The Distinctive Features behind the Aggressiveness of Oral and Cutaneous Squamous Cell Carcinomas
Abstract
:Simple Summary
Abstract
1. Introduction
1.1. Oral Squamous Cell Carcinomas
1.2. Cutaneous Squamous Cell Carcinoma
2. The Process of Carcinogenesis: Gene Mutations in Oral and Cutaneous Squamous Cell Carcinomas
3. Tumor Microenvironment
3.1. Extracellular Matrix
3.2. Cancer Associated Fibroblasts
3.3. Immune Cells
3.4. The Importance of the TME for the Treatment of OSCC and CSCC
3.4.1. CAF-Targeting Strategies
3.4.2. Immunotherapy
4. Conclusions and Future Directions
Supplementary Materials
Author Contributions
Funding
Conflicts of Interest
References
- Ferlay, J.; Colombet, M.; Soerjomataram, I.; Mathers, C.; Parkin, D.M.; Piñeros, M.; Znaor, A.; Bray, F. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int. J. Cancer 2019, 144, 1941–1953. [Google Scholar] [CrossRef] [Green Version]
- Dotto, G.P.; Rustgi, A.K. Squamous Cell Cancers: A Unified Perspective on Biology and Genetics. Cancer Cell 2016, 29, 622–637. [Google Scholar] [CrossRef] [Green Version]
- Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Primers 2020, 6, 92. [Google Scholar] [CrossRef]
- Alam, M.; Ratner, D. Cutaneous Squamous-Cell Carcinoma. N. Engl. J. Med. 2001, 344, 975–983. [Google Scholar] [CrossRef] [PubMed]
- Chow, L.Q.M. Head and Neck Cancer. N. Engl. J. Med. 2020, 382, 60–72. [Google Scholar] [CrossRef] [PubMed]
- Koyfman, S.A.; Ismaila, N.; Crook, D.; D’Cruz, A.; Rodriguez, C.P.; Sher, D.J.; Silbermins, D.; Sturgis, E.M.; Tsue, T.T.; Weiss, J.; et al. Management of the Neck in Squamous Cell Carcinoma of the Oral Cavity and Oropharynx: ASCO Clinical Practice Guideline. J. Clin. Oncol. 2019, 37, 1753–1774. [Google Scholar] [CrossRef]
- Chen, S.H.; Hsiao, S.Y.; Chang, K.Y.; Chang, J.Y. New Insights Into Oral Squamous Cell Carcinoma: From Clinical Aspects to Molecular Tumorigenesis. Int. J. Mol. Sci. 2021, 22, 2252. [Google Scholar] [CrossRef]
- Andrews, E.; Seaman, W.T.; Webster-Cyriaque, J. Oropharyngeal carcinoma in non-smokers and non-drinkers: A role for HPV. Oral Oncol. 2009, 45, 486–491. [Google Scholar] [CrossRef]
- Zur Hausen, H. Papillomaviruses and cancer: From basic studies to clinical application. Nat. Rev. Cancer 2002, 2, 342–350. [Google Scholar] [CrossRef] [PubMed]
- Sánchez-Danés, A.; Blanpain, C. Deciphering the cells of origin of squamous cell carcinomas. Nat. Rev. Cancer. 2018, 18, 549–561. [Google Scholar] [CrossRef]
- Velleuer, E.; Dietrich, R. Fanconi anemia: Young patients at high risk for squamous cell carcinoma. Mol. Cell. Pediatr. 2014, 1, 9. [Google Scholar] [CrossRef] [Green Version]
- Cramer, J.D.; Burtness, B.; Le, Q.T.; Ferris, R.L. The changing therapeutic landscape of head and neck cancer. Nat. Rev. Clin. Oncol. 2019, 16, 669–683. [Google Scholar] [CrossRef]
- Bossi, P.; Resteghini, C.; Paielli, N.; Licitra, L.; Pilotti, S.; Perrone, F. Prognostic and predictive value of EGFR in head and neck squamous cell carcinoma. Oncotarget 2016, 7, 74362–74379. [Google Scholar] [CrossRef] [Green Version]
- Bonner, J.A.; Harari, P.M.; Giralt, J.; Azarnia, N.; Shin, D.M.; Cohen, R.B.; Jones, C.U.; Sur, R.; Raben, D.; Jassem, J.; et al. Radiotherapy plus Cetuximab for Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2006, 354, 567–578. [Google Scholar] [CrossRef] [Green Version]
- Seiwert, T.Y.; Burtness, B.; Mehra, R.; Weiss, J.; Berger, R.; Eder, J.P.; Heath, K.; McClanahan, T.; Lunceford, J.; Gause, C.; et al. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): An open-label, multicentre, phase 1b trial. Lancet Oncol. 2016, 17, 956–965. [Google Scholar] [CrossRef]
- Naimi, A.; Mohammed, R.N.; Raji, A.; Chupradit, S.; Yumashev, A.V.; Suksatan, W.; Shalaby, M.N.; Thangavelu, L.; Kamrava, S.; Shomali, N.; et al. Tumor immunotherapies by immune checkpoint inhibitors (ICIs); the pros and cons. Cell Commun. Signal. 2022, 20, 44. [Google Scholar] [CrossRef] [PubMed]
