CGE_2024v12n5

Cancer Genetics and Epigenetics 2024, Vol.12, No.5, 234-253 http://medscipublisher.com/index.php/cge 246 In conclusion, addressing the challenges of immune resistance, enhancing immune memory, personalizing immunotherapy approaches, and integrating multi-omics data are critical for advancing the field of immunotherapy in colon cancer. These efforts hold the potential to significantly improve patient outcomes and pave the way for more effective and durable cancer treatments. 8 Concluding Remarks In this study, we have comprehensively examined the critical role of the immune microenvironment in colon cancer therapy. Key findings highlight the importance of various immune cells, cytokines, and the extracellular matrix in modulating tumor progression and response to treatment. We discussed the mechanisms of immune evasion employed by colon cancer cells, including the upregulation of immune checkpoints, recruitment of immunosuppressive cells like tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), and impaired tumor antigen presentation. Therapeutic strategies to modulate the immune microenvironment, such as immune checkpoint inhibitors, adoptive cell transfer therapies, cancer vaccines, and oncolytic viruses, were explored for their potential to enhance anti-tumor immunity and improve clinical outcomes . The insights gathered from recent research underscore the necessity of targeting the immune microenvironment in colon cancer therapy. The efficacy of immune checkpoint inhibitors, for instance, can be significantly enhanced when combined with therapies that modulate the immune microenvironment, such as TAM reprogramming and MDSC targeting agents. Additionally, personalized immunotherapy approaches that utilize biomarkers such as tumor mutational burden (TMB) and microsatellite instability (MSI) can better stratify patients and tailor treatments to achieve optimal outcomes. Future research should continue to focus on overcoming the challenges associated with immune resistance and enhancing immune memory to ensure long-term protection against tumor recurrence. Integrating multi-omics data will be vital for a deeper understanding of the tumor microenvironment and for the development of novel therapeutic targets. Personalized immunotherapy approaches, guided by detailed molecular and immunological profiling, hold great promise for improving patient outcomes. By addressing these challenges and leveraging emerging technologies, we can pave the way for more effective and durable cancer treatments. Acknowledgments Thank you to the peer reviewers for their valuable feedback. Funding This work was supported by the National Natural Science Foundation of China (grant no. 62372141, 82303742). Conflict of Interest Disclosure The authors affirm that this research was conducted without any commercial or financial relationships that could be construed as a potential conflict of interest. References André T., Shiu K.K., Kim T.W., Jensen B.V., Jensen L.H., Punt C., Smith D., Garcia-Carbonero R., Benavides M., Gibbs P., de la Fouchardiere C., Rivera F., Elez E., Bendell J., Le D.T., Yoshino T., Van Cutsem E., Yang P., Farooqui M.Z.H., Marinello P., and Jr Diaz L.A., 2020, Pembrolizumab in microsatellite-instability-high advanced colorectal cancer, The New England Journal of Medicine, 383(23): 2207-2318. Antoniotti C., Rossini D., Pietrantonio F., Catteau A., Salvatore L., Lonardi S., Boquet I., Tamberi S., Marmorino F., Moretto R.,,Ambrosini M., Tamburini E., Tortora G., Passardi A., Bergamo F., Kassambara A., Sbarrato T., Morano F., Ritorto G., Borelli B., Boccaccino A., Conca V., Giordano M., Ugolini C., Fieschi J., Alexia Papadopulos A., Clémentine Massoué C., Aprile G., Antonuzzo L., Fabio Gelsomino F., Erika Martinelli E., Nicoletta Pella N., Gianluca Masi G., Fontanini G., Boni L., Galon J., and Cremolini C., 2022, Upfront FOLFOXIRI plus bevacizumab with or without atezolizumab in the treatment of patients with metastatic colorectal cancer (AtezoTRIBE): a multicentre, open-label, randomised, controlled, phase 2 trial, The Lancet Oncology 23(7): 876-887. https://doi.org/10.1016/S1470-2045(22)00274-1 Aparicio C., Belver M., Enríquez L., Espeso F., Núñez L., Sánchez A., de la Fuente M., and González-Vallinas M., 2021, Cell therapy for colorectal cancer: the promise of chimeric antigen receptor (CAR)-T cells, International Journal of Molecular Sciences, 22(21): 11781. https://doi.org/10.3390/ijms222111781 PMID: 34769211 PMCID: PMC8583883

RkJQdWJsaXNoZXIy MjQ4ODYzNQ==