CGE_2024v12n3

Cancer Genetics and Epigenetics 2024, Vol.12, No.3, 115-124 http://medscipublisher.com/index.php/cge 122 prevalence of specific gene methylation patterns and histone modifications may vary between populations, affecting the efficacy of targeted therapies (Qu et al., 2013). Large-scale epidemiological studies can also identify environmental and lifestyle factors that interact with genetic predispositions to influence gastric cancer risk. By integrating data from global studies, researchers can develop more effective prevention and treatment strategies tailored to specific populations, ultimately improving patient outcomes on a global scale. The future of gastric cancer research lies in the integration of emerging technologies, multi-omics approaches, and global epidemiological studies. These strategies will provide a deeper understanding of the genetic and epigenetic mechanisms underlying gastric cancer, paving the way for innovative therapeutic interventions and personalized medicine. 8 Concluding Remarks The research on genetic and epigenetic regulation in gastric cancer (GC) has revealed significant insights into the mechanisms driving this malignancy. Key findings include the identification of somatic mutations in epigenetic regulation genes such as KMT2D, KMT2C, ARID1A, and CHD7, which are associated with reduced overall survival and metastasis in GC patients. DNA methylation, particularly in promoter regions, has been highlighted as a critical epigenetic modification leading to the inactivation of tumor suppressor genes and the activation of oncogenes, contributing to gastric carcinogenesis. Additionally, the PI3K/Akt/mTOR signaling pathway has been identified as a significant player in GC pathogenesis, with potential for targeted pharmacologic interventions. The role of BET inhibitors in targeting epigenetic regulators like BRD4 has also shown promise as a therapeutic approach. Continued research in the field of genetic and epigenetic regulation in gastric cancer is crucial for several reasons. Firstly, understanding the intricate mechanisms of epigenetic alterations can lead to the identification of novel biomarkers for early diagnosis and prognosis, which is essential given the typically late diagnosis and poor prognosis associated with GC. Secondly, exploring the therapeutic potential of targeting epigenetic modifications, such as DNA methylation and histone modifications, can pave the way for the development of more effective and personalized treatment strategies. Furthermore, investigating the interplay between genetic mutations and epigenetic changes can provide a comprehensive understanding of GC pathogenesis, potentially leading to the discovery of new drug targets and the improvement of existing therapies. In conclusion, the integration of genetic and epigenetic research holds significant promise for advancing our understanding and treatment of gastric cancer. The identification of key epigenetic alterations and their impact on gene expression and tumor behavior underscores the potential of epigenetic therapies in improving patient outcomes. As research progresses, it is imperative to continue exploring the molecular underpinnings of GC and to translate these findings into clinical practice. By doing so, we can move closer to achieving more effective diagnostic tools and therapeutic options, ultimately improving the prognosis and quality of life for patients with gastric cancer. Acknowledgments I extend my sincere thanks to two anonymous peer reviewers for their invaluable feedback on the study. Conflict of Interest Disclosure The author affirms that this research was conducted without any commercial or financial relationships that could be construed as a potential conflict of interest. References Askari N., Esfahani B.S., Parvizpour S., Shafieipour S., and Hadizadeh M., 2023, Long non-coding RNAs as potential biomarkers or therapeutic targets in gastric cancer, Gastroenterology and Hepatology from Bed to Bench, 16(3): 297. Biswas S., and Rao C.M., 2017, Epigenetics in cancer: fundamentals and beyond, Pharmacology and Therapeutics, 173: 118-134. https://doi.org/10.1016/j.pharmthera.2017.02.011 Cai W.Y., Dong Z.N., Fu X.T., Lin L.Y., Wang L., Ye G.D., Luo Q., and Chen Y.C., 2020, Identification of a tumor microenvironment-relevant gene set-based prognostic signature and related therapy targets in gastric cancer, Theranostics, 10(19): 8633. https://doi.org/10.7150/thno.47938

RkJQdWJsaXNoZXIy MjQ4ODYzNQ==