GET THE APP

Genetic Screening in the Genomic Era: Opportunities and Challenges
..

Cancer Science & Therapy

ISSN: 1948-5956

Open Access

Mini Review - (2024) Volume 16, Issue 3

Genetic Screening in the Genomic Era: Opportunities and Challenges

Christina Shaheed*
*Correspondence: Christina Shaheed, Department of Public Health, University of Sydney, Sydney, New South Wales, Australia, Email:
Department of Public Health, University of Sydney, Sydney, New South Wales, Australia

Received: 29-Apr-2024, Manuscript No. jcst-24-138790; Editor assigned: 01-May-2024, Pre QC No. P-138790; Reviewed: 15-May-2024, QC No. Q-138790; Revised: 20-May-2024, Manuscript No. R-138790; Published: 27-May-2024 , DOI: 10.37421/1948-5956.2024.16.642
Citation: Shaheed, Christina. “Genetic Screening in the Genomic Era: Opportunities and Challenges.” J Cancer Sci Ther 16 (2024): 642.
Copyright: © 2024 Shaheed C. 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

Genetic screening has undergone a profound transformation in the genomic era, fueled by advancements in sequencing technologies and bioinformatics. This review examines the opportunities and challenges presented by genetic screening in the context of the rapidly evolving landscape of genomics. We discuss the expanding applications of genetic screening, from prenatal testing to population-wide initiatives, and the implications for healthcare, including disease prevention and personalized medicine. Additionally, we explore the ethical, legal, and social considerations surrounding genetic screening, emphasizing the need for informed decision-making and equitable access to genetic services. By analyzing recent research and emerging trends, this review provides insights into the future directions of genetic screening and its role in shaping healthcare policies and practices.

Keywords

Genetic screening • Genomics • Bioinformatics

Introduction

The advent of high-throughput sequencing technologies and the exponential growth of genomic data have revolutionized the field of genetic screening. Genetic screening, once confined to target testing for specific genetic conditions, has now expanded to encompass a wide range of applications, from prenatal diagnosis to population-wide initiatives aimed at disease prevention. The genomic era offers unprecedented opportunities to identify individuals at risk of genetic disorders, tailor interventions to their specific genetic makeup, and ultimately improve health outcomes. However, along with these opportunities come significant challenges, including ethical, legal, and social implications, as well as issues related to data privacy, equity, and access to genetic services. This review examines the evolving landscape of genetic screening in the genomic era, highlighting both the potential benefits and the complex challenges associated with its widespread implementation [1].

Literature Review

Advancements in sequencing technologies have enabled the expansion of genetic screening beyond traditional methods. Whole-genome and whole-exome sequencing now allow for comprehensive analysis of an individual's genetic makeup, facilitating the identification of disease-causing variants and the prediction of disease risk. Genetic screening is increasingly being integrated into clinical practice for various purposes, including prenatal testing, carrier screening, pharmacogenomics, and predisposition testing for common complex diseases. Genetic screening holds immense promise for improving healthcare outcomes through personalized medicine and disease prevention. Early identification of individuals at risk of genetic disorders allows for targeted interventions, such as preventive measures, surveillance, and tailored treatment strategies. Population-wide genetic screening initiatives, coupled with advances in risk prediction algorithms and decision support tools, have the potential to transform public health efforts and reduce the burden of genetic diseases [2].

Despite its potential benefits, genetic screening raises complex ethical, legal, and social issues that must be addressed. These include concerns about privacy and confidentiality, informed consent, discrimination, stigmatization, and the equitable distribution of genetic services. Moreover, the interpretation of genetic data poses challenges due to the complexity of genetic information, the uncertain penetrance of genetic variants, and the potential for unintended consequences. Genetic screening is a powerful tool that allows for the identification of genetic variations or mutations associated with specific diseases or conditions. It serves various purposes, including identifying individuals at risk of inherited disorders, guiding treatment decisions, enabling early detection and intervention, and informing reproductive choices. Genetic screening encompasses a range of techniques, from targeted testing for known genetic mutations to comprehensive analysis of an individual's entire genome [3].