- Barton, V.; Armeson, K.; Hampras, S.; Ferris, L.K.; Visvanathan, K.; Rollison, D.; Alberg, A.J. Nonmelanoma skin cancer and risk of all-cause and cancer-related mortality: A systematic review. Arch. Dermatol. Res. 2017, 309, 243–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schmults, C.D.; Karia, P.S.; Carter, J.B.; Han, J.; Qureshi, A.A. Factors Predictive of Recurrence and Death From Cutaneous Squamous Cell Carcinoma. JAMA Dermatol. 2013, 149, 541. [Google Scholar] [CrossRef] [Green Version]
- Clayman, G.L.; Lee, J.J.; Holsinger, F.C.; Zhou, X.; Duvic, M.; El-Naggar, A.K.; Prieto, V.G.; Altamirano, E.; Tucker, S.L.; Strom, S.S.; et al. Mortality Risk From Squamous Cell Skin Cancer. J. Clin. Oncol. 2005, 23, 759–765. [Google Scholar] [CrossRef]
- Shelton, M.E.; Adamson, A.S. Review and Update on Evidence-Based Surgical Treatment Recommendations for Nonmelanoma Skin Cancer. Dermatol. Clin. 2019, 37, 425–433. [Google Scholar] [CrossRef]
- Juarranz, Á.; Jaén, P.; Sanz-Rodríguez, F.; Cuevas, J.; González, S. Photodynamic therapy of cancer. Basic principles and applications. Clin. Transl. Oncol. 2008, 10, 148–154. [Google Scholar] [CrossRef] [PubMed]
- Patel, R.; Chang, A.L.S. Immune Checkpoint Inhibitors for Treating Advanced Cutaneous Squamous Cell Carcinoma. Am. J. Clin. Dermatol. 2019, 20, 477–482. [Google Scholar] [CrossRef] [PubMed]
- US Food and Drug Administration. FDA Approves Cemiplimab-Rwlc for Metastatic or Locally Advanced Cutaneous Squamous Cell Carcinoma. FDA Website. Available online: https://www.fda.gov/drugs/drugapprovals-and-databases (accessed on 24 January 2023).
- Tang, X.H.; Scognamiglio, T.; Gudas, L.J. Basal stem cells contribute to squamous cell carcinomas in the oral cavity. Carcinogenesis 2013, 34, 1158–1164. [Google Scholar] [CrossRef]
- De Gruijl, F.R.; Tensen, C.P. Pathogenesis of Skin Carcinomas and a Stem Cell as Focal Origin. Front. Med. 2018, 5, 165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pickering, C.R.; Zhang, J.; Yoo, S.Y.; Bengtsson, L.; Moorthy, S.; Neskey, D.M.; Zhao, M.; Alves, M.V.O.; Chang, K.; Drummond, J.; et al. Integrative Genomic Characterization of Oral Squamous Cell Carcinoma Identifies Frequent Somatic Drivers. Cancer Discov. 2013, 3, 770–781. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.Y.; Hanna, G.J.; Laga, A.C.; Haddad, R.I.; Lorch, J.H.; Hammerman, P.S. Genomic Analysis of Metastatic Cutaneous Squamous Cell Carcinoma. Clin. Cancer Res. 2015, 21, 1447–1456. [Google Scholar] [CrossRef] [Green Version]
- Inman, G.J.; Wang, J.; Nagano, A.; Alexandrov, L.B.; Purdie, K.J.; Taylor, R.G.; Sherwood, V.; Thomson, J.; Hogan, S.; Spender, L.C.; et al. The genomic landscape of cutaneous SCC reveals drivers and a novel azathioprine associated mutational signature. Nat. Commun. 2018, 9, 3667. [Google Scholar] [CrossRef]
- Chang, D.; Shain, A.H. The landscape of driver mutations in cutaneous squamous cell carcinoma. Nat. Genom. Med. 2021, 6, 61. [Google Scholar] [CrossRef]
- Hedberg, M.L.; Berry, C.T.; Moshiri, A.S.; Xiang, Y.; Yeh, C.J.; Attilasoy, C.; Capell, B.C.; Seykora, J.T. Molecular Mechanisms of Cutaneous Squamous Cell Carcinoma. Int. J. Mol. Sci. 2022, 23, 3478. [Google Scholar] [CrossRef]
- Kim, Y.; Jung, S.; Park, Y.M.; Park, G.S.; Kim, H.S.; Maeng, L.; Chung, Y. Targeted deep sequencing reveals genomic alterations of actinic keratosis/cutaneous squamous cell carcinoma in situ and cutaneous squamous cell carcinoma. Exp. Dermatol. 2022, 32, 447–456. [Google Scholar] [CrossRef]
- Piipponen, M.; Riihilä, P.; Nissinen, L.; Kähäri, V.M. The role of p53 in Progression of Cutaneous Squamous Cell Carcinoma. Cancers 2021, 7, 4507. [Google Scholar] [CrossRef]
- Leemans, C.R.; Snijders, P.J.F.; Brakenhoff, R.H. The molecular landscape of head and neck cancer. Nat. Rev. Cancer 2018, 18, 269–282. [Google Scholar] [CrossRef]
- Farah, C.S. Molecular landscape of head and neck cancer and implications for therapy. Ann. Transl. Med. 2021, 9, 915. [Google Scholar] [CrossRef]
- Wils, L.J.; Poell, J.B.; Brink, A.; Evren, I.; Brouns, E.R.; de Visscher, J.G.; Bloemena, E.; Brakenhoff, R.H. Elucidating the Genetic Landscape of Oral Leukoplakia to Predict Malignant Transformation. Clin. Cancer Res. 2023, 29, 602–613. [Google Scholar] [CrossRef]
