Rapamycin Induces Phenotypic Alterations in Oral Cancer Cells That May Facilitate Antitumor T Cell Responses
Abstract
:1. Introduction
2. Materials and Methods
2.1. Tissue Samples
2.2. Immunohistochemistry
2.3. Human and Mouse Oral Squamous Cell Carcinoma Cell Lines
2.4. Cell Proliferation Assay
2.5. Migration Assay
2.6. Assessment of Cellular Apoptosis
2.7. Mice and Tumor Model
2.8. In Vivo Rapamycin Treatment
2.9. Tumor Dissociation
2.10. Antibodies and Reagents
2.11. Flow Cytometry
2.12. Intracellular Cytokine Staining of Foxp3
2.13. Mixed Lymphocyte Reaction (MLR) and Intracellular Cytokine Staining
2.14. Statistical Analysis
3. Results
3.2. Rapamycin Exerted a Direct Antitumor Effect in OSCC Cells
3.3. Rapamycin Altered Surface Antigen Expression in OSCC Cells
3.4. Rapamycin Modulated the Distribution of Immune Cell Populations in In Vivo Mouse OSCC Model
3.5. In Vivo Rapamycin Administration Induced Phenotypic Alterations of Dendritic Cells in OSCC Tumor-Bearing Mice
3.6. In Vivo Rapamycin Administration Induced Phenotypic Alterations of Tumor Cells That Facilitated Antitumor T Cell Responses
4. Discussion
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Montero, P.H.; Patel, S.G. Cancer of the oral cavity. Surg. Oncol. Clin. N. Am. 2015, 24, 491–508. [Google Scholar] [CrossRef] [PubMed]
- Ong, W.; Zhao, R.; Lui, B.; Tan, W.; Ebrahimi, A.; Clark, J.R.; Soo, K.C.; Tan, N.C.; Tan, H.K.; Iyer, N.G. Prognostic significance of lymph node density in squamous cell carcinoma of the tongue. Head. Neck. 2016, 38, E859–E866. [Google Scholar] [CrossRef]
- Chakraborty, D.; Natarajan, C.; Mukherjee, A. Advances in oral cancer detection. Adv. Clin. Chem. 2019, 91, 181–200. [Google Scholar] [PubMed]
- Ang, K.K.; Berkey, B.A.; Tu, X.; Zhang, H.Z.; Katz, R.; Hammond, E.H.; Fu, K.K.; Milas, L. Impact of epidermal growth factor receptor expression on survival and pattern of relapse in patients with advanced head and neck carcinoma. Cancer Res. 2002, 62, 7350–7356. [Google Scholar] [PubMed]
- Moy, J.D.; Moskovitz, J.M.; Ferris, R.L. Biological mechanisms of immune escape and implications for immunotherapy in head and neck squamous cell carcinoma. Eur. J. Cancer 2017, 76, 152–166. [Google Scholar] [CrossRef] [PubMed]
- Ciuffreda, L.; Di Sanza, C.; Incani, U.C.; Milella, M. The mTOR pathway: A new target in cancer therapy. Curr. Cancer Drug Targets 2010, 10, 484–495. [Google Scholar] [CrossRef] [PubMed]
- Mayer, I.A.; Arteaga, C.L. The PI3K/AKT Pathway as a Target for Cancer Treatment. Annu. Rev. Med. 2016, 67, 11–28. [Google Scholar] [CrossRef] [PubMed]
- Amornphimoltham, P.; Patel, V.; Sodhi, A.; Nikitakis, N.G.; Sauk, J.J.; Sausville, E.A.; Molinolo, A.A.; Gutkind, J.S. Mammalian target of rapamycin, a molecular target in squamous cell carcinomas of the head and neck. Cancer Res. 2005, 65, 9953–9961. [Google Scholar] [CrossRef]
- Faivre, S.; Kroemer, G.; Raymond, E. Current development of mTOR inhibitors as anticancer agents. Nat. Rev. Drug Discov. 2006, 5, 671–688. [Google Scholar] [CrossRef]
- Huang, J.; Wu, S.; Wu, C.L.; Manning, B.D. Signaling events downstream of mammalian target of rapamycin complex 2 are attenuated in cells and tumors deficient for the tuberous sclerosis complex tumor suppressors. Cancer Res. 2009, 69, 6107–6114. [Google Scholar] [CrossRef]
