GET THE APP

Genomic Medicine in Oncology: Harnessing Clinical Genomics for Cancer Treatment
..

Journal of Bioengineering & Biomedical Science

ISSN: 2155-9538

Open Access

Brief Report - (2023) Volume 13, Issue 2

Genomic Medicine in Oncology: Harnessing Clinical Genomics for Cancer Treatment

Balyasin Andrey*
*Correspondence: Balyasin Andrey, Department of Medicinal Chemistry and Masonic Cancer Center, University of Minnesota, Minneapolis, USA, Email:
Department of Medicinal Chemistry and Masonic Cancer Center, University of Minnesota, Minneapolis, USA

Received: 27-Mar-2023, Manuscript No. jbbs-23-109792; Editor assigned: 29-Mar-2023, Pre QC No. P-109792; Reviewed: 13-Apr-2023, QC No. Q-109792; Revised: 18-Apr-2023, Manuscript No. R-109792; Published: 25-Apr-2023 , DOI: 10.37421/2155-9538.2023.13.351
Citation: Andrey, Balyasin. “Genomic Medicine in Oncology: Harnessing Clinical Genomics for Cancer Treatment.” J Bioengineer & Biomedical Sci 13 (2023): 351.
Copyright: © 2023 Andrey B. 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.

Introduction

Cancer is a complex and heterogeneous disease that arises due to genetic alterations and mutations in the DNA of cells. The advent of genomic medicine has revolutionized cancer research and treatment by providing unprecedented insights into the molecular basis of cancer. Clinical genomics, the application of genomic information in healthcare, plays a crucial role in oncology by enabling personalized and targeted therapies, improving patient outcomes, and paving the way for precision cancer medicine. This article explores how genomic medicine is transforming cancer treatment and the challenges and opportunities it presents [1].

Description

Genomic profiling involves analyzing the genetic makeup of cancer cells to identify specific mutations and alterations that drive tumor growth and progression. Techniques such as Next-Generation Sequencing (NGS) have enabled comprehensive profiling of cancer genomes, leading to the identification of driver mutations and potential therapeutic targets. We discuss the importance of genomic profiling in cancer diagnosis, its impact on understanding tumor heterogeneity and the implications for treatment decision-making. One of the most significant advancements in cancer treatment is the development of targeted therapies. These drugs are designed to specifically target the molecular abnormalities present in cancer cells, sparing healthy cells from damage and reducing treatment-related side effects. Genomic medicine has facilitated the identification of actionable mutations that can be targeted by these therapies. We explore the success stories of targeted therapies in different cancer types and the challenges in overcoming resistance mechanisms [2].

Immunogenomics combines the fields of genomics and immunology to understand the interaction between the immune system and cancer cells. Genomic profiling of tumors can help identify tumor-specific antigens that can be recognized by the immune system. This knowledge has paved the way for immunotherapies, such as immune checkpoint inhibitors, which enhance the body's immune response against cancer cells. One of the key applications of genomic medicine in oncology is the identification of driver mutations. Driver mutations are genetic alterations that play a causative role in tumorigenesis by promoting cell proliferation, inhibiting apoptosis, or facilitating other hallmarks of cancer. By distinguishing driver mutations from passenger mutations (random genetic changes with no functional consequence), clinicians can prioritize specific targets for treatment [3].

For instance, the discovery of mutations in the Epidermal Growth Factor Receptor (EGFR) gene in non-small cell lung cancer has led to the development of targeted therapies like EGFR tyrosine kinase inhibitors, resulting in improved outcomes for patients with these mutations. We delve into the role of immunogenomics in cancer treatment, the concept of tumor immunogenicity, and the potential of combination therapies for improved outcomes. Genomic medicine also contributes to the emerging field of immunotherapy, which harnesses the body's immune system to target and eliminate cancer cells. The identification of tumor-specific antigens through genomic analysis enables the development of personalized cancer vaccines and adoptive T cell therapies. These approaches leverage the patient's own immune system to recognize and attack cancer cells with precision, potentially leading to long-lasting remissions. Traditional tissue biopsies provide valuable information about the genomic landscape of cancer cells. However, they can be invasive and may not always capture the full genomic heterogeneity of tumors. Liquid biopsies, on the other hand, offer a non-invasive and dynamic approach to monitor cancer genomics through the analysis of circulating tumor DNA (ctDNA) and other biomarkers in blood samples. Despite significant advancements, several challenges remain in the clinical implementation of genomic medicine in oncology. These include data interpretation, standardization of genomic testing, ethical considerations, and access to genomic profiling for all patients. We also explore ongoing research and future directions in cancer genomics, such as single-cell genomics, epigenetics, and the integration of artificial intelligence in data analysis and treatment decision-making [4,5].

Conclusion

Genomic medicine has transformed oncology by providing a deeper understanding of the genetic drivers of cancer and paving the way for targeted and personalized therapies. By harnessing clinical genomics, healthcare providers can tailor treatment plans to each patient's unique genetic profile, leading to more effective and less toxic therapies. As technology and research continue to advance, the future of cancer treatment lies in precision medicine, where genomic information will play an increasingly crucial role in improving patient outcomes and ultimately moving us closer to conquering cancer. However, addressing challenges related to data interpretation, ethical considerations, and equitable access to genomic testing is vital to ensuring the widespread and responsible implementation of genomic medicine in oncology.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Delaney, Susan K., Michael L. Hultner, Howard J. Jacob and David H. Ledbetter, et al. "Toward clinical genomics in everyday medicine: Perspectives and recommendations." Expert Rev Mol Diagn 16 (2016): 521-532.
  2. Google Scholar, Crossref, Indexed at

  3. Patel, Jai N. "Cancer pharmacogenomics, challenges in implementation and patient-focused perspectives." Pharmgenomics Pers Med (2016): 65-77.
  4. Google Scholar, Crossref,  Indexed at

  5. Jessri, M. and C. S. Farah. "Harnessing massively parallel sequencing in personalized head and neck oncology." J Dent Res 93 (2014): 437-444.
  6. Google Scholar, Crossref, Indexed at

  7. Moore, Donald C. and Andrew S. Guinigundo. "Revolutionizing cancer treatment: Harnessing the power of biomarkers to improve patient outcomes." JAdPrO 14 (2023): 4.
  8. Google Scholar, Crossref,  Indexed at

  9. Tsimberidou, Apostolia M., Ulrik Ringborg and Richard L. Schilsky. "Strategies to overcome clinical, regulatory and financial challenges in the implementation of personalized medicine." ASCO 33 (2013): 118-125.
  10. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 307

Journal of Bioengineering & Biomedical Science received 307 citations as per Google Scholar report

Journal of Bioengineering & Biomedical Science peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward