Programmed death-ligand 1 regulates ameloblastoma growth and recurrence

Linzhou Zhang , Hao Lin , Jiajie Liang , Xuanhao Liu , Chenxi Zhang , Qiwen Man , Ruifang Li , Yi Zhao , Bing Liu

International Journal of Oral Science ›› 2025, Vol. 17 ›› Issue (1) : 29

PDF
International Journal of Oral Science ›› 2025, Vol. 17 ›› Issue (1) : 29 DOI: 10.1038/s41368-025-00364-w
Article

Programmed death-ligand 1 regulates ameloblastoma growth and recurrence

Author information +
History +
PDF

Abstract

Tumor cell-intrinsic programmed death-ligand 1 (PD-L1) signals mediate tumor initiation, progression and metastasis, but their effects in ameloblastoma (AM) have not been reported. In this comprehensive study, we observed marked upregulation of PD-L1 in AM tissues and revealed the robust correlation between elevated PD-L1 expression and increased tumor growth and recurrence rates. Notably, we found that PD-L1 overexpression markedly increased self-renewal capacity and promoted tumorigenic processes and invasion in hTERT+-AM cells, whereas genetic ablation of PD-L1 exerted opposing inhibitory effects. By performing high-resolution single-cell profiling and thorough immunohistochemical analyses in AM patients, we delineated the intricate cellular landscape and elucidated the mechanisms underlying the aggressive phenotype and unfavorable prognosis of these tumors. Our findings revealed that hTERT+-AM cells with upregulated PD-L1 expression exhibit increased proliferative potential and stem-like attributes and undergo partial epithelial‒mesenchymal transition. This phenotypic shift is induced by the activation of the PI3K-AKT-mTOR signaling axis; thus, this study revealed a crucial regulatory mechanism that fuels tumor growth and recurrence. Importantly, targeted inhibition of the PD-L1-PI3K-AKT-mTOR signaling axis significantly suppressed the growth of AM patient-derived tumor organoids, highlighting the potential of PD-L1 blockade as a promising therapeutic approach for AM.

Cite this article

Download citation ▾
Linzhou Zhang, Hao Lin, Jiajie Liang, Xuanhao Liu, Chenxi Zhang, Qiwen Man, Ruifang Li, Yi Zhao, Bing Liu. Programmed death-ligand 1 regulates ameloblastoma growth and recurrence. International Journal of Oral Science, 2025, 17(1): 29 DOI:10.1038/s41368-025-00364-w

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

EffiomOA, OgundanaOM, AkinshipoAO, AkintoyeSO. Ameloblastoma: current etiopathological concepts and management. Oral. Dis., 2018, 24: 307-316.

[2]

KreppelM, ZollerJ. Ameloblastoma-Clinical, radiological, and therapeutic findings. Oral. Dis., 2018, 24: 63-66.

[3]

Almeida RdeA, AndradeES, BarbalhoJC, VajgelA, VasconcelosBC. Recurrence rate following treatment for primary multicystic ameloblastoma: systematic review and meta-analysis. Int. J. Oral. Maxillofac. Surg., 2016, 45: 359-367.

[4]

DandriyalR, GuptaA, PantS, BawejaHH. Surgical management of ameloblastoma: conservative or radical approach. Natl. J. Maxillofac. Surg., 2011, 2: 22-27.

[5]

PogrelMA, MontesDM. Is there a role for enucleation in the management of ameloblastoma?. Int. J. Oral. Maxillofac. Surg., 2009, 38: 807-812.

[6]

ShirsatPM, BansalS, PrasadP, DesaiRS. Low frequency of BRAF V600E immunoexpression in mandibular ameloblastomas: an institutional study. J. Oral. Maxillofac. Pathol., 2018, 22: 353-359.

[7]

DerakhshanS, et al. . High frequency of BRAF V600E mutation in Iranian population ameloblastomas. Med. Oral. Patol. Oral. Cir. Bucal., 2020, 25: e502-e507.

[8]

FernandesGS, GirardiDM, BernardesJPG, FonsecaFP, FregnaniER. Clinical benefit and radiological response with BRAF inhibitor in a patient with recurrent ameloblastoma harboring V600E mutation. BMC Cancer, 2018, 18. 887

[9]

ChaJH, ChanLC, LiCW, HsuJL, HungMC. Mechanisms controlling PD-L1 expression in cancer. Mol. Cell, 2019, 76: 359-370.

[10]

SunC, MezzadraR, SchumacherTN. Regulation and function of the PD-L1 checkpoint. Immunity, 2018, 48: 434-452.

[11]

KornepatiAVR, VadlamudiRK, CurielTJ. Programmed death ligand 1 signals in cancer cells. Nat. Rev. Cancer, 2022, 22: 174-189.

