Cyclin Dependent Kinase Inhibitor 2A Genetic and Epigenetic Alterations Interfere with Several Immune Components and Predict Poor Clinical Outcome
Abstract
:1. Introduction
2. Materials and Methods
2.1. CDKN2A Differential Expression Analysis
2.2. Assessment of Differential CDKN2A Protein Levels
2.3. Survival Analysis
2.4. Investigation of CDKN2A Genetic Alterations under Tumor Conditions
2.5. Epigenetic Modulation of CDKN2A under Tumor Conditions
2.6. Interference of Altered CDKN2A with the Infiltration and Status of Different Immune Components
2.7. CDKN2A Enrichment Analysis
3. Results
3.1. CDKN2A Upregulation in Tumor Tissue Is Positively Correlated with the Stage, Grade, and Metastasis of Specific Human Tumors
3.2. CDKN2A Differential Protein Level Analysis and IHC Staining
3.3. CDKN2A Overexpression Predicts a Poor Clinical Outcome in Several Human Tumors
3.4. CDKN2A Genetic Alteration Adversely Affects Patient’s Survival
3.5. CDKN2A Hypermethylation in Most of the Analyzed Human Tumors
3.6. Genetic Alterations in CDKN2A Stimulate Immunosuppressive Components and Inhibit Cytotoxic CD8 T Cell Infiltration
3.7. CDKN2A Related Genes Enrichment Analysis
4. Discussion
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Soltan, M.A.; Eldeen, M.A.; Eid, R.A.; Alyamani, N.M.; Alqahtani, L.S.; Albogami, S.; Jafri, I.; Park, M.N.; Alsharif, G.; Fayad, E.; et al. A pan-cancer analysis reveals CHD1L as a prognostic and immunological biomarker in several human cancers. Front. Mol. Biosci. 2023, 10, 1017148. [Google Scholar] [CrossRef] [PubMed]
- Brown, V.L.; Harwood, C.A.; Crook, T.; Cronin, J.G.; Kelsell, D.R.; Proby, C.M. p16INK4a and p14ARF tumor suppressor genes are commonly inactivated in cutaneous squamous cell carcinoma. J. Investig. Dermatol. 2004, 122, 1284–1292. [Google Scholar] [CrossRef] [Green Version]
- Boutelle, A.M.; Attardi, L.D. p53 and Tumor Suppression: It Takes a Network. Trends Cell Biol. 2021, 31, 298–310. [Google Scholar] [CrossRef] [PubMed]
- Soltan, M.A.; Eldeen, M.A.; Sajer, B.H.; Abdelhameed, R.F.A.; Al-Salmi, F.A.; Fayad, E.; Jafri, I.; Ahmed, H.E.M.; Eid, R.A.; Hassan, H.M.; et al. Integration of Chemoinformatics and Multi-Omics Analysis Defines ECT2 as a Potential Target for Cancer Drug Therapy. Biology 2023, 12, 613. [Google Scholar] [CrossRef] [PubMed]
- Park, J.; Lee, J.O.; Lee, M.; Chung, Y.J. AS-CMC: A pan-cancer database of alternative splicing for molecular classification of cancer. Sci. Rep. 2022, 12, 21074. [Google Scholar] [CrossRef]
- Ramos, M.; Geistlinger, L.; Oh, S.; Schiffer, L.; Azhar, R.; Kodali, H.; de Bruijn, I.; Gao, J.; Carey, V.J.; Morgan, M.; et al. Multiomic Integration of Public Oncology Databases in Bioconductor. JCO Clin. Cancer Inform. 2020, 1, 958–971. [Google Scholar] [CrossRef]
- Eid, R.A.; Soltan, M.A.; Eldeen, M.A.; Shati, A.A.; Dawood, S.A.; Eissa, M.; Zaki, M.S.A.; Algahtani, M.; Theyab, A.; Abdel-Daim, M.M.; et al. Assessment of RACGAP1 as a Prognostic and Immunological Biomarker in Multiple Human Tumors: A Multiomics Analysis. Int. J. Mol. Sci. 2022, 23, 14102. [Google Scholar] [CrossRef]
- Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013, 6, pl1. [Google Scholar] [CrossRef] [Green Version]
- Wu, K.; Yi, M.; Qin, S.; Chu, Q.; Zheng, X.; Wu, K. The efficacy and safety of combination of PD-1 and CTLA-4 inhibitors: A meta-analysis. Exp. Hematol. Oncol. 2019, 8, 26. [Google Scholar] [CrossRef] [Green Version]
- Eid, R.A.; Eldeen, M.A.; Soltan, M.A.; Al-Shraim, M.; Aldehri, M.; Alqahtani, L.S.; Alsharif, G.; Albogami, S.; Jafri, I.; Fayad, E.; et al. Integrative analysis of WDR12 as a potential prognostic and immunological biomarker in multiple human tumors. Front. Genet. 2023, 13, 1008502. [Google Scholar] [CrossRef]
- Epstein, R.J. A periodic table for cancer. Future Oncol. 2015, 11, 785–800. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.H.; Wu, C.F.; Rajasekaran, N.; Shin, Y.K. Loss of tumor suppressor gene function in human cancer: An overview. Cell. Physiol. Biochem. 2019, 51, 2647–2693. [Google Scholar] [CrossRef] [PubMed]
- Baxter, E.; Windloch, K.; Gannon, F.; Lee, J.S. Epigenetic regulation in cancer progression. Cell Biosci. 2014, 4, 45. [Google Scholar] [CrossRef] [Green Version]
- Macaluso, M.; Paggi, M.G.; Giordano, A. Genetic and epigenetic alterations as hallmarks of the intricate road to cancer. Oncogene 2003, 22, 6472–6478. [Google Scholar] [CrossRef] [Green Version]
- Sun, W.; Yang, J. Functional mechanisms for human tumor suppressors. J. Cancer 2010, 1, 136. [Google Scholar] [CrossRef]
- Hamid, A.A.; Gray, K.P.; Shaw, G.; MacConaill, L.E.; Evan, C.; Bernard, B.; Loda, M.; Corcoran, N.M.; Van Allen, E.M.; Choudhury, A.D.; et al. Compound Genomic Alterations of TP53, PTEN, and RB1 Tumor Suppressors in Localized and Metastatic Prostate Cancer. Eur. Urol. 2019, 76, 89–97. [Google Scholar] [CrossRef]
- Quetel, L.; Meiller, C.; Assié, J.B.; Blum, Y.; Imbeaud, S.; Montagne, F.; Tranchant, R.; de Wolf, J.; Caruso, S.; Copin, M.C.; et al. Genetic alterations of malignant pleural mesothelioma: Association with tumor heterogeneity and overall survival. Mol. Oncol. 2020, 14, 1207–1223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Q.; Lei, J.H.; Bao, J.; Wang, H.; Hao, W.; Li, L.; Peng, C.; Masuda, T.; Miao, K.; Xu, J.; et al. BRCA1 Deficiency Impairs Mitophagy and Promotes Inflammasome Activation and Mammary Tumor Metastasis. Adv. Sci. 2020, 7, 1903616. [Google Scholar] [CrossRef] [Green Version]
- Morris, L.G.T.; Chan, T.A. Therapeutic targeting of tumor suppressor genes. Cancer 2015, 121, 1357–1368. [Google Scholar] [CrossRef] [Green Version]
- Sherr, C.J. The INK4a/ARF network in tumour suppression. Nat. Rev. Mol. Cell Biol. 2001, 2, 731–737. [Google Scholar] [CrossRef]
- Rubin, S.M. Deciphering the retinoblastoma protein phosphorylation code. Trends Biochem. Sci. 2013, 38, 12–19. [Google Scholar] [CrossRef] [Green Version]
- Jiang, Y.Z.; Ma, D.; Suo, C.; Shi, J.; Xue, M.; Hu, X.; Xiao, Y.; Yu, K.D.; Liu, Y.R.; Yu, Y.; et al. Genomic and Transcriptomic Landscape of Triple-Negative Breast Cancers: Subtypes and Treatment Strategies. Cancer Cell 2019, 35, 428–440. [Google Scholar] [CrossRef] [Green Version]