- O’Donnell, A.; Faivre, S.; Burris, H.A., III; Rea, D.; Papadimitrakopoulou, V.; Shand, N.; Lane, H.A.; Hazell, K.; Zoellner, U.; Kovarik, J.M.; et al. Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors. J. Clin. Oncol. 2008, 26, 1588–1595. [Google Scholar] [CrossRef] [PubMed]
- Besse, B.; Leighl, N.; Bennouna, J.; Papadimitrakopoulou, V.A.; Blais, N.; Traynor, A.M.; Soria, J.C.; Gogov, S.; Miller, N.; Jehl, V.; et al. Phase II study of everolimus-erlotinib in previously treated patients with advanced non-small-cell lung cancer. Ann. Oncol. 2014, 25, 409–415. [Google Scholar] [CrossRef] [PubMed]
- Benjamin, D.; Colombi, M.; Moroni, C.; Hall, M.N. Rapamycin passes the torch: A new generation of mTOR inhibitors. Nat. Rev. Drug Discov. 2011, 10, 868–880. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, K.; Jinhua, P.; Omura, K.; Azuma, M. Multipotency of CD11bhigh Gr-1+ immature myeloid cells accumulating in oral squamous cell carcinoma-bearing mice. Oral. Oncol. 2007, 43, 586–592. [Google Scholar] [CrossRef] [PubMed]
- Tomihara, K.; Fuse, H.; Heshiki, W.; Takei, R.; Zhang, B.; Arai, N.; Nakamori, K.; Noguchi, M. Gemcitabine chemotherapy induces phenotypic alterations of tumor cells that facilitate antitumor T cell responses in a mouse model of oral cancer. Oral. Oncol. 2014, 50, 457–467. [Google Scholar] [CrossRef] [PubMed]
- Fuse, H.; Tomihara, K.; Heshiki, W.; Yamazaki, M.; Akyu-Takei, R.; Tachinami, H.; Furukawa, K.I.; Sakurai, K.; Rouwan, M.; Noguchi, M. Enhanced expression of PD-L1 in oral squamous cell carcinoma-derived CD11b+ Gr-1+ cells and its contribution to immunosuppressive activity. Oral. Oncol. 2016, 59, 20–29. [Google Scholar] [CrossRef] [PubMed]
- Sekido, K.; Tomihara, K.; Tachinami, H.; Heshiki, W.; Sakurai, K.; Moniruzzaman, R.; Imaue, S.; Fujiwara, K.; Noguchi, M. Alterations in composition of immune cells and impairment of anti-tumor immune response in aged oral cancer-bearing mice. Oral. Oncol. 2019, 99, 104462. [Google Scholar] [CrossRef] [PubMed]
- Day, T.A.; Shirai, K.; O’Brien, P.E.; Matheus, M.G.; Godwin, K.; Sood, A.J.; Kompelli, A.; Vick, J.A.; Martin, D.; Vitale-Cross, L.; et al. Inhibition of mTOR Signaling and Clinical Activity of Rapamycin in Head and Neck Cancer in a Window of Opportunity Trial. Clin. Cancer Res. 2019, 25, 1156–1164. [Google Scholar] [CrossRef]
- Kawauchi, T. Cell adhesion and its endocytic regulation in cell migration during neural development and cancer metastasis. Int. J. Mol. Sci. 2012, 13, 4564–4590. [Google Scholar] [CrossRef]
- Zhou, H.; Huang, S. Role of mTOR signaling in tumor cell motility, invasion and metastasis. Curr. Protein Pept. Sci. 2011, 12, 30–42. [Google Scholar]
- Wang, M.H.; Sun, R.; Zhou, X.M.; Zhang, M.Y.; Lu, J.B.; Yang, Y.; Zeng, L.S.; Yang, X.Z.; Shi, L.; Xiao, R.W.; et al. Epithelial cell adhesion molecule overexpression regulates epithelial-mesenchymal transition, stemness and metastasis of nasopharyngeal carcinoma cells via the PTEN/AKT/mTOR pathway. Cell Death Dis. 2018, 9, 2. [Google Scholar] [CrossRef] [PubMed]
- Martin-Belmonte, F.; Gassama, A.; Datta, A.; Yu, W.; Rescher, U.; Gerke, V.; Mostov, K. PTEN-mediated apical segregation of phosphoinositides controls epithelial morphogenesis through Cdc42. Cell 2007, 128, 383–397. [Google Scholar] [CrossRef] [PubMed]
- Araki, K.; Turner, A.P.; Shaffer, V.O.; Gangappa, S.; Keller, S.A.; Bachmann, M.F.; Larsen, C.P.; Ahmed, R. mTOR regulates memory CD8 T-cell differentiation. Nature 2009, 460, 108–112. [Google Scholar] [CrossRef] [PubMed]
