GET THE APP

Harnessing Multi-omics and Machine Learning for Predictive Modeling of Cancer Drug Response: Advancing Precision Medicine
..

Journal of Bioanalysis & Biomedicine

ISSN: 1948-593X

Open Access

Mini Review - (2024) Volume 16, Issue 2

Harnessing Multi-omics and Machine Learning for Predictive Modeling of Cancer Drug Response: Advancing Precision Medicine

Richard Fernandes*
*Correspondence: Richard Fernandes, Department of Pediatric Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China, Email:
Department of Pediatric Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China

Received: 29-Mar-2024, Manuscript No. jbabm-24-139032; Editor assigned: 01-Apr-2024, Pre QC No. P-139032; Reviewed: 15-Apr-2024, QC No. Q-139032; Revised: 20-Apr-2024, Manuscript No. R-139032; Published: 29-Apr-2024 , DOI: 10.37421/1948-593X.2024.16.432
Citation: Fernandes, Richard. “Harnessing Multi-omics and Machine Learning for Predictive Modeling of Cancer Drug Response: Advancing Precision Medicine.” J Bioanal Biomed 16 (2024): 432.
Copyright: © 2024 Fernandes R. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

The advent of precision medicine has revolutionized oncology by promising tailored therapeutic strategies based on individual patient characteristics. Central to this advancement is the integration of multi-omics data—genomics, transcriptomics, proteomics, and metabolomics— providing a comprehensive understanding of cancer's molecular underpinnings. This study explores the integration of machine learning algorithms for predictive modeling of drug response in cancer patients using a multi-omics approach. By leveraging advanced computational techniques and vast multi-omics datasets, the research aims to enhance the accuracy and efficacy of predicting patient-specific responses to cancer treatments, thereby facilitating personalized medicine. Key challenges such as cancer heterogeneity, high dimensionality of data, and integration of disparate data types are addressed using multi-view learning, data integration frameworks, and feature fusion strategies. Explainable AI methods are employed to interpret the models and uncover potential biomarkers and therapeutic targets. The ultimate goal is to develop a predictive modeling framework for clinical use, guiding treatment decisions and improving patient outcomes.

Keywords

Precision medicine • Oncology • Multi-omics • Genomics

Introduction

The advent of precision medicine has revolutionized the field of oncology, promising tailored therapeutic strategies based on individual patient characteristics. Central to this advancement is the integration of multi-omics data, which includes genomics, transcriptomics, proteomics, and metabolomics. By leveraging the wealth of information from these diverse biological layers, researchers can gain a comprehensive understanding of cancer's molecular underpinnings. This study explores the integration of machine learning algorithms for predictive modeling of drug response in cancer patients using a multi-omics approach. By harnessing the power of advanced computational techniques and vast multi-omics datasets, this research aims to improve the accuracy and efficacy of predicting patient-specific responses to cancer treatments, thereby facilitating personalized medicine [1,2].

Literature Review

The integration of machine learning algorithms with multi-omics data for predicting drug response in cancer patients involves several critical steps. First, multi-omics data from various sources, including DNA sequencing, RNA sequencing, protein expression profiles, and metabolite concentrations, are collected and preprocessed. This preprocessing includes normalization, missing data imputation, and feature selection to reduce dimensionality while retaining relevant biological information [3]. Next, the processed data are used to train machine learning models. Various algorithms, such as random forests, support vector machines, neural networks, and ensemble methods, are employed to capture the complex relationships between multi-omics features and drug response outcomes. These models are rigorously validated using cross-validation techniques and independent test datasets to ensure their robustness and generalizability [4].

Discussion

Key challenges addressed in this study include the heterogeneity of cancer, the high dimensionality of multi-omics data, and the integration of disparate data types. Advanced techniques like multi-view learning, data integration frameworks, and feature fusion strategies are utilized to overcome these challenges. Additionally, explainable AI methods are applied to interpret the models and uncover potential biomarkers and therapeutic targets [5]. By combining multi-omics data and machine learning, this approach not only aims to predict drug responses with high accuracy but also to provide insights into the underlying biological mechanisms driving these responses. The ultimate goal is to develop a predictive modeling framework that can be used in clinical settings to guide treatment decisions and improve patient outcomes [6].

Conclusion

The integration of machine learning algorithms for predictive modeling of drug response in cancer patients using a multi-omics approach represents a significant advancement in personalized medicine. This multi-disciplinary strategy, combining computational power with comprehensive biological data, offers a promising pathway to more accurate and individualized cancer treatment plans. By addressing key challenges such as data heterogeneity and high dimensionality, and utilizing advanced integration techniques, this approach can enhance our understanding of drug responses and lead to the identification of novel biomarkers and therapeutic targets. As this field progresses, the collaboration between data scientists, biologists, and clinicians will be crucial in translating these predictive models into practical tools that improve the precision and efficacy of cancer treatments, ultimately leading to better patient outcomes and advancements in oncology.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Donne, Romain and Amaia Lujambio. "The liver cancer immune microenvironment: Therapeutic implications for hepatocellular carcinoma." Hepatol 77 (2023): 1773-1796.
  2. Google Scholar, Crossref, Indexed at

  3. Younossi, Zobair M., Grace Wong, Quentin M. Anstee and Linda Henry. "The global burden of liver disease." Clin Gastroenterol Hepatol 21 (2023): 1978-1991.
  4. Google Scholar, Crossref, Indexed at

  5. Li, Xin, Pierluigi Ramadori, Dominik Pfister and Marco Seehawer, et al. "The immunological and metabolic landscape in primary and metastatic liver cancer." Nat Rev Cancer 21 (2021): 541-557.
  6. Google Scholar, Crossref, Indexed at

  7. Ma, Yibao, Sarah M. Temkin, Adam M. Hawkridge and Chunqing Guo, et al. "Fatty acid oxidation: An emerging facet of metabolic transformation in cancer." Cancer Lett 435 (2018): 92-100.
  8. Google Scholar, Crossref, Indexed at

  9. Yang, Tingting, Chanping You, Shuhui Meng and Zhengquan Lai, et al. "EBV infection and its regulated metabolic reprogramming in nasopharyngeal tumorigenesis." Front Cell Infect Microbiol 12 (2022): 935205.
  10. Google Scholar, Crossref, Indexed at

  11. Zhu, Yifei, Xinyan Li, Lei Wang and Xiwei Hong, et al. "Metabolic reprogramming and crosstalk of cancer-related fibroblasts and immune cells in the tumor microenvironment." Front Endocrinol 13 (2022): 988295.
  12. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 3099

Journal of Bioanalysis & Biomedicine received 3099 citations as per Google Scholar report

Journal of Bioanalysis & Biomedicine peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward
https://www.olimpbase.org/1937/