- Hayward, N.K.; Wilmott, J.S.; Waddell, N.; Johansson, P.A.; Field, M.A.; Nones, K.; Patch, A.M.; Kakavand, H.; Alexandrov, L.B.; Burke, H.; et al. Whole-genome landscapes of major melanoma subtypes. Nature 2017, 545, 175–180. [Google Scholar] [CrossRef] [PubMed]
- Hodis, E.; Watson, I.R.; Kryukov, G.V.; Arold, S.T.; Imielinski, M.; Theurillat, J.P.; Nickerson, E.; Auclair, D.; Li, L.; Place, C.; et al. A landscape of driver mutations in melanoma. Cell 2012, 150, 251–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, J.; Gu, Y.; Liu, J.; Wu, R.; Fu, L.; Zhao, J.; Guan, Y. Coexistence of p16/CDKN2A homozygous deletions and activating EGFR mutations in lung adenocarcinoma patients signifies a poor response to EGFR-TKIs. Lung Cancer 2016, 102, 101–107. [Google Scholar] [CrossRef]
- Delfau-Larue, M.H.; Klapper, W.; Berger, F.; Jardin, F.; Briere, J.; Salles, G.; Casasnovas, O.; Feugier, P.; Haioun, C.; Ribrag, V.; et al. High-dose cytarabine does not overcome the adverse prognostic value of CDKN2A and TP53 deletions in mantle cell lymphoma. Blood 2015, 126, 604–611. [Google Scholar] [CrossRef] [Green Version]
- Cen, L.; Carlson, B.L.; Schroeder, M.A.; Ostrem, J.L.; Kitange, G.J.; Mladek, A.C.; Fink, S.R.; Decker, P.A.; Wu, W.; Kim, J.S.; et al. P16-Cdk4-Rb axis controls sensitivity to a cyclin-dependent kinase inhibitor PD0332991 in glioblastoma xenograft cells. Neuro-Oncology 2012, 14, 870–881. [Google Scholar] [CrossRef] [Green Version]
- Bui, N.Q.; Przybyl, J.; Trabucco, S.E.; Frampton, G.; Hastie, T.; van de Rijn, M.; Ganjoo, K.N. A clinico-genomic analysis of soft tissue sarcoma patients reveals CDKN2A deletion as a biomarker for poor prognosis. Clin. Sarcoma Res. 2019, 9, 12. [Google Scholar] [CrossRef] [Green Version]
- Qiao, J.; Tian, Y.; Cheng, X.; Liu, Z.; Zhou, J.; Gu, L.; Zhang, B.; Zhang, L.; Ji, J.; Xing, R.; et al. CDKN2A Deletion Leading to Hematogenous Metastasis of Human Gastric Carcinoma. Front. Oncol. 2021, 11, 801219. [Google Scholar] [CrossRef]
- Li, T.; Fu, J.; Zeng, Z.; Cohen, D.; Li, J.; Chen, Q.; Li, B.; Liu, X.S. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 2020, 48, W509–W514. [Google Scholar] [CrossRef]
- Tang, Z.; Kang, B.; Li, C.; Chen, T.; Zhang, Z. GEPIA2: An enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 2019, 47, W556–W560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ru, B.; Wong, C.N.; Tong, Y.; Zhong, J.Y.; Zhong, S.S.W.; Wu, W.C.; Chu, K.C.; Wong, C.Y.; Lau, C.Y.; Chen, I.; et al. TISIDB: An integrated repository portal for tumor-immune system interactions. Bioinformatics 2019, 35, 4200–4202. [Google Scholar] [CrossRef] [PubMed]
- Bartha, Á.; Győrffy, B. Tnmplot.Com: A web tool for the comparison of gene expression in normal, tumor and metastatic tissues. Int. J. Mol. Sci. 2021, 22, 2622. [Google Scholar] [CrossRef] [PubMed]
- Chandrashekar, D.S.; Bashel, B.; Balasubramanya, S.A.H.; Creighton, C.J.; Ponce-Rodriguez, I.; Chakravarthi, B.V.S.K.; Varambally, S. UALCAN: A Portal for Facilitating Tumor Subgroup Gene Expression and Survival Analyses. Neoplasia 2017, 19, 649–658. [Google Scholar] [CrossRef]
- Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef]
- Lánczky, A.; Győrffy, B. Web-based survival analysis tool tailored for medical research (KMplot): Development and implementation. J. Med. Internet Res. 2021, 23, e27633. [Google Scholar] [CrossRef]
- Garinis, G.A.; Patrinos, G.P.; Spanakis, N.E.; Menounos, P.G. DNA hypermethylation: When tumour suppressor genes go silent. Hum. Genet. 2002, 111, 115–127. [Google Scholar] [CrossRef] [PubMed]
- Chandrashekar, D.S.; Karthikeyan, S.K.; Korla, P.K.; Patel, H.; Shovon, A.R.; Athar, M.; Netto, G.J.; Qin, Z.S.; Kumar, S.; Manne, U.; et al. UALCAN: An update to the integrated cancer data analysis platform. Neoplasia 2022, 25, 18–27. [Google Scholar] [CrossRef]
- Li, Y.; Ge, D.; Lu, C. The SMART App: An interactive web application for comprehensive DNA methylation analysis and visualization. Epigenet. Chromatin 2019, 12, 71. [Google Scholar] [CrossRef] [Green Version]
- Smyth, M.J.; Dunn, G.P.; Schreiber, R.D. Cancer Immunosurveillance and Immunoediting: The Roles of Immunity in Suppressing Tumor Development and Shaping Tumor Immunogenicity. Adv. Immunol. 2006, 90, 1–50. [Google Scholar]
- Upadhyay, S.; Sharma, N.; Gupta, K.B.; Dhiman, M. Role of immune system in tumor progression and carcinogenesis. J. Cell. Biochem. 2018, 119, 5028–5042. [Google Scholar] [CrossRef]
- Dolina, J.S.; Van Braeckel-Budimir, N.; Thomas, G.D.; Salek-Ardakani, S. CD8+ T Cell Exhaustion in Cancer. Front. Immunol. 2021, 12, 715234. [Google Scholar] [CrossRef]
- Li, L.; Liu, Y.D.; Zhan, Y.T.; Zhu, Y.H.; Li, Y.; Xie, D.; Guan, X.Y. High levels of CCL2 or CCL4 in the tumor microenvironment predict unfavorable survival in lung adenocarcinoma. Thorac. Cancer 2018, 9, 775–784. [Google Scholar] [CrossRef]
- Kadomoto, S.; Izumi, K.; Mizokami, A. The CCL20-CCR6 axis in cancer progression. Int. J. Mol. Sci. 2020, 21, 5186. [Google Scholar] [CrossRef]
- Szklarczyk, D.; Gable, A.L.; Nastou, K.C.; Lyon, D.; Kirsch, R.; Pyysalo, S.; Doncheva, N.T.; Legeay, M.; Fang, T.; Bork, P.; et al. The STRING database in 2021: Customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021, 49, D605–D612. [Google Scholar] [CrossRef] [PubMed]
- Sherman, B.T.; Hao, M.; Qiu, J.; Jiao, X.; Baseler, M.W.; Lane, H.C.; Imamichi, T.; Chang, W. DAVID: A web server for functional enrichment analysis and functional annotation of gene lists (2021 update). Nucleic Acids Res. 2022, 50, W216–W221. [Google Scholar] [CrossRef]
- Eid, R.A.; Alaa Edeen, M.; Shedid, E.M.; Kamal, A.S.S.; Warda, M.M.; Mamdouh, F.; Khedr, S.A.; Soltan, M.A.; Jeon, H.W.; Zaki, M.S.A.; et al. Targeting Cancer Stem Cells as the Key Driver of Carcinogenesis and Therapeutic Resistance. Int. J. Mol. Sci. 2023, 24, 1786. [Google Scholar] [CrossRef]
- Serrano, J.; Goebel, S.U.; Peghini, P.L.; Lubensky, I.A.; Gibril, F.; Jensen, R.T. Alterations in the p16INK4a/CDKN2A tumor suppressor gene in gastrinomas. J. Clin. Endocrinol. Metab. 2000, 85, 4146–4156. [Google Scholar] [CrossRef]