- Liang, L.; Li, Y.; Ying, B.; Huang, X.; Liao, S.; Yang, J.; Liao, G. Mutation-associated transcripts reconstruct the prognostic features of oral tongue squamous cell carcinoma. Int. J. Oral Sci. 2023, 15, 1–11. [Google Scholar] [CrossRef]
- Mitsui, H.; Suárez-Fariñas, M.; Gulati, N.; Shah, K.R.; Cannizzaro, M.V.; Coats, I.; Felsen, D.; Krueger, J.G.; Carucci, J.A. Gene expression profiling of the leading edge of cutaneous squamous cell carcinoma: IL-24-driven MMP-7. J. Investig. Dermatol. 2014, 134, 1418–1427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steffens Reinhardt, L.; Groen, K.; Newton, C.; Avery-Kiejda, K.A. The role of truncated p53 isoforms in the DNA damage response. Biochim. Biophys. Acta Rev. Cancer 2023, 1878, 188882. [Google Scholar] [CrossRef] [PubMed]
- Nappi, A.; Miro, C.; Pezone, A.; Tramontano, A.; Di Cicco, E.; Sagliocchi, S.; Cicatiello, A.G.; Murolo, M.; Torabinejad, S.; Abbotto, E.; et al. Loss of p53 activates thyroid hormone via type 2 deiodinase and enhances DNA damage. Nat. Commun. 2023, 14, 1244. [Google Scholar] [CrossRef] [PubMed]
- Shah, P.A.; Huang, C.; Li, Q.; Kazi, S.A.; Byers, L.A.; Wang, J.; Johnson, F.M.; Frederick, M.J. NOTCH1 Signaling in Head and Neck Squamous Cell Carcinoma. Cells 2020, 9, 2677. [Google Scholar] [CrossRef]
- Fukusumi, T.; Califano, J.A. The NOTCH Pathway in Head and Neck Squamous Cell Carcinoma. J. Dent. Res. 2018, 97, 645–653. [Google Scholar] [CrossRef]
- Nowell, C.S.; Radtke, F. Notch as a tumour suppressor. Nat. Rev. Cancer 2017, 17, 145–159. [Google Scholar] [CrossRef]
- Sherwood, V.; Thomson, J.; Hogan, S. Cancer Genome Atlas Research Network. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015, 517, 576–582. [Google Scholar] [CrossRef] [Green Version]
- Pastushenko, I.; Mauri, F.; Song, Y.; de Cock, F.; Meeusen, B.; Swedlund, B.; Impens, F.; Van Haver, D.; Opitz, M.; Thery, M.; et al. Fat1 deletion promotes hybrid EMT state, tumour stemness and metastasis. Nature 2020, 589, 448–455. [Google Scholar] [CrossRef] [PubMed]
- Martin, D.; Degese, M.S.; Vitale-Cross, L.; Iglesias-Bartolome, R.; Valera, J.L.C.; Wang, Z.; Feng, X.; Yeerna, H.; Vadmal, V.; Moroishi, T.; et al. Assembly and activation of the Hippo signalome by FAT1 tumor suppressor. Nat. Commun. 2018, 9, 2372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ashford, B.G.; Clark, J.; Gupta, R.; Iyer, N.G.; Yu, B.; Ranson, M. Reviewing the genetic alterations in high-risk cutaneous squamous cell carcinoma: A search for prognostic markers and therapeutic targets. Head Neck 2017, 39, 1462–1469. [Google Scholar] [CrossRef]
- Lazar, A.D.; Dinescu, S.; Costache, M. Deciphering the Molecular Landscape of Cutaneous Squamous Cell Carcinoma for Better Diagnosis and Treatment. J. Clin. Med. 2020, 9, 2228. [Google Scholar] [CrossRef]
- Thomson, J.; Bewicke-Copley, F.; Anene, C.A.; Gulati, A.; Nagano, A.; Purdie, K.; Inman, G.J.; Proby, C.M.; Leigh, I.M.; Harwood, C.A.; et al. The Genomic Landscape of Actinic Keratosis. J. Investig. Dermatol. 2021, 141, 1664–1674.e7. [Google Scholar] [CrossRef]
- Ko, E.K.; Capell, B.C. Methyltransferases in the Pathogenesis of Keratinocyte Cancers. Cancers 2021, 13, 3402. [Google Scholar] [CrossRef]
- Anderson, N.M.; Simón, M.C. The tumor microenvironment. Curr. Biol. 2020, 30, R921–R925. [Google Scholar] [CrossRef]
- Walker, C.; Mojares, E.; del Río Hernández, A. Role of Extracellular Matrix in Development and Cancer Progression. Int. J. Mol. Sci. 2018, 19, 3028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Najafi, M.; Farhood, B.; Mortezaee, K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J. Cell. Biochem. 2019, 120, 2782–2790. [Google Scholar] [CrossRef] [PubMed]
- Harada, T.; Shinohara, M.; Nakamura, S.; Oka, M. An immunohistochemical study of the extracellular matrix in oral squamous cell carcinoma and its association with invasive and metastatic potential. Virchows Archiv. 1994, 424, 257–266. [Google Scholar] [CrossRef] [PubMed]
