IJCCR_2024v14n1

International Journal of Clinical Case Reports 2024, Vol.14, No.1, 40-47 http://medscipublisher.com/index.php/ijccr 46 Some hereditary diseases result from the functional abnormalities of specific genes. In such cases, gene knockout techniques can be employed to restore the normal function of the gene. For example, in the case of certain monogenic diseases, gene knockout or gene editing technologies can be used to repair or replace the patient's abnormal genes, thereby achieving therapeutic effects. 3.3 Precision medication and prediction of drug side effects The goal of genome-driven personalized medication is to conduct precise treatment and assess the risk of side effects based on individual genomic information. Through genomic analysis and related predictive models, it is possible to better understand the patient's response to drugs and the risk of side effects, thereby optimizing treatment plans, improving treatment outcomes, and reducing unnecessary drug side effects. By analyzing a patient's genome, gene variations related to drug metabolism, drug targets, or drug transport can be identified. These gene variations may impact the speed of drug metabolism, the affinity of drug targets, or the distribution of drugs in the body, thereby influencing both the efficacy and side effects of medications. Through associating these variations with clinical data, it is possible to predict a patient's responsiveness to specific drugs, thereby achieving precision medication (Xu et al., 2019). Certain gene variations may impact the metabolic pathways of drugs, leading to changes in the clearance speed of medications. This could result in either excessively high or low concentrations of drugs in the body, thereby affecting both the efficacy and side effects of the medications. Through genomic analysis, gene variations related to drug metabolism can be identified, allowing for the adjustment of drug dosages to ensure that the concentration of the medication in the patient's body remains within the effective range. Genome-driven personalized drug therapy can also leverage predictive models to forecast drug targets and mechanisms of action. By analyzing a patient's genomic data alongside drug databases, it becomes possible to predict the sensitivity of a patient's targets to specific medications. This aids in determining which drugs are more effective for the patient's condition and avoids the use of drugs insensitive to the patient's genomic characteristics. Genomic information can also be used to predict a patient's sensitivity to drug side effects. Some genetic variations are associated with adverse reactions to specific drugs. By analyzing these variations, it's possible to predict whether a patient is prone to drug-related adverse reactions and make corresponding adjustments in the treatment plan. This helps reduce the risk of adverse drug reactions for patients and enhances the safety of the treatment. 4 Summary and Outlook Genome-driven personalized drug therapy is an advanced and highly promising field that achieves precision medicine and predicts drug side effects by analyzing patients' genomic information. Through genomic analysis, associations between patients' genetic variations and drug responses can be identified, allowing the prediction of patient responsiveness and the risk of side effects to specific drugs. This brings significant breakthroughs to the medical field, enabling the optimization of treatment plans, improving efficacy, and reducing unnecessary drug side effects (Yan and Wang, 2022). The development of genome-driven personalized drug therapy relies on technological advancements and continuous research deepening. The rapid progress and cost reduction of high-throughput sequencing technologies have made acquiring genomic data more convenient and economical, providing more opportunities for personalized treatment. At the same time, people's understanding of the relationship between genes and diseases is constantly deepening, and new gene markers and targeted therapy methods are emerging. However, genome-driven personalized drug therapy still faces challenges. The analysis and interpretation of genomic data require further research and the development of standardized methods to more accurately predict drug efficacy and side effects. The complexity and diversity of individual genomic variations, as well as the interaction between genes and the environment, make the formulation of personalized treatment strategies more intricate. Additionally, issues related to personal privacy and ethics need to be thoroughly considered to ensure the security and compliance of genomic data.

RkJQdWJsaXNoZXIy MjQ4ODYzNQ==