Molecular Genetics in Precision Medicine: From Bench to Bedside

*Corresponding author: 
           Darand Martin, 
           Department of Medical Genetics and Genomic Medicine, University of Campinas (UNICAMP), Barão Geraldo, Campinas - SP, Brazil, 
           E-mail: martin.darand@uniamp.br 

Received date: February 01, 2025; Accepted date: February 20, 2025; Published date: February 28, 2025

Citation:  Martin D (2025) Molecular Genetics in Precision Medicine: From Bench to Bedside. J Mol Genet Med. 9 No.1:003

Visit for more related articles at Journal of Molecular Genetics and Medicine

Introduction

The advent of precision medicine has transformed the landscape of modern healthcare by shifting the focus from generalized treatment approaches to strategies tailored to an individualâ??s genetic and molecular profile. Molecular genetics lies at the core of this paradigm, providing insights into genetic variations, mutations, and regulatory networks that shape disease risk, progression, and therapeutic response. Through genomic sequencing, transcriptomics, and molecular diagnostics, clinicians can now identify patient-specific biomarkers that inform diagnosis, predict outcomes, and guide treatment choices with unprecedented accuracy. From oncology to cardiology and rare genetic disorders, the translation of molecular genetics into clinical practice is reshaping the way diseases are understood and managed. Precision medicine not only emphasizes early detection and targeted therapies but also integrates data from pharmacogenomics, gene editing, and molecular diagnostics to improve patient outcomes. Bridging the gap between laboratory discoveries and bedside applications requires multidisciplinary collaboration, robust clinical trials, and technological innovations that ensure accessibility, safety, and cost-effectiveness [1].

Description

Molecular genetics provides the fundamental blueprint for precision medicine by revealing genetic determinants that underlie health and disease. Techniques such as Whole-Genome Sequencing (WGS), whole-exome sequencing (WES), and Genome-Wide Association Studies (GWAS) have identified variants associated with susceptibility to cancers, autoimmune disorders, cardiovascular diseases, and metabolic conditions. For instance, mutations in BRCA1 and BRCA2 genes are widely recognized for increasing the risk of breast and ovarian cancers, guiding preventive measures and therapeutic interventions. Similarly, genetic profiling in cardiovascular disease has identified variants in PCSK9 and LDLR genes that influence cholesterol regulation, leading to the development of targeted lipid-lowering therapies. These examples illustrate how genetic discoveries move from bench research to practical, patient-centered solutions [2].

The translation of genetic discoveries into clinical applications relies heavily on pharmacogenomics, which studies how genetic variation affects drug metabolism and response. Genes such as CYP2D6, CYP2C9, and VKORC1 are known to modulate how individuals metabolize antidepressants, anticoagulants, and chemotherapeutics. By incorporating pharmacogenomic data into treatment protocols, clinicians can optimize drug choice and dosage, minimizing adverse reactions and improving therapeutic efficacy. For example, targeted cancer therapies such as tyrosine kinase inhibitors are prescribed based on the presence of EGFR or ALK mutations, while immune checkpoint inhibitors are guided by biomarkers like PD-L1 expression and microsatellite instability. This integration of molecular genetics ensures treatments are not only more effective but also safer for patients [3].

Beyond pharmacogenomics, advancements in gene editing technologies such as CRISPR-Cas9 have opened new frontiers in precision medicine. These tools enable the correction of disease-causing mutations at their source, offering potential cures for genetic disorders such as sickle cell anemia, beta-thalassemia, and Duchenne muscular dystrophy. Early clinical trials have demonstrated promising results, with some patients achieving sustained remission or functional cures. In addition, molecular therapies such as Antisense Oligonucleotides (ASOs) and RNA interference (RNAi) are being developed to modulate gene expression, expanding therapeutic possibilities for conditions once considered untreatable. As these innovations progress, the challenge lies in ensuring safety, scalability, and ethical oversight in their clinical adoption [4].

The integration of molecular genetics into healthcare also requires robust data infrastructure, bioinformatics tools, and interdisciplinary collaboration. Large-scale initiatives such as the All of Us Research Program and the UK Biobank have generated massive genomic datasets, accelerating discoveries and their clinical translation. Artificial intelligence and machine learning are increasingly employed to analyze complex genomic data and predict disease risk or treatment outcomes. Furthermore, ethical considerations including data privacy, equitable access, and informed consent remain central to the successful implementation of precision medicine. Addressing these challenges ensures that the benefits of molecular genetics extend beyond specialized centers to broader patient populations, closing the gap between bench discoveries and bedside practice. One of the most groundbreaking developments in precision medicine is Casgevy (exagamglogene autotemcel): the first CRISPR/Cas9-based gene-editing therapy approved by the U.S. FDA. In December 2023, Casgevy was authorized for treating sickle cell disease (SCD) in patients aged 12 and older. The therapy involves editing a patientâ??s own hematopoietic stem cells to increase fetal hemoglobin and reduce vaso-occlusive crises. Shortly thereafter, in January 2024, its approval expanded to include transfusion-dependent beta thalassemia (TDT) [5].

Conclusion

Molecular genetics has become a cornerstone of precision medicine, enabling a shift toward more personalized, predictive, and preventive healthcare. From identifying genetic risk factors and guiding pharmacogenomic decisions to pioneering gene editing therapies, the integration of genetic insights into clinical care has already improved patient outcomes across diverse medical fields. The journey from bench to bedside highlights the importance of interdisciplinary collaboration, technological innovation, and ethical stewardship in realizing the full potential of genetic medicine. As advances continue, precision medicine promises to redefine the future of healthcare, offering targeted solutions that align with each patientâ??s unique genetic and molecular profile. None.

Acknowledgment

None.

Conflict of Interest

None.

References

  1. Taverna S, Cammarata G, Colomba P, Sciarrino S, Zizzo C, et al. (2020). Pompe disease: Pathogenesis, molecular genetics and diagnosis. Aging 12: 15856.

                          Google Scholar Cross Ref Indexed at

  1. Myerowitz R, Puertollano R, Raben N (2021). Impaired autophagy: The collateral damage of lysosomal storage disorders. EBioMedicine: 63.

                          Google Scholar Cross Ref Indexed at

  1. Farah BL, Yen PM, Koeberl DD (2020). Links between autophagy and disorders of glycogen metabolism–perspectives on pathogenesis and possible treatments. Mol Genet Metab 129: 3-12.

                          Google Scholar Cross Ref Indexed at

  1. Wang H, Zhang C, Dong Z, Zhu X, Zheng X, et al. (2024). Using an in vivo mouse model to determine the exclusion criteria of preexisting anti-AAV9 neutralizing antibody titer of pompe disease patients in clinical trials. Viruses 16: 400.

                          Google Scholar Cross Ref Indexed at

  1. Liang Q, Vlaar EC, Pijnenburg JM, Rijkers E, Demmers JA, et al. (2024). Lentiviral gene therapy with IGF2-tagged GAA normalizes the skeletal muscle proteome in murine Pompe disease. J Proteom 291: 105037.

                          Google Scholar Cross Ref Indexed at

Select your language of interest to view the total content in your interested language

Viewing options

Flyer image

Share This Article