[12]

YuJ, et al. . Regulation of sister chromatid cohesion by nuclear PD-L1. Cell Res., 2020, 30: 590-601.

[13]

HouJ, et al. . PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis. Nat. Cell Biol., 2020, 22: 1264-1275.

[14]

YadollahiP, JeonYK, NgWL, ChoiI. Current understanding of cancer-intrinsic PD-L1: regulation of expression and its protumoral activity. BMB Rep., 2021, 54: 12-20.

[15]

TuX, et al. . PD-L1 (B7-H1) competes with the RNA exosome to regulate the DNA damage response and can be targeted to sensitize to radiation or chemotherapy. Mol. Cell, 2019, 74: 1215-1226 e1214.

[16]

XieXQ, et al. . Targeting ATAD3A-PINK1-mitophagy axis overcomes chemoimmunotherapy resistance by redirecting PD-L1 to mitochondria. Cell Res., 2023, 33: 215-228.

[17]

GaoY, et al. . Acetylation-dependent regulation of PD-L1 nuclear translocation dictates the efficacy of anti-PD-1 immunotherapy. Nat. Cell Biol., 2020, 22: 1064-1075.

[18]

KornepatiAVR, et al. . Tumor intrinsic PD-L1 promotes DNA repair in distinct cancers and suppresses PARP inhibitor-induced synthetic lethality. Cancer Res., 2022, 82: 2156-2170.

[19]

OhKY. Treatment options for advanced ameloblastoma in the era of precision medicine: a brief review. Oral. Oncol., 2023, 146. 106585

[20]

KarpathiouG, et al. . Autophagy and immune microenvironment in craniopharyngioma and ameloblastoma. Exp. Mol. Pathol., 2021, 123. 104712

[21]

GlavianoA, et al. . PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol. Cancer, 2023, 22. 138

[22]

OrtegaMA, et al. . PD-1/PD-L1 axis: implications in immune regulation, cancer progression, and translational applications. J. Mol. Med., 2024, 102: 987-1000.

[23]

LastwikaKJ, et al. . Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung cancer. Cancer Res., 2016, 76: 227-238.

[24]

O’DonnellJS, MassiD, TengMWL, MandalaM. PI3K-AKT-mTOR inhibition in cancer immunotherapy, redux. Semin. Cancer Biol., 2018, 48: 91-103.

[25]

ZhaoR, et al. . PD-1/PD-L1 blockade rescue exhausted CD8+ T cells in gastrointestinal stromal tumours via the PI3K/Akt/mTOR signalling pathway. Cell Prolif., 2019, 52. e12571

[26]

HurnikP, et al. . Metastasising ameloblastoma or ameloblastic carcinoma? A case report with mutation analyses. BMC Oral. Health, 2023, 23. 563

[27]

EricH, et al. . High expression of PD-L1 on conventional dendritic cells in tumour-draining lymph nodes is associated with poor prognosis in oral cancer. Cancer Immunol. Immunother., 2024, 73: 165.

[28]

SoopanitT, LaokulrathN, ChayopasakulV, PongsapichW. Prognostic value and clinicopathological status of PD-L1 expression and CD8+ TILs in oral squamous cell cancer patients with or without traditional risk factors. Head. Neck, 2023, 45: 1017-1025.

[29]

MaoY, et al. . The landscape of objective response rate of anti-PD-1/L1 monotherapy across 31 types of cancer: a system review and novel biomarker investigating. Cancer Immunol. Immunother., 2023, 72: 2483-2498.

[30]

CarlisleJW, SteuerCE, OwonikokoTK, SabaNF. An update on the immune landscape in lung and head and neck cancers. CA Cancer J. Clin., 2020, 70: 505-517.

[31]

ChangCH, et al. . Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell, 2015, 162: 1229-1241.

[32]

GuptaHB, et al. . Tumor cell-intrinsic PD-L1 promotes tumor-initiating cell generation and functions in melanoma and ovarian cancer. Signal Transduct. Target Ther., 2016, 1. 16030-

[33]

ChenC, ZhaoS, KarnadA, FreemanJW. The biology and role of CD44 in cancer progression: therapeutic implications. J. Hematol. Oncol., 2018, 11: 64.

[34]

SmithLM, et al. . CD133/prominin-1 is a potential therapeutic target for antibody-drug conjugates in hepatocellular and gastric cancers. Br. J. Cancer, 2008, 99: 100-109.

[35]

MaI, AllanAL. The role of human aldehyde dehydrogenase in normal and cancer stem cells. Stem Cell Rev. Rep., 2011, 7: 292-306.

