CGE_2024v12n3

Cancer Genetics and Epigenetics 2024, Vol.12, No.3, 144-156 http://medscipublisher.com/index.php/cge 144 Feature Review Open Access Integrative Approaches for Predicting Treatment Response in Advanced Solid Tumors Hui Xu Tianjin Medical University Cancer Institute and Hospital, Hexi, 300210, Tianjin, China Corresponding email: xuhui@163.com Cancer Genetics and Epigenetics, 2024, Vol.12, No.3 doi: 10.5376/cge.2024.12.0017 Received: 28 Apr., 2024 Accepted: 30 May, 2024 Published: 13 Jun., 2024 Copyright © 2024 Xu, This is an open access article published under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Preferred citation for this article: Xu H., 2024, Integrative approaches for predicting treatment response in advanced solid tumors, Cancer Genetics and Epigenetics, 12(3):144-156 (doi: 10.5376/cge.2024.12.0017) Abstract This study explores a comprehensive approach for predicting treatment response in advanced solid tumors, with a focus on the effectiveness of combining molecular, imaging, and clinical data to improve prediction accuracy. Emphasis was placed on the role of advanced computational models, including machine learning and artificial intelligence, in improving predictive capabilities. By integrating diverse data sources, these methods provide a comprehensive understanding of tumor biology and treatment response, ultimately leading to more personalized and effective treatment strategies. The significance of these comprehensive methods for research and clinical practice was also discussed, pointing out their potential to improve patient prognosis. The future directions include molecular analysis, advances in computational algorithms, collaborative research, and data sharing programs, all aimed at improving the accuracy and applicability of predictive models in precision oncology. Keywords Integrative approaches; Predictive models; Advanced solid tumors; Precision oncology; Personalized treatment strategies 1 Introduction Treatment response in advanced solid tumors is a critical factor influencing patient outcomes and overall survival rates. Advanced solid tumors, which include malignancies such as breast, lung, colorectal, and pancreatic cancers, are often characterized by their complex biology and resistance to conventional therapies. The heterogeneous nature of these tumors means that patients may exhibit vastly different responses to the same treatment regimen. This variability poses a significant challenge for oncologists who aim to personalize treatment plans to maximize efficacy and minimize adverse effects. Understanding the mechanisms underlying treatment response is essential for improving therapeutic strategies and patient care (Wheeler et al., 2020). The complexity and heterogeneity of advanced solid tumors necessitate the use of integrative methods to predict treatment response accurately. Integrative approaches combine data from multiple sources, including molecular and genetic profiling, imaging techniques, and clinical data. These methods enable a more comprehensive understanding of the tumor biology and the factors influencing treatment outcomes. For instance, combining genomic sequencing data with imaging biomarkers and clinical variables can provide a multidimensional view of the tumor, facilitating more precise predictions of how it will respond to specific treatments (Uzilov et al., 2016). The integration of machine learning and artificial intelligence further enhances the predictive power of these methods by identifying patterns and correlations that may not be apparent through traditional analysis. Despite the promise of integrative methods, several challenges remain in predicting treatment response in advanced solid tumors. One of the primary challenges is the heterogeneity of the data, which can vary widely in type, quality, and source. Standardizing these diverse data sets for meaningful analysis is a complex task. Additionally, the high-dimensional nature of the data, including genomic, proteomic, and imaging information, requires sophisticated computational tools and substantial computational resources (Davis et al., 2020). Ethical and privacy concerns also arise from the integration and sharing of patient data. Moreover, translating predictive models from research settings to clinical practice involves addressing issues of model validation, reproducibility,

RkJQdWJsaXNoZXIy MjQ4ODYzNQ==