- Zhao, R.; Choi, B.Y.; Lee, M.H.; Bode, A.M.; Dong, Z. Implications of Genetic and Epigenetic Alterations of CDKN2A (p16INK4a) in Cancer. EBioMedicine 2016, 8, 30–39. [Google Scholar] [CrossRef] [Green Version]
- Ladanyi, M. Implications of P16/CDKN2A deletion in pleural mesotheliomas. Lung Cancer 2005, 49, S95–S98. [Google Scholar] [CrossRef]
- Alhejaily, A.; Day, A.G.; Feilotter, H.E.; Baetz, T.; LeBrun, D.P. Inactivation of the CDKN2A tumor-suppressor gene by deletion or methylation is common at diagnosis in follicular lymphoma and associated with poor clinical outcome. Clin. Cancer Res. 2014, 20, 1676–1686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roy, S.; LaFramboise, W.A.; Liu, T.C.; Cao, D.; Luvison, A.; Miller, C.; Lyons, M.A.; O’Sullivan, R.J.; Zureikat, A.H.; Hogg, M.E.; et al. Loss of Chromatin-Remodeling Proteins and/or CDKN2A Associates with Metastasis of Pancreatic Neuroendocrine Tumors and Reduced Patient Survival Times. Gastroenterology 2018, 154, 2060–2063.e8. [Google Scholar] [CrossRef]
- Padhi, S.S.; Roy, S.; Kar, M.; Saha, A.; Roy, S.; Adhya, A.; Baisakh, M.; Banerjee, B. Role of CDKN2A/p16 expression in the prognostication of oral squamous cell carcinoma. Oral Oncol. 2017, 73, 27–35. [Google Scholar] [CrossRef]
- Sievers, P.; Hielscher, T.; Schrimpf, D.; Stichel, D.; Reuss, D.E.; Berghoff, A.S.; Neidert, M.C.; Wirsching, H.G.; Mawrin, C.; Ketter, R.; et al. CDKN2A/B homozygous deletion is associated with early recurrence in meningiomas. Acta Neuropathol. 2020, 140, 409–413. [Google Scholar] [CrossRef] [PubMed]
- Guyot, A.; Duchesne, M.; Robert, S.; Lia, A.S.; Derouault, P.; Scaon, E.; Lemnos, L.; Salle, H.; Durand, K.; Labrousse, F. Analysis of CDKN2A gene alterations in recurrent and non-recurrent meningioma. J. Neurooncol. 2019, 145, 449–459. [Google Scholar] [CrossRef]
- Sargen, M.R.; Merrill, S.L.; Chu, E.Y.; Nathanson, K.L. CDKN2A mutations with p14 loss predisposing to multiple nerve sheath tumours, melanoma, dysplastic naevi and internal malignancies: A case series and review of the literature. Br. J. Dermatol. 2016, 175, 785–789. [Google Scholar] [CrossRef] [PubMed]
- Cicenas, J.; Kvederaviciute, K.; Meskinyte, I.; Meskinyte-Kausiliene, E.; Skeberdyte, A. KRAS, TP53, CDKN2A, SMAD4, BRCA1, and BRCA2 mutations in pancreatic cancer. Cancers 2017, 9, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heaton, C.M.; Durr, M.L.; Tetsu, O.; Van Zante, A.; Wang, S.J. TP53 and CDKN2a mutations in never-smoker oral tongue squamous cell carcinoma. Laryngoscope 2014, 124, E267–E273. [Google Scholar] [CrossRef]
- Helgadottir, H.; Hoiom, V.; Tuominen, R.; Nielsen, K.; Jonsson, G.; Olsson, H.; Hansson, J. Germline CDKN2A mutation status and survival in familial melanoma cases. J. Natl. Cancer Inst. 2016, 108, djw135. [Google Scholar] [CrossRef] [Green Version]