- Knowles, L.M.; Gurski, L.A.; Engel, C.; Gnarra, J.R.; Maranchie, J.K.; Pilch, J. Integrin αvβ3 and Fibronectin Upregulate Slug in Cancer Cells to Promote Clot Invasion and Metastasis. Cancer Res. 2013, 73, 6175–6184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scanlon, C.S.; Van Tubergen, E.A.; Inglehart, R.C.; D’Silva, N.J. Biomarkers of Epithelial-Mesenchymal Transition in Squamous Cell Carcinoma. J. Dent. Res. 2013, 92, 114–121. [Google Scholar] [CrossRef] [Green Version]
- Janes, S.M.; Watt, F.M. New roles for integrins in squamous-cell carcinoma. Nat. Rev. Cancer 2006, 6, 175–183. [Google Scholar] [CrossRef] [PubMed]
- Thulabandu, V.; Chen, D. Atit RP. Dermal fibroblast in cutaneous development and healing. WIREs Dev. Biol. 2018, 7, 307. [Google Scholar] [CrossRef]
- Driskell, R.R.; Watt, F.M. Understanding fibroblast heterogeneity in the skin. Trends Cell Biol. 2015, 25, 92–99. [Google Scholar] [CrossRef]
- Teichgräber, V.; Monasterio, C.; Chaitanya, K.; Boger, R.; Gordon, K.; Dieterle, T.; Jäger, D.; Bauer, S. Specific inhibition of fibroblast activation protein (FAP)-alpha prevents tumor progression in vitro. Adv. Med. Sci. 2015, 60, 264–272. [Google Scholar] [CrossRef]
- Nurmik, M.; Ullmann, P.; Rodriguez, F.; Haan, S.; Letellier, E. In search of definitions: Cancer-associated fibroblasts and their markers. Int. J. Cancer 2020, 146, 895–905. [Google Scholar] [CrossRef] [Green Version]
- Sasaki, K.; Sugai, T.; Ishida, K.; Osakabe, M.; Amano, H.; Kimura, H.; Sakuraba, M.; Kashiwa, K.; Kobayashi, S. Analysis of cancer-associated fibroblasts and the epithelial-mesenchymal transition in cutaneous basal cell carcinoma, squamous cell carcinoma, and malignant melanoma. Hum. Pathol. 2018, 79, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef]
- Kumar, D.; New, J.; Vishwakarma, V.; Joshi, R.; Enders, J.; Lin, F.; Dasari, S.; Gutierrez, W.R.; Leef, G.; Ponnurangam, S.; et al. Cancer-Associated Fibroblasts Drive Glycolysis in a Targetable Signaling Loop Implicated in Head and Neck Squamous Cell Carcinoma Progression. Cancer Res 2018, 78, 3769–3782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuzet, S.E.; Gaggioli, C. Fibroblast activation in cancer: When seed fertilizes soil. Cell Tissue Res. 2016, 365, 607–619. [Google Scholar] [CrossRef] [PubMed]
- Smithmyer, M.E.; Spohn, J.B.; Kloxin, A.M. Probing Fibroblast Activation in Response to Extracellular Cues with Whole Protein- or Peptide-Functionalized Step-Growth Hydrogels. ACS Biomater. Sci. Eng. 2018, 4, 3304–3316. [Google Scholar] [CrossRef]
- Joshi, R.S.; Kanugula, S.S.; Sudhir, S.; Pereira, M.P.; Jain, S.; Aghi, M.K. The Role of Cancer-Associated Fibroblasts in Tumor Progression. Cancers 2021, 13, 1399. [Google Scholar] [CrossRef] [PubMed]
- Eke, I.; Storch, K.; Krause, M.; Cordes, N. Cetuximab Attenuates Its Cytotoxic and Radiosensitizing Potential by Inducing Fibronectin Biosynthesis. Cancer Res. 2013, 73, 5869–5879. [Google Scholar] [CrossRef] [Green Version]
- Grauel, A.L.; Nguyen, B.; Ruddy, D.; Laszewski, T.; Schwartz, S.; Chang, J.; Chen, J.; Piquet, M.; Pelletier, M.; Yan, Z.; et al. TGFβ-blockade uncovers stromal plasticity in tumors by revealing the existence of a subset of interferon-licensed fibroblasts. Nat. Commun. 2020, 11, 6315. [Google Scholar] [CrossRef]
- Ghahremanifard, P.; Chanda, A.; Bonni, S.; Bose, P. TGF-β Mediated Immune Evasion in Cancer—Spotlight on Cancer-Associated Fibroblasts. Cancers 2020, 12, 3650. [Google Scholar] [CrossRef]
- Xiao, L.; Zhu, H.; Shu, J.; Gong, D.; Zheng, D.; Gao, J. Overexpression of TGF-β1 and SDF-1 in cervical cancer-associated fibroblasts promotes cell growth, invasion and migration. Arch. Gynecol. Obstet. 2022, 305, 179–192. [Google Scholar] [CrossRef]
- Gallego-Rentero, M.; Gutiérrez-Pérez, M.; Fernández-Guarino, M.; Mascaraque, M.; Portillo-Esnaola, M.; Gilaberte, Y.; Carrasco, E.; Juarranz, Á. TGFβ1 Secreted by Cancer-Associated Fibroblasts as an Inductor of Resistance to Photodynamic Therapy in Squamous Cell Carcinoma Cells. Cancers 2021, 13, 5613. [Google Scholar] [CrossRef]
- Guo, S.; Deng, C.X. Effect of Stromal Cells in Tumor Microenvironment on Metastasis Initiation. Int. J. Biol. Sci. 2018, 14, 2083–2093. [Google Scholar] [CrossRef]