[36]

KalogirouEM, et al. . Decoding a gene expression program that accompanies the phenotype of sporadic and basal cell nevus syndrome-associated odontogenic keratocyst. J. Oral. Pathol. Med., 2022, 51: 649-658.

[37]

VanjeMM, TanveerS, AhmedSA, KumarS, VanjeT. Immunoexperssion of cancer stem cell marker (CD44) in ameloblastoma. J. Oral. Maxillofac. Pathol., 2019, 23: 400-406.

[38]

KhanW, et al. . Stem cell markers SOX-2 and OCT-4 enable to resolve the diagnostic dilemma between ameloblastic carcinoma and aggressive solid multicystic ameloblastoma. Adv. Biomed. Res., 2018, 7: 149.

[39]

JuuriE, IsakssonS, JussilaM, HeikinheimoK, ThesleffI. Expression of the stem cell marker, SOX2, in ameloblastoma and dental epithelium. Eur. J. Oral. Sci., 2013, 121: 509-516.

[40]

XiongG, et al. . Single-cell transcriptomics reveals cell atlas and identifies cycling tumor cells responsible for recurrence in ameloblastoma. Int. J. Oral. Sci., 2024, 16: 21.

[41]

DongreA, WeinbergRA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat. Rev. Mol. Cell Biol., 2019, 20: 69-84.

[42]

VerstappeJ, BerxG. A role for partial epithelial-to-mesenchymal transition in enabling stemness in homeostasis and cancer. Semin Cancer Biol., 2023, 90: 15-28.

[43]

HuangRY, et al. . An EMT spectrum defines an anoikis-resistant and spheroidogenic intermediate mesenchymal state that is sensitive to e-cadherin restoration by a src-kinase inhibitor, saracatinib (AZD0530). Cell Death Dis., 2013, 4. e915

[44]

NietoMA, HuangRY, JacksonRA, ThieryJP. Emt: 2016. Cell, 2016, 166: 21-45.

[45]

ZhangJ, et al. . Interleukin-8/beta-catenin mediates epithelial-mesenchymal transition in ameloblastoma. Oral. Dis., 2019, 25: 1964-1971.

[46]

JiangC, et al. . Mesenchymal stromal cell-derived interleukin-6 promotes epithelial-mesenchymal transition and acquisition of epithelial stem-like cell properties in ameloblastoma epithelial cells. Stem Cells, 2017, 35: 2083-2094.

[47]

LiaoC, et al. . Partial EMT in squamous cell carcinoma: a snapshot. Int. J. Biol. Sci., 2021, 17: 3036-3047.

[48]

SiarCH, NgKH. Epithelial-to-mesenchymal transition in ameloblastoma: focus on morphologically evident mesenchymal phenotypic transition. Pathology, 2019, 51: 494-501.

[49]

GrynbergS, et al. . Neoadjuvant BRAF-targeted therapy for ameloblastoma of the mandible: an organ preservation approach. J. Natl. Cancer Inst., 2024, 116: 539-546.

[50]

YouZ, LiuSP, DuJ, WuYH, ZhangSZ. Advancements in MAPK signaling pathways and MAPK-targeted therapies for ameloblastoma: a review. J. Oral. Pathol. Med., 2019, 48: 201-205.

[51]

OberholzerPA, et al. . RAS mutations are associated with the development of cutaneous squamous cell tumors in patients treated with RAF inhibitors. J. Clin. Oncol., 2012, 30: 316-321.

[52]

BrunetM, KhalifaE, ItalianoA. Enabling precision medicine for rare head and neck tumors: the example of BRAF/MEK targeting in patients with metastatic ameloblastoma. Front. Oncol., 2019, 9: 1204.

[53]

LuSW, et al. . IL-20 antagonist suppresses PD-L1 expression and prolongs survival in pancreatic cancer models. Nat. Commun., 2020, 11. 4611

[54]

Dong, X. et al. Consensus statement on extracellular vesicles in liquid biopsy for advancing laboratory medicine. Clin. Chem. Lab Med.63, 465–482 (2024).

[55]

ZhangLZ, et al. . PD-1 carried on small extracellular vesicles leads to OSCC metastasis. J. Dent. Res, 2023, 102: 795-805.

[56]

WangR, et al. . Blockade of dual immune checkpoint inhibitory signals with a CD47/PD-L1 bispecific antibody for cancer treatment. Theranostics, 2023, 13: 148-160.

[57]

ChangTH, et al. . LGR5(+) epithelial tumor stem-like cells generate a 3D-organoid model for ameloblastoma. Cell Death Dis., 2020, 11. 338

RIGHTS & PERMISSIONS

The Author(s)

AI Summary AI Mindmap
PDF

163

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/