- Betti, M.; Aspesi, A.; Biasi, A.; Casalone, E.; Ferrante, D.; Ogliara, P.; Gironi, L.C.; Giorgione, R.; Farinelli, P.; Grosso, F.; et al. CDKN2A and BAP1 germline mutations predispose to melanoma and mesothelioma. Cancer Lett. 2016, 378, 120–130. [Google Scholar] [CrossRef]
- Trietsch, M.D.; Spaans, V.M.; Ter Haar, N.T.; Osse, E.M.; Peters, A.A.W.; Gaarenstroom, K.N.; Fleuren, G.J. CDKN2A(p16) and HRAS are frequently mutated in vulvar squamous cell carcinoma. Gynecol. Oncol. 2014, 135, 149–155. [Google Scholar] [CrossRef] [Green Version]
- Ng, J.M.K.; Yu, J. Promoter hypermethylation of tumour suppressor genes as potential biomarkers in colorectal cancer. Int. J. Mol. Sci. 2015, 16, 2472–2496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arya, A.K.; Bhadada, S.K.; Singh, P.; Sachdeva, N.; Saikia, U.N.; Dahiya, D.; Behera, A.; Bhansali, A.; Rao, S.D. Promoter hypermethylation inactivates CDKN2A, CDKN2B and RASSF1A genes in sporadic parathyroid adenomas. Sci. Rep. 2017, 7, 3123. [Google Scholar] [CrossRef] [Green Version]
- Spitzwieser, M.; Entfellner, E.; Werner, B.; Pulverer, W.; Pfeiler, G.; Hacker, S.; Cichna-Markl, M. Hypermethylation of CDKN2A exon 2 in tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients. BMC Cancer 2017, 17, 260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liau, J.Y.; Liao, S.L.; Hsiao, C.H.; Lin, M.C.; Chang, H.C.; Kuo, K.T. Hypermethylation of the CDKN2A gene promoter is a frequent epigenetic change in periocular sebaceous carcinoma and is associated with younger patient age. Hum. Pathol. 2014, 45, 533–539. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Wang, X.-b.; Qiu, X.-p.; Zhang, S.; Wang, C.; Zheng, F. CDKN2A promoter methylation and hepatocellular carcinoma risk: A meta-analysis. Clin. Res. Hepatol. Gastroenterol. 2018, 42, 529–541. [Google Scholar] [CrossRef]
- Mei, X.; Cheng, M.; Chen, W.; Wu, X.; Xie, M. The hypermethylation of the CDKN2A and CHFR promoter region is a key regulatory mechanism of CDKN2A and CHFR expression in esophageal squamous cell carcinoma. Transl. Cancer Res. 2019, 8, 770–778. [Google Scholar] [CrossRef] [PubMed]
- Pierini, S.; Jordanov, S.H.; Mitkova, A.V.; Chalakov, I.J.; Melnicharov, M.B.; Kunev, K.V.; Mitev, V.I.; Kaneva, R.P.; Goranova, T.E. Promoter hypermethylation of CDKN2A, MGMT, MLH1, and DAPK genes in laryngeal squamous cell carcinoma and their associations with clinical profiles of the patients. Head Neck 2014, 36, 1103–1108. [Google Scholar] [CrossRef]
- Wijetunga, N.A.; Belbin, T.J.; Burk, R.D.; Whitney, K.; Abadi, M.; Greally, J.M.; Einstein, M.H.; Schlecht, N.F. Novel epigenetic changes in CDKN2A are associated with progression of cervical intraepithelial neoplasia. Gynecol. Oncol. 2016, 142, 566–573. [Google Scholar] [CrossRef] [Green Version]
- Tang, B.; Li, Y.; Qi, G.; Yuan, S.; Wang, Z.; Yu, S.; Li, B.; He, S. Clinicopathological Significance of CDKN2A Promoter Hypermethylation Frequency with Pancreatic Cancer. Sci. Rep. 2015, 5, srep13563. [Google Scholar] [CrossRef] [Green Version]
- Lei, X.; Lei, Y.; Li, J.K.; Du, W.X.; Li, R.G.; Yang, J.; Li, J.; Li, F.; Tan, H.B. Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Lett. 2020, 470, 126–133. [Google Scholar] [CrossRef] [PubMed]