- Delgoffe, G.M.; Pollizzi, K.N.; Waickman, A.T.; Heikamp, E.; Meyers, D.J.; Horton, M.R.; Xiao, B.; Worley, P.F.; Powell, J.D. The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat. Immunol. 2011, 12, 295–303. [Google Scholar] [CrossRef] [PubMed]
- Pollizzi, K.N.; Patel, C.H.; Sun, I.H.; Oh, M.H.; Waickman, A.T.; Wen, J.; Delgoffe, G.M.; Powell, J.D. mTORC1 and mTORC2 selectively regulate CD8+ T cell differentiation. J. Clin. Investig. 2015, 125, 2090–2108. [Google Scholar] [CrossRef] [PubMed]
- Ye, L.; Lee, J.; Xu, L.; Mohammed, A.U.; Li, W.; Hale, J.S.; Tan, W.G.; Wu, T.; Davis, C.W.; Ahmed, R.; et al. mTOR Promotes antiviral humoral immunity by differentially regulating CD4 helper T cell and B cell responses. J. Virol. 2017, 91, e01653-16. [Google Scholar] [CrossRef] [PubMed]
- Macedo, C.; Turnquist, H.R.; Castillo-Rama, M.; Zahorchak, A.F.; Shapiro, R.; Thomson, A.W.; Metes, D. Rapamycin augments human DC IL-12p70 and IL27 secretion to promote allogeneic Type 1 polarization modulated by NK cells. Am. J. Transpl. 2013, 13, 2322–2333. [Google Scholar] [CrossRef] [PubMed]
- Waickman, A.T.; Powell, J.D. mTOR, metabolism, and the regulation of T-cell differentiation and function. Immunol. Rev. 2012, 249, 43–58. [Google Scholar] [CrossRef] [PubMed]
- Irelli, A.; Sirufo, M.M.; Scipioni, T.; De Pietro, F.; Pancotti, A.; Ginaldi, L.; De Martinis, M. mTOR Links Tumor Immunity and Bone Metabolism: What are the Clinical Implications? Int. J. Mol. Sci. 2019, 20, 5841. [Google Scholar] [CrossRef]
- Lastwika, K.J.; Wilson, W., III; Li, Q.K.; Norris, J.; Xu, H.; Ghazarian, S.R.; Kitagawa, H.; Kawabata, S.; Taube, J.M.; Yao, S.; 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. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, Z.; Tian, M.; Hu, X.; Wang, L.; Ji, J.; Liao, A. The altered PD-1/PD-L1 pathway delivers the ‘one-two punch’ effects to promote the Treg/Th17 imbalance in pre-eclampsia. Cell Mol. Immunol. 2018, 15, 710–723. [Google Scholar] [CrossRef] [PubMed]
- Zhao, R.; Song, Y.; Wang, Y.; Huang, Y.; Li, Z.; Cui, Y.; Yi, M.; Xia, L.; Zhuang, W.; Wu, X.; 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. [Google Scholar] [CrossRef] [PubMed]
- Scala, S. Molecular Pathways: Targeting the CXCR4-CXCL12 Axis--Untapped Potential in the Tumor Microenvironment. Clin. Cancer Res. 2015, 21, 4278–4285. [Google Scholar] [CrossRef] [PubMed]
- Reikvam, H.; Nepstad, I.; Bruserud, O.; Hatfield, K.J. Correction: Pharmacologic targeting of the PI3K/mTOR pathway controls release of angioregulators from primary human acute myeloid leukemia cells and their neighboring stromal cells. Oncotarget. 2017, 8, 5638–5639. [Google Scholar] [CrossRef] [PubMed]
- Hong, C.H.; Lin, S.H.; Lee, C.H. CCL21 Induces mTOR-dependent MALAT1 Expression, Leading to Cell Migration in Cutaneous T-Cell Lymphoma. Vivo 2019, 33, 793–800. [Google Scholar] [CrossRef] [PubMed]
- Esfahani, K.; Al-Aubodah, T.A.; Thebault, P.; Lapointe, R.; Hudson, M.; Johnson, N.A.; Baran, D.; Bhulaiga, N.; Takano, T.; Cailhier, J.F.; et al. Targeting the mTOR pathway uncouples the efficacy and toxicity of PD-1 blockade in renal transplantation. Nat. Commun. 2019, 10, 4712. [Google Scholar] [CrossRef] [PubMed]
- Cramer, J.D.; Burtness, B.; Ferris, R.L. Immunotherapy for head and neck cancer: Recent advances and future directions. Oral. Oncol. 2019, 99, 104460. [Google Scholar] [CrossRef] [PubMed]