- Wheeler, S.E.; Shi, H.; Lin, F.; Dasari, S.; Bednash, J.; Thorne, S.; Watkins, S.; Joshi, R.; Thomas, S.M. Enhancement of head and neck squamous cell carcinoma proliferation, invasion, and metastasis by tumor-associated fibroblasts in preclinical models. Head Neck 2013, 36, 385–392. [Google Scholar] [CrossRef] [Green Version]
- Hwang, Y.S.; Xianglan, Z.; Park, K.K.; Chung, W.Y. Functional invadopodia formation through stabilization of the PDPN transcript by IMP-3 and cancer-stromal crosstalk for PDPN expression. Carcinogenesis 2012, 33, 2135–2146. [Google Scholar] [CrossRef] [Green Version]
- Ruffin, A.T.; Li, H.; Vujanovic, L.; Zandberg, D.P.; Ferris, R.L.; Bruno, T.C. Improving head and neck cancer therapies by immunomodulation of the tumour microenvironment. Nat. Rev. Cancer 2023, 23, 173–188. [Google Scholar] [CrossRef]
- Ansary, T.M.; Hossain, M.R.; Komine, M.; Ohtsuki, M. Immunotherapy for the Treatment of Squamous Cell Carcinoma: Potential Benefits and Challenges. Int. J. Mol. Sci. 2022, 23, 8530. [Google Scholar] [CrossRef]
- Fang, J.; Li, X.; Da Ma, D.; Liu, X.; Chen, Y.; Wang, Y.; Lui, V.W.Y.; Xia, J.; Bin Cheng, B.; Wang, Z. Prognostic significance of tumor infiltrating immune cells in oral squamous cell carcinoma. BMC Cancer 2017, 17, 375. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Zeng, Y.; Qu, Q.; Zhu, J.; Liu, Z.; Ning, W.; Zeng, H.; Zhang, N.; Du, W.; Chen, C.; et al. PD-L1 induced by IFN-γ from tumor-associated macrophages via the JAK/STAT3 and PI3K/AKT signaling pathways promoted progression of lung cancer. Int. J. Clin. Oncol. 2017, 22, 1026–1033. [Google Scholar] [CrossRef] [PubMed]
- Woolaver, R.; Wang, X.; Krinsky, A.L.; Waschke, B.C.; Chen, S.M.Y.; Popolizio, V.; Nicklawsky, A.G.; Gao, D.; Chen, Z.; Jimeno, A.; et al. Differences in TCR repertoire and T cell activation underlie the divergent outcomes of antitumor immune responses in tumor-eradicating versus tumor-progressing hosts. J. Immunother. Cancer 2021, 9, e001615. [Google Scholar] [CrossRef]
- Gurin, D.; Slavik, M.; Hermanova, M.; Selingerova, I.; Kazda, T.; Hendrych, M.; Shatokhina, T.; Vesela, M. The tumor immune microenvironment and its implications for clinical outcome in patients with oropharyngeal squamous cell carcinoma. J. Oral Pathol. Med. 2020, 49, 886–896. [Google Scholar] [CrossRef] [PubMed]
- Nasti, T.H.; Iqbal, O.; Tamimi, I.A.; Geise, J.T.; Katiyar, S.K.; Yusuf, N. Differential Roles of T-cell Subsets in Regulation of Ultraviolet Radiation Induced Cutaneous Photocarcinogenesis. Photochem. Photobiol. 2011, 87, 387–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hladíková, K.; Koucký, V.; Bouček, J.; Laco, J.; Grega, M.; Hodek, M.; Zábrodský, M.; Vošmik, M.; Rozkošová, K.; Vošmiková, H.; 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. [Google Scholar] [CrossRef]
- Fogarty, G.B.; Bayne, M.; Bedford, P.; Bond, R.; Kannourakis, G. Three Cases of Activation of Cutaneous Squamous-cell Carcinoma During Treatment with Prolonged Administration of Rituximab. Clin. Oncol. 2006, 18, 155–156. [Google Scholar] [CrossRef] [PubMed]
- Sautès-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] [PubMed]
- Schumacher, T.N.; Thommen, D.S. Tertiary lymphoid structures in cancer. Science 2022, 375, eabf9419. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Z.; Ding, H.; Lin, Z.-B.; Qiu, S.-H.; Zhang, Y.-R.; Guo, Y.-G.; Chu, X.-D.; I Sam, L.; 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]
- Domblides, C.; Rochefort, J.; Riffard, C.; Panouillot, M.; Lescaille, G.; Teillaud, J.-L.; Mateo, V.; Dieu-Nosjean, M.-C. Tumor-Associated Tertiary Lymphoid Structures: From Basic and Clinical Knowledge to Therapeutic Manipulation. Front. Immunol. 2021, 12, 698604. [Google Scholar] [CrossRef]
- Wu, Y.; Wu, F.; Yan, G.; Zeng, Q.; Jia, N.; Zheng, Z.; Fang, S.; Liu, Y.; Zhang, G.; Wang, X. Features and clinical significance of tertiary lymphoid structure in cutaneous squamous cell carcinoma. J. Eur. Acad. Dermatol. Venereol. 2022, 36, 2043–2050. [Google Scholar] [CrossRef]
- Lerman, I.; Mitchell, D.C.; Richardson, C.T. Human cutaneous B cells: What do we really know? Ann. Transl. Med. 2021, 9, 440–449. [Google Scholar] [CrossRef]