- Kishore, C.; Bhadra, P. Current advancements and future perspectives of immunotherapy in colorectal cancer research. Eur. J. Pharmacol. 2021, 893, 173819. [Google Scholar] [CrossRef] [PubMed]
- Siddiqui, I.; Schaeuble, K.; Chennupati, V.; Fuertes Marraco, S.A.; Calderon-Copete, S.; Pais Ferreira, D.; Carmona, S.J.; Scarpellino, L.; Gfeller, D.; Pradervand, S.; et al. Intratumoral Tcf1 + PD-1 + CD8 + T Cells with Stem-like Properties Promote Tumor Control in Response to Vaccination and Checkpoint Blockade Immunotherapy. Immunity 2019, 50, 195–211.e10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, B.; Chan, H.L.; Chen, P. Immune Checkpoint Inhibitors: Basics and Challenges. Curr. Med. Chem. 2017, 26, 3009–3025. [Google Scholar] [CrossRef]
- Mognol, G.P.; Spreafico, R.; Wong, V.; Scott-Browne, J.P.; Togher, S.; Hoffmann, A.; Hogan, P.G.; Rao, A.; Trifari, S. Exhaustion-associated regulatory regions in CD8+ tumor-infiltrating T cells. Proc. Natl. Acad. Sci. USA 2017, 114, E2776–E2785. [Google Scholar] [CrossRef]
- Budimir, N.; Thomas, G.D.; Dolina, J.S.; Salek-Ardakani, S. Reversing T-cell Exhaustion in Cancer: Lessons Learned from PD-1/PD-L1 Immune Checkpoint Blockade. Cancer Immunol. Res. 2022, 10, 146–153. [Google Scholar] [CrossRef]
- Hajifathali, A.; Parkhideh, S.; Kazemi, M.H.; Chegeni, R.; Roshandel, E.; Gholizadeh, M. Immune checkpoints in hematologic malignancies: What made the immune cells and clinicians exhausted! J. Cell. Physiol. 2020, 235, 9080–9097. [Google Scholar] [CrossRef]
- Zarour, H.M. Reversing T-cell dysfunction and exhaustion in cancer. Clin. Cancer Res. 2016, 22, 1856–1864. [Google Scholar] [CrossRef] [Green Version]
- Hua, F.; Tian, Y. CCL4 promotes the cell proliferation, invasion and migration of endometrial carcinoma by targeting the VEGF-A signal pathway. Int. J. Clin. Exp. Pathol. 2017, 10, 11288–11299. [Google Scholar]
- Wang, Y.; Liu, T.; Yang, N.; Xu, S.; Li, X.; Wang, D. Hypoxia and macrophages promote glioblastoma invasion by the CCL4-CCR5 axis. Oncol. Rep. 2016, 36, 3522–3528. [Google Scholar] [CrossRef] [Green Version]
- Nandi, B.; Shapiro, M.; Samur, M.K.; Pai, C.; Frank, N.Y.; Yoon, C.; Prabhala, R.H.; Munshi, N.C.; Gold, J.S. Stromal CCR6 drives tumor growth in a murine transplantable colon cancer through recruitment of tumor-promoting macrophages. Oncoimmunology 2016, 5, e1189052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boyle, S.T.; Faulkner, J.W.; McColl, S.R.; Kochetkova, M. The chemokine receptor CCR6 facilitates the onset of mammary neoplasia in the MMTV-PyMT mouse model via recruitment of tumor-promoting macrophages. Mol. Cancer 2015, 14, 115. [Google Scholar] [CrossRef] [Green Version]
