facilitate TLS formation. Notably, CXCL10 and CXCR3 were favourable prognostic factors for cancer patients, and closely related with immune cells infiltration. Inhibiting CXCL10/CXCR3 reduced TCF1+CD8+ T cells and granzyme B expression, and impaired oHSV-mediated TLS formation. Furthermore, oHSV-mediated TLS formation revealed superior response and survival rate when combined with αPD-1 treatment. Collectively, these findings indicate that oHSV recruits stem-like TCF1+CD8+ T cells through CXCL10/CXCR3 pathway to propagate TLS formation, and warrants future antitumor immunity development.

Cite this article

Download citation ▾
Meng-Jie Zhang, Wen-Ping Lin, Qing Wang, Shuo Wang, An Song, Yuan-Yuan Wang, Hao Li, Zhi-Jun Sun. Oncolytic herpes simplex virus propagates tertiary lymphoid structure formation via CXCL10/CXCR3 to boost antitumor immunity. Cell Proliferation, 2025, 58(1): e13740 DOI:10.1111/cpr.13740

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

SharmaP, Goswami S, RaychaudhuriD, et al. Immune checkpoint therapy-current perspectives and future directions. Cell. 2023;186(8):1652-1669.

[2]

RuffinAT, LiH, VujanovicL, Zandberg DP, FerrisRL, BrunoTC. Improving head and neck cancer therapies by immunomodulation of the tumour microenvironment. Nat Rev Cancer. 2023;23(3):173-188.

[3]

YiM, ZhengX, NiuM, ZhuS, GeH, WuK. Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Mol Cancer. 2022;21(1):28.

[4]

MoradG, Helmink BA, SharmaP, WargoJA. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell. 2022;185(3):576.

[5]

BruniD, AngellHK, GalonJ. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat Rev Cancer. 2020;20(11):662-680.

[6]

SchumacherTN, Thommen DS. Tertiary lymphoid structures in cancer. Science. 2022;375(6576):eabf9419.

[7]

Sautes-FridmanC, Petitprez F, CalderaroJ, FridmanWH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019;19(6):307-325.

[8]

PengY, XiaoL, RongH, et al. Single-cell profiling of tumor-infiltrating TCF1/TCF7+ T cells reveals a T lymphocyte subset associated with tertiary lymphoid structures/organs and a superior prognosis in oral cancer. Oral Oncol. 2021;119:105348.

[9]

Di ModugnoF, Di Carlo A, SpadaS, et al. Tumoral and stromal hMENA isoforms impact tertiary lymphoid structure localization in lung cancer and predict immune checkpoint blockade response in patients with cancer. EBioMedicine. 2024;101:105003.

[10]

LiH, DingJY, ZhangMJ, Yu HJ, SunZJ. Tertiary lymphoid structures and cytokines interconnections: the implication in cancer immunotherapy. Cancer Lett. 2023;568:216293.

[11]

XuM, WangY, XiaR, WeiY, WeiX. Role of the CCL2-CCR2 signalling axis in cancer: mechanisms and therapeutic targeting. Cell Prolif. 2021;54(10):e13115.

[12]

ZhangC, WangXY, ZuoJL, 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.

[13]

ItalianoA, Bessede A, PulidoM, et al. Pembrolizumab in soft-tissue sarcomas with tertiary lymphoid structures: a phase 2 PEMBROSARC trial cohort. Nat Med. 2022;28(6):1199-1206.

[14]

ShalhoutSZ, MillerDM, EmerickKS, Kaufman HL. Therapy with oncolytic viruses: progress and challenges. Nat Rev Clin Oncol. 2023;20(3):160-177.

[15]

ZhangB, ChengP. Improving antitumor efficacy via combinatorial regimens of oncolytic virotherapy. Mol Cancer. 2020;19(1):158.