- Anfray, C.; Ummarino, A.; Torres-Andón, F.; Allavena, P. Current Strategies to Target Tumor-Associated-Macrophages to Improve Anti-Tumor Immune Responses. Cells 2019, 9, 46. [Google Scholar] [CrossRef] [Green Version]
- Mantovani, A.; Biswas, S.K.; Galdiero, M.R.; Sica, A.; Locati, M. Macrophage plasticity and polarization in tissue repair and remodelling. J. Pathol. 2013, 229, 176–185. [Google Scholar] [CrossRef]
- Makita, N.; Hizukuri, Y.; Yamashiro, K.; Murakawa, M.; Hayashi, Y. IL-10 enhances the phenotype of M2 macrophages induced by IL-4 and confers the ability to increase eosinophil migration. Int. Immunol. 2015, 27, 131–141. [Google Scholar] [CrossRef] [Green Version]
- Hu, Y.; He, M.-Y.; Zhu, L.-F.; Yang, C.-C.; Zhou, M.-L.; Wang, Q.; Zhang, W.; Zheng, Y.-Y.; Wang, D.-M.; Xu, Z.-Q.; et al. Tumor-associated macrophages correlate with the clinicopathological features and poor outcomes via inducing epithelial to mesenchymal transition in oral squamous cell carcinoma. J. Exp. Clin. Cancer Res. 2016, 35, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seminerio, I.; Kindt, N.; Descamps, G.; Bellier, J.; Lechien, J.R.; Mat, Q.; Pottier, C.; Journé, F.; Saussez, S. High infiltration of CD68+ macrophages is associated with poor prognoses of head and neck squamous cell carcinoma patients and is influenced by human papillomavirus. Oncotarget 2018, 9, 11046–11059. [Google Scholar] [CrossRef] [Green Version]
- Pang, X.; Fan, H.-Y.; Tang, Y.-L.; Wang, S.-S.; Cao, M.-X.; Wang, H.-F.; Dai, L.-L.; Wang, K.; Yu, X.-H.; Wu, J.-B.; et al. Myeloid derived suppressor cells contribute to the malignant progression of oral squamous cell carcinoma. PLoS ONE 2020, 15, e0229089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Youn, J.I.; Nagaraj, S.; Collazo, M.; Gabrilovich, D.I. Subsets of Myeloid-Derived Suppressor Cells in Tumor-Bearing Mice. J. Immunol. 2008, 181, 5791–5802. [Google Scholar] [CrossRef] [Green Version]
- Bruno, A.; Mortara, L.; Baci, D.; Noonan, D.M.; Albini, A. Myeloid Derived Suppressor Cells Interactions With Natural Killer Cells and Pro-angiogenic Activities: Roles in Tumor Progression. Front. Immunol. 2019, 10, 771. [Google Scholar] [CrossRef] [Green Version]
- Vasquez-Dunddel, D.; Pan, F.; Zeng, Q.; Gorbounov, M.; Albesiano, E.; Fu, J.; Blosser, R.L.; Tam, A.J.; Bruno, T.; Zhang, H.; et al. STAT3 regulates arginase-I in myeloid-derived suppressor cells from cancer patients. J. Clin. Investig. 2013, 123, 1580–1589. [Google Scholar] [CrossRef] [Green Version]
- Bottomley, M.J.; Thomson, J.; Harwood, C.; Leigh, I. The Role of the Immune System in Cutaneous Squamous Cell Carcinoma. Int. J. Mol. Sci. 2019, 20, 2009. [Google Scholar] [CrossRef] [Green Version]
- Seddon, A.; Hock, B.; Miller, A.; Frei, L.; Pearson, J.; McKenzie, J.; Simcock, J.; Currie, M. Cutaneous squamous cell carcinomas with markers of increased metastatic risk are associated with elevated numbers of neutrophils and/or granulocytic myeloid derived suppressor cells. J. Dermatol. Sci. 2016, 83, 124–130. [Google Scholar] [CrossRef]
- Du, R.; Lu, K.V.; Petritsch, C.; Liu, P.; Ganss, R.; Passegué, E.; Song, H.; VandenBerg, S.; Johnson, R.S.; Werb, Z.; et al. HIF1α Induces the Recruitment of Bone Marrow-Derived Vascular Modulatory Cells to Regulate Tumor Angiogenesis and Invasion. Cancer Cell 2008, 13, 206–220. [Google Scholar] [CrossRef] [Green Version]
- Komi, D.E.A.; Redegeld, F.A. Role of Mast Cells in Shaping the Tumor Microenvironment. Clin. Rev. Allergy. Immunol. 2020, 58, 313–325. [Google Scholar] [CrossRef] [Green Version]
- Lätti, S.; Leskinen, M.; Shiota, N.; Wang, Y.; Kovanen, P.T.; Lindstedt, K.A. Mast cell-mediated apoptosis of endothelial cells in vitro: A paracrine mechanism involving TNF-α-mediated down-regulation of bcl-2 expression. J. Cell Physiol. 2003, 195, 130–138. [Google Scholar] [CrossRef]
- Gudiseva, S.; Santosh, A.B.R.; Chitturi, R.; Anumula, V.; Poosarla, C.; Baddam, V.R.R. The role of mast cells in oral squamous cell carcinoma. Contemp. Oncol. 2017, 1, 21–29. [Google Scholar] [CrossRef] [Green Version]