- Zhu, C.-C.; Chen, C.; Xu, Z.-Q.; Zhao, J.-K.; Ou, B.-C.; Sun, J.; Zheng, M.-H.; Zong, Y.-P.; Lu, A.-G. CCR6 promotes tumor angiogenesis via the AKT/NF-κB/VEGF pathway in colorectal cancer. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 387–397. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.G.; Song, B.T.; Liu, F.J.; Sun, D.; Wang, K.X.; Qu, H. CCR6 overexpression predicted advanced biological behaviors and poor prognosis in patients with gastric cancer. Clin. Transl. Oncol. 2016, 18, 700–707. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Ke, F.; Xu, Z.; Liu, Z.; Zhang, L.; Yan, S.; Wang, Z.; Wang, H.; Wang, H. CCR6 is a prognostic marker for overall survival in patients with colorectal cancer, and its overexpression enhances metastasis in vivo. PLoS ONE 2014, 9, e101137. [Google Scholar] [CrossRef] [Green Version]
- Xie, D.; Pei, Q.; Li, J.; Wan, X.; Ye, T. Emerging Role of E2F Family in Cancer Stem Cells. Front. Oncol. 2021, 11, 723137. [Google Scholar] [CrossRef]
- Sun, Y.; Cheng, Z.; Liu, S. MCM2 in human cancer: Functions, mechanisms, and clinical significance. Mol. Med. 2022, 28, 128. [Google Scholar]
- Yates, K.B.; Tonnerre, P.; Martin, G.E.; Gerdemann, U.; Al Abosy, R.; Comstock, D.E.; Weiss, S.A.; Wolski, D.; Tully, D.C.; Chung, R.T.; et al. Epigenetic scars of CD8+ T cell exhaustion persist after cure of chronic infection in humans. Nat. Immunol. 2021, 22, 1020–1029. [Google Scholar] [CrossRef]
- Abdel-Hakeem, M.S.; Manne, S.; Beltra, J.C.; Stelekati, E.; Chen, Z.; Nzingha, K.; Ali, M.A.; Johnson, J.L.; Giles, J.R.; Mathew, D.; et al. Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation. Nat. Immunol. 2021, 22, 1008–1019. [Google Scholar] [CrossRef]
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
Soltan, M.A.; Alhanshani, A.A.; Shati, A.A.; Alqahtani, Y.A.; Alshaya, D.S.; Alharthi, J.; Altalhi, S.A.; Fayad, E.; Zaki, M.S.A.; Eid, R.A. Cyclin Dependent Kinase Inhibitor 2A Genetic and Epigenetic Alterations Interfere with Several Immune Components and Predict Poor Clinical Outcome. Biomedicines 2023, 11, 2254. https://doi.org/10.3390/biomedicines11082254
Soltan MA, Alhanshani AA, Shati AA, Alqahtani YA, Alshaya DS, Alharthi J, Altalhi SA, Fayad E, Zaki MSA, Eid RA. Cyclin Dependent Kinase Inhibitor 2A Genetic and Epigenetic Alterations Interfere with Several Immune Components and Predict Poor Clinical Outcome. Biomedicines. 2023; 11(8):2254. https://doi.org/10.3390/biomedicines11082254
Chicago/Turabian StyleSoltan, Mohamed A., Ahmad A. Alhanshani, Ayed A. Shati, Youssef A. Alqahtani, Dalal Sulaiman Alshaya, Jawaher Alharthi, Sarah Awwadh Altalhi, Eman Fayad, Mohamed Samir A. Zaki, and Refaat A. Eid. 2023. "Cyclin Dependent Kinase Inhibitor 2A Genetic and Epigenetic Alterations Interfere with Several Immune Components and Predict Poor Clinical Outcome" Biomedicines 11, no. 8: 2254. https://doi.org/10.3390/biomedicines11082254
APA StyleSoltan, M. A., Alhanshani, A. A., Shati, A. A., Alqahtani, Y. A., Alshaya, D. S., Alharthi, J., Altalhi, S. A., Fayad, E., Zaki, M. S. A., & Eid, R. A. (2023). Cyclin Dependent Kinase Inhibitor 2A Genetic and Epigenetic Alterations Interfere with Several Immune Components and Predict Poor Clinical Outcome. Biomedicines, 11(8), 2254. https://doi.org/10.3390/biomedicines11082254