- Tachinami, H.; Tomihara, K.; Yamada, S.I.; Ikeda, A.; Imaue, S.; Hirai, H.; Nakai, H.; Sonoda, T.; Kurohara, K.; Yoshioka, Y.; et al. Neutrophil-to-lymphocyte ratio as an early marker of outcomes in patients with recurrent oral squamous cell carcinoma treated with nivolumab. Br. J. Oral. Maxillofac. Surg. 2023, 61, 320–326. [Google Scholar] [CrossRef] [PubMed]
- Principe, D.R.; Chiec, L.; Mohindra, N.A.; Munshi, H.G. Regulatory T-Cells as an Emerging Barrier to Immune Checkpoint Inhibition in Lung Cancer. Front. Oncol. 2021, 11, 684098. [Google Scholar] [CrossRef]
- Christmas, B.J.; Rafie, C.I.; Hopkins, A.C.; Scott, B.A.; Ma, H.S.; Cruz, K.A.; Woolman, S.; Armstrong, T.D.; Connolly, R.M.; Azad, N.A.; et al. Entinostat converts immune-resistant breast and pancreatic cancers into checkpoint-responsive tumors by reprogramming tumor-infiltrating MDSCs. Cancer Immunol. Res. 2018, 6, 1561–1577. [Google Scholar] [CrossRef]
- Park, S.M.; Youn, J.I. Role of myeloid-derived suppressor cells in immune checkpoint inhibitor therapy in cancer. Arch. Pharm. Res. 2019, 42, 560–566. [Google Scholar] [CrossRef] [PubMed]
mTOR (H Score) | |||
---|---|---|---|
n | Median (Interquartile Range) | p-Values | |
Age, years | 0.29 | ||
<65 | 11 | 131.80 (97.00–161.40) | |
≧65 | 16 | 167.70 (100.43–192.90) | |
Sex | 0.54 | ||
Male | 15 | 153.80 (97.00–221.00) | |
Female | 12 | 142.35 (108.33–180.65) | |
Primary site | 0.71 | ||
Tongue | 10 | 135.15 (96.28–162.73) | |
Mandibular gingiva | 7 | 161.40 (129.10–193.10) | |
Maxillary gingiva | 5 | 183.00 (96.25–232.20) | |
Buccal mucosa | 3 | 192.30 (92.60–236.30) | |
Floor of mouth | 2 | 135.30 (97.00–173.60) | |
Differentiation | 0.63 | ||
Well | 8 | 115.25 (81.88–186.15) | |
Moderate | 9 | 138.50 (94.80–228.65) | |
Poor | 10 | 163.70 (128.03–190.20) | |
Tumor stage | 0.65 | ||
I | 5 | 129.10 (57.75–216.90) | |
II | 8 | 161.6 (151.78–187.63) | |
III | 7 | 101.40 (85.90–189.50) | |
IV | 7 | 133.60 (97.00–193.10) | |
Smoking | 0.98 | ||
Absence | 14 | 147.50 (112.88–190.20 | |
Presence | 13 | 151.10 (94.80–207.05) | |
Drinking | 0.22 | ||
Absence | 13 | 161.80 (124.25–193.35) | |
Presence | 14 | 133.80 (90.93–185.53) |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 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
Yonesi, A.; Tomihara, K.; Takatsuka, D.; Tachinami, H.; Yamazaki, M.; Jadidi, A.R.Y.; Takaichi, M.; Imaue, S.; Fujiwara, K.; Yamada, S.-I.; et al. Rapamycin Induces Phenotypic Alterations in Oral Cancer Cells That May Facilitate Antitumor T Cell Responses. Biomedicines 2024, 12, 1078. https://doi.org/10.3390/biomedicines12051078
Yonesi A, Tomihara K, Takatsuka D, Tachinami H, Yamazaki M, Jadidi ARY, Takaichi M, Imaue S, Fujiwara K, Yamada S-I, et al. Rapamycin Induces Phenotypic Alterations in Oral Cancer Cells That May Facilitate Antitumor T Cell Responses. Biomedicines. 2024; 12(5):1078. https://doi.org/10.3390/biomedicines12051078
Chicago/Turabian StyleYonesi, Amirmoezz, Kei Tomihara, Danki Takatsuka, Hidetake Tachinami, Manabu Yamazaki, Amir Reza Younesi Jadidi, Mayu Takaichi, Shuichi Imaue, Kumiko Fujiwara, Shin-Ichi Yamada, and et al. 2024. "Rapamycin Induces Phenotypic Alterations in Oral Cancer Cells That May Facilitate Antitumor T Cell Responses" Biomedicines 12, no. 5: 1078. https://doi.org/10.3390/biomedicines12051078