- Shrestha, A.; Keshwar, S.; Raut, T. Evaluation of Mast Cells in Oral Potentially Malignant Disorders and Oral Squamous Cell Carcinoma. Int. J. Dent. 2021, 2021, 5609563. [Google Scholar] [CrossRef]
- Yegodayev, K.M.; Novoplansky, O.; Golden, A.; Prasad, M.; Levin, L.; Jagadeeshan, S.; Zorea, J.; Dimitstein, O.; Joshua, B.-Z.; Cohen, L.; et al. TGF-Beta-Activated Cancer-Associated Fibroblasts Limit Cetuximab Efficacy in Preclinical Models of Head and Neck Cancer. Cancers 2020, 12, 339. [Google Scholar] [CrossRef] [Green Version]
- Johansson, A.-C.; Ansell, A.; Jerhammar, F.; Lindh, M.B.; Grénman, R.; Munck-Wikland, E.; Östman, A.; Roberg, K. Cancer-Associated Fibroblasts Induce Matrix Metalloproteinase–Mediated Cetuximab Resistance in Head and Neck Squamous Cell Carcinoma Cells. Mol. Cancer Res. 2012, 10, 1158–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stylianou, A.; Gkretsi, V.; Stylianopoulos, T. Transforming growth factor-β modulates pancreatic cancer associated fibroblasts cell shape, stiffness and invasion. Biochim. Biophys. Acta Gen. Subj. 2018, 1862, 1537–1546. [Google Scholar] [CrossRef]
- Quan, J.; Elhousiny, M.; Johnson, N.W.; Gao, J. Transforming growth factor-β1 treatment of oral cancer induces epithelial-mesenchymal transition and promotes bone invasion via enhanced activity of osteoclasts. Clin. Exp. Metastasis 2013, 30, 659–670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lebrun, J.J. The Dual Role of TGFβ in Human Cancer: From Tumor Suppression to Cancer Metastasis. ISRN Mol. Biol. 2012, 2012, 381428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ludwig, N.; Wieteska, Ł.; Hinck, C.S.; Yerneni, S.S.; Azambuja, J.H.; Bauer, R.J.; Reichert, T.E.; Hinck, A.P.; Whiteside, T.L. Novel TGFβ Inhibitors Ameliorate Oral Squamous Cell Carcinoma Progression and Improve the Antitumor Immune Response of Anti–PD-L1 Immunotherapy. Mol. Cancer Ther. 2021, 20, 1102–1111. [Google Scholar] [CrossRef]
- Mao, X.; Xu, J.; Wang, W.; Liang, C.; Hua, J.; Liu, J.; Zhang, B.; Meng, Q.; Yu, X.; Shi, S. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol. Cancer 2021, 20, 1–30. [Google Scholar] [CrossRef]
- Hanley, C.J.; Mellone, M.; Ford, K.; Thirdborough, S.M.; Mellows, T.; Frampton, S.J.; Smith, D.M.; Harden, E.; Szyndralewiez, C.; Bullock, M.; et al. Targeting the Myofibroblastic Cancer-Associated Fibroblast Phenotype Through Inhibition of NOX4. J. Natl. Cancer Inst. 2018, 110, 109–120. [Google Scholar] [CrossRef] [Green Version]
- Feig, C.; Jones, J.O.; Kraman, M.; Wells, R.J.; Deonarine, A.; Chan, D.S.; Connell, C.M.; Roberts, E.W.; Zhao, Q.; Caballero, O.L.; et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 20212–20217. [Google Scholar] [CrossRef] [Green Version]
- Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased Survival in Pancreatic Cancer with nab-Paclitaxel plus Gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef] [Green Version]
- Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and Its Ligands in Tolerance and Immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [Green Version]
- Flies, D.B.; Sandler, B.J.; Sznol, M.; Chen, L. Blockade of the B7-H1/PD-1 pathway for cancer immunotherapy. Yale J. Biol. Med. 2011, 84, 409–421. [Google Scholar] [PubMed]
- Fisher, J.; Zeitouni, N.; Fan, W.; Samie, F.H. Immune checkpoint inhibitor therapy in solid organ transplant recipients: A patient-centered systematic review. J. Am. Acad. Dermatol. 2020, 82, 1490–1500. [Google Scholar] [CrossRef] [PubMed]
- Long, J.; Qi, Z.; Rongxin, Z. PD-1/PD-L1 pathway blockade works as an effective and practical therapy for cancer immunotherapy. Cancer Biol. Med. 2018, 15, 116. [Google Scholar] [CrossRef] [Green Version]
- Ferrarotto, R.; Bell, D.; Rubin, M.L.; Hutcheson, K.A.; Johnson, J.M.; Goepfert, R.P.; Phan, J.; Elamin, Y.Y.; Torman, D.K.; Warneke, C.L.; et al. Impact of Neoadjuvant Durvalumab with or without Tremelimumab on CD8+ Tumor Lymphocyte Density, Safety, and Efficacy in Patients with Ovropharynx Cancer: CIAO Trial Results. Clin. Cancer Res. 2020, 26, 3211–3219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duhen, R.; Ballesteros-Merino, C.; Frye, A.K.; Tran, E.; Rajamanickam, V.; Chang, S.-C.; Koguchi, Y.; Bifulco, C.B.; Bernard, B.; Leidner, R.S.; et al. Neoadjuvant anti-OX40 (MEDI6469) therapy in patients with head and neck squamous cell carcinoma activates and expands antigen-specific tumor-infiltrating T cells. Nat. Commun. 2021, 12, 1–14. [Google Scholar] [CrossRef] [PubMed]