[16]

MaJ, Ramachandran M, JinC, et al. Characterization of virus-mediated immunogenic cancer cell death and the consequences for oncolytic virus-based immunotherapy of cancer. Cell Death Dis. 2020;11(1):48.

[17]

MelcherA, Harrington K, VileR. Oncolytic virotherapy as immunotherapy. Science. 2021;374(6573):1325-1326.

[18]

ClubbJHA, Kudling TV, HeinioC, et al. Adenovirus encoding tumor necrosis factor alpha and interleukin 2 induces a tertiary lymphoid structure signature in immune checkpoint inhibitor refractory head and neck cancer. Front Immunol. 2022;13:794251.

[19]

HeT, HaoZ, LinM, et al. Oncolytic adenovirus promotes vascular normalization and nonclassical tertiary lymphoid structure formation through STING-mediated DC activation. Onco Targets Ther. 2022;11(1):2093054.

[20]

HouelA, Foloppe J, Dieu-NosjeanMC. Harnessing the power of oncolytic virotherapy and tertiary lymphoid structures to amplify antitumor immune responses in cancer patients. Semin Immunol. 2023;69:101796.

[21]

ThomasS, Kuncheria L, RoulstoneV, et al. Development of a new fusion-enhanced oncolytic immunotherapy platform based on herpes simplex virus type 1. J Immunother Cancer. 2019;7(1):214.

[22]

XieJ, WangS, ZhongY, et al. Oncolytic herpes simplex virus armed with a bacterial GBP1 degrader improves antitumor activity. Mol Ther Oncolytics. 2023;29:61-76.

[23]

WangZ, WuVH, AllevatoMM, et al. Syngeneic animal models of tobacco-associated oral cancer reveal the activity of in situ anti-CTLA-4. Nat Commun. 2019;10(1):5546.

[24]

ZhengDW, DengWW, SongWF, et al. Biomaterial-mediated modulation of oral microbiota synergizes with PD-1 blockade in mice with oral squamous cell carcinoma. Nat Biomed Eng. 2022;6(1):32-43.

[25]

WangS, WuZZ, ZhuSW, et al. CTLA-4 blockade induces tumor pyroptosis via CD8+ T cells in head and neck squamous cell carcinoma. Mol Ther. 2023;31(7):2154-2168.

[26]

LiH, LiuH, FuH, et al. Peritumoral tertiary lymphoid structures correlate with protective immunity and improved prognosis in patients with hepatocellular carcinoma. Front Immunol. 2021;12:648812.

[27]

ChenL, WanSC, MaoL, HuangCF, BuLL, SunZJ. NLRP3 in tumor-associated macrophages predicts a poor prognosis and promotes tumor growth in head and neck squamous cell carcinoma. Cancer Immunol Immunother. 2023;72(6):1647-1660.

[28]

DangXTT, PhungCD, LimCMH, et al. Dendritic cell-targeted delivery of antigens using extracellular vesicles for anti-cancer immunotherapy. Cell Prolif. 2024;57(7):e13622.

[29]

CabritaR, LaussM, SannaA, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577(7791):561-565.

[30]

NtostoglouK, Theodorou SDP, ProctorT, et al. Distinct profiles of proliferating CD8+/TCF1+ T cells and CD163+/PD-L1+ macrophages predict risk of relapse differently among treatment-naive breast cancer subtypes. Cancer Immunol Immunother. 2024;73(3):46.

[31]

ZhangB, LiH, LiuYT, Xiong D, ZhangL, SunZJ. Single-cell chemokine receptor profiles delineate the immune contexture of tertiary lymphoid structures in head and neck squamous cell carcinoma. Cancer Lett. 2023;558:216105.

[32]

MarklF, HuynhD, EndresS, Kobold S. Utilizing chemokines in cancer immunotherapy. Trends Cancer. 2022;8(8):670-682.