Type of Gene | Gene | Protein | Function | Mutation Frequency (%) HNSCC/CSCC |
---|---|---|---|---|
Tumor suppressors | TCP53 | p53 | Cell survival and proliferation | 68/76 |
CDKN2A | p16 INK4A | Cell cycle and survival | 20/34 | |
PTEN | PTEN | Cell cycle | 3.1/5.4 | |
FAT1 | Protocadherin FAT1 |
Cell adhesion
(Wnt/β-catenin pathway) | 20.3/55.5 | |
AJUBA | AJUBA |
Cell adhesion
(Wnt/β-catenin pathway) | 6.4/10.7 | |
NOTCH (1) | NOTCH (1) |
Cell adhesion and differentiation
(Delta/Notch pathway) | 16.3/55 | |
KMT2D | Histone-lysine N-methyltransferase KMT2D | Epigenetic regulation | 14.5/41 | |
LRP1B | LRP1 | (Wnt/β-catenin pathway) | 16.7/56.3 | |
Oncogenes | HRAS | HRAS |
Cell proliferation
(MAP/ERK pathway) | 6/12.6 |
PI3KCA | p110 |
Cell proliferation
(PI3K/AKT pathway) | 15.5/13.2 | |
EGFR | EGFR |
Cell proliferation
(PI3K/AKT and MAP/ERK pathway) | 3.7/8.6 | |
TP63 | p63 | Cell cycle | 2.9/9.3 | |
CCND1 | G1–S-specific cyclin D1 | Cell proliferation | 0.6/2.9 | |
TGFBR2 | TGFBR | 4.6/5.7 |
Product Name | Target | Phase | Status | Identifier | |
---|---|---|---|---|---|
OSCC | Anti-EGFR monoclonal antibody | EGFR | II | Recruiting | NCT04508829 |
Cetuximab | EGFR | II | Completed | NCT00933387 | |
Anti-OX40 antibody | OX40 | I | Active, not recruiting | NCT02274155 | |
Cemiplimab | PD-1 | II | Recruiting | NCT04398524 | |
Nivolumab | PD-1 | I/II | Completed | NCT03021993 | |
Sintilimab | PD-1 | II | Not yet recruiting | NCT05000892 | |
Toripalimab | PD-1 | II/III | Recruiting | NCT05125055 | |
Camrelizumab + Apatinib | PD-1 + VEGFR | II | Recruiting | NCT05069857 | |
Durvalumab + Tremelimumab | PD-L1 + CTLA-4 | II | Active, not recruiting | NCT03410615 | |
Bevacizumab | VEGF | I | Active, not recruiting | NCT01552434 | |
CSCC | Erlotinib | EGFR | II | Completed | NCT01059305 |
Cemiplimab | PD-1 | II | Completed | NCT04242173 | |
Cemiplimab | PD-1 | I | Recruiting | NCT03889912 | |
Cemiplimab | PD-1 | I | Recruiting | NCT04339062 | |
Pembrolizumab | PD-1 | II | Recruiting | NCT04808999 | |
Pembrolizumab | PD-1 | II | Active, not recruiting | NCT03284424 | |
Pembrolizumab | PD-1 | III | Recruiting | NCT03833167 | |
Pembrolizumab + Cetuximab | PD-1 + EGFR | II | Unknown | NCT03666325 | |
Atezolizumab | PD-L1 | I | Recruiting | NCT05085496 | |
Avelumab + Cetuximab | PDL-1 + EGFR | II | Recruiting | NCT03944941 |
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. |
© 2023 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
Alonso-Juarranz, M.; Mascaraque, M.; Carrasco, E.; Gracia-Cazaña, T.; De La Sen, O.; Gilaberte, Y.; Gonzalez, S.; Juarranz, Á.; Falahat, F. The Distinctive Features behind the Aggressiveness of Oral and Cutaneous Squamous Cell Carcinomas. Cancers 2023, 15, 3227. https://doi.org/10.3390/cancers15123227
Alonso-Juarranz M, Mascaraque M, Carrasco E, Gracia-Cazaña T, De La Sen O, Gilaberte Y, Gonzalez S, Juarranz Á, Falahat F. The Distinctive Features behind the Aggressiveness of Oral and Cutaneous Squamous Cell Carcinomas. Cancers. 2023; 15(12):3227. https://doi.org/10.3390/cancers15123227
Chicago/Turabian StyleAlonso-Juarranz, Miguel, Marta Mascaraque, Elisa Carrasco, Tamara Gracia-Cazaña, Oscar De La Sen, Yolanda Gilaberte, Salvador Gonzalez, Ángeles Juarranz, and Farzin Falahat. 2023. "The Distinctive Features behind the Aggressiveness of Oral and Cutaneous Squamous Cell Carcinomas" Cancers 15, no. 12: 3227. https://doi.org/10.3390/cancers15123227
APA StyleAlonso-Juarranz, M., Mascaraque, M., Carrasco, E., Gracia-Cazaña, T., De La Sen, O., Gilaberte, Y., Gonzalez, S., Juarranz, Á., & Falahat, F. (2023). The Distinctive Features behind the Aggressiveness of Oral and Cutaneous Squamous Cell Carcinomas. Cancers, 15(12), 3227. https://doi.org/10.3390/cancers15123227