[33]

Moreno AyalaMA, Campbell TF, ZhangC, et al. CXCR3 expression in regulatory T cells drives interactions with type I dendritic cells in tumors to restrict CD8+ T cell antitumor immunity. Immunity. 2023;56(7):1613-1630.e5.

[34]

LiX, LuM, YuanM, et al. CXCL10-armed oncolytic adenovirus promotes tumor-infiltrating T-cell chemotaxis to enhance anti-PD-1 therapy. Onco Targets Ther. 2022;11(1):2118210.

[35]

PandeyV, Fleming-Martinez A, BasteaL, et al. CXCL10/CXCR3 signaling contributes to an inflammatory microenvironment and its blockade enhances progression of murine pancreatic precancerous lesions. Elife. 2021;10:e60646.

[36]

FridmanWH, MeylanM, PetitprezF, Sun CM, ItalianoA, Sautes-FridmanC. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat Rev Clin Oncol. 2022;19(7):441-457.

[37]

WangM, ZhaiR, WangM, et al. Tertiary lymphoid structures in head and neck squamous cell carcinoma improve prognosis by recruiting CD8+ T cells. Mol Oncol. 2023;17(8):1514-1530.

[38]

LinD, ShenY, LiangT. Oncolytic virotherapy: basic principles, recent advances and future directions. Signal Transduct Target Ther. 2023;8(1):156.

[39]

HarringtonKJ, Hingorani M, TanayMA, et al. Phase I/II study of oncolytic HSV GM-CSF in combination with radiotherapy and cisplatin in untreated stage III/IV squamous cell cancer of the head and neck. Clin Cancer Res. 2010;16(15):4005-4015.

[40]

MongeC, XieC, MyojinY, et al. Phase I/II study of PexaVec in combination with immune checkpoint inhibition in refractory metastatic colorectal cancer. J Immunother Cancer. 2023;11(2):e005640.

[41]

RamachandranM, Vaccaro A, van de WalleT, et al. Tailoring vascular phenotype through AAV therapy promotes anti-tumor immunity in glioma. Cancer Cell. 2023;41(6):1134-1151.e10.

[42]

LinWP, LiH, SunZJ. T cell exhaustion initiates tertiary lymphoid structures and turbocharges cancer-immunity cycle. EBioMedicine. 2024;104:105154.

[43]

Sautes-FridmanC, Fridman WH. TLS in tumors: what lies within. Trends Immunol. 2016;37(1):1-2.

[44]

ChowMT, OzgaAJ, ServisRL, et al. Intratumoral activity of the CXCR3 chemokine system is required for the efficacy of anti-PD-1 therapy. Immunity. 2019;50(6):1498-1512.e5.

[45]

TokunagaR, ZhangW, NaseemM, et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation -a target for novel cancer therapy. Cancer Treat Rev. 2018;63:40-47.

[46]

WangT, ShenY, LuytenS, Yang Y, JiangX. Tissue-resident memory CD8+ T cells in cancer immunology and immunotherapy. Pharmacol Res. 2020;159:104876.

[47]

WeigelinB, den Boer AT, WagenaE, et al. Cytotoxic T cells are able to efficiently eliminate cancer cells by additive cytotoxicity. Nat Commun. 2021;12(1):5217.

[48]

DangajD, BruandM, GrimmAJ, et al. Cooperation between constitutive and inducible chemokines enables T cell engraftment and immune attack in solid tumors. Cancer Cell. 2019;35(6):885-900.e10.

[49]

SatoY, SilinaK, van den BroekM, HiraharaK, Yanagita M. The roles of tertiary lymphoid structures in chronic diseases. Nat Rev Nephrol. 2023;19(8):525-537.

[50]

CottrellTR, Thompson ED, FordePM, 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(8):1853-1860.

RIGHTS & PERMISSIONS

2024 The Author(s). Cell Proliferation published by Beijing Institute for Stem Cell and Regenerative Medicine and John Wiley & Sons Ltd.

AI Summary AI Mindmap
PDF

234

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/