One of the primary applications of genetic screening is in the realm of inherited genetic disorders. By analyzing an individual's genetic makeup, healthcare providers can identify mutations associated with conditions such as cystic fibrosis, sickle cell anemia, Huntington's disease, and hereditary cancers. Screening individuals who are at higher risk due to family history or ethnic background can enable early diagnosis, proactive management, and informed reproductive decisions. In addition to diagnosing genetic disorders, genetic screening plays a crucial role in guiding treatment decisions, particularly in the field of pharmacogenomics. By analysing a patient's genetic profile, healthcare providers can predict how an individual will respond to specific medications, allowing for personalized treatment plans tailored to each patient's genetic makeup. This approach can improve treatment outcomes, minimize adverse drug reactions, and optimize drug efficacy [4].

Genetic screening also has significant implications for disease prevention and public health. Population-wide screening initiatives, such as new-born screening programs, aim to identify individuals with genetic conditions early in life, allowing for timely interventions to prevent or mitigate the effects of these disorders. Similarly, screening for hereditary conditions in adults can enable targeted preventive measures, such as lifestyle modifications, surveillance, or prophylactic surgeries, to reduce the risk of developing certain diseases [5].

Furthermore, advances in genetic screening have expanded its applications beyond diagnosing genetic disorders to predicting an individual's risk of developing common complex diseases, such as cardiovascular disease, diabetes, and certain cancers. By analyzing genetic markers associated with disease risk, healthcare providers can stratify individuals based on their genetic predisposition and tailor preventive interventions accordingly. This approach, known as predictive or predispositional genetic screening, has the potential to empower individuals to take proactive steps to reduce their risk of developing these diseases through lifestyle modifications and regular monitoring [6].

Discussion

Despite its many benefits, genetic screening also raises ethical, legal, and social considerations that must be carefully navigated. These include concerns about privacy and confidentiality, informed consent, the potential for discrimination or stigmatization based on genetic information, and the equitable access to genetic services. Additionally, the interpretation of genetic data can be complex, requiring specialized expertise and resources to ensure accurate analysis and appropriate counseling of individuals undergoing genetic screening. The widespread implementation of genetic screening in the genomic era offers immense opportunities to improve healthcare outcomes and advance our understanding of human genetics. However, realizing the full potential of genetic screening requires addressing a myriad of challenges, including ethical, legal, and social considerations, as well as technical and practical issues related to data interpretation, counseling, and resource allocation. Collaborative efforts among stakeholders, including healthcare providers, policymakers, researchers, and patient advocacy groups, are essential to navigate these challenges and ensure the responsible and equitable integration of genetic screening into clinical practice.

Conclusion

Genetic screening in the genomic era holds great promise for advancing precision medicine, disease prevention, and public health. By leveraging the power of genomics to identify individuals at risk of genetic disorders, genetic screening has the potential to revolutionize healthcare delivery and improve patient outcomes. However, the realization of this potential requires careful consideration of the ethical, legal, and social implications of genetic screening, as well as the development of robust infrastructure, policies, and guidelines to support its responsible implementation. With continued research, innovation, and collaboration, genetic screening will play an increasingly important role in shaping the future of healthcare.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Libby, Peter, Paul M. Ridker and Attilio Maseri. "Inflammation and atherosclerosis." Circulation 105 (2002): 1135-1143.
  2. Google Scholar, Crossref, Indexed at

  3. Clark, B., J. Sitzia and W. Harlow. "Incidence and risk of arm oedema following treatment for breast cancer: A three-year follow-up study." Qjm 98 (2005): 343-348.
  4. Google Scholar, Crossref, Indexed at

  5. Olsson Möller, Ulrika, Ingela Beck, L. Rydén and M. Malmström. "A comprehensive approach to rehabilitation interventions following breast cancer treatment-A systematic review of systematic reviews." BMC Cancer 19 (2019): 1-20.
  6. Google Scholar, Crossref, Indexed at

  7. Armer, Jane M., M. Elise Radina, Davina Porock and Scott D. Culbertson. "Predicting breast cancer-related lymphedema using self-reported symptoms." Nurs Res 52 (2003): 370-379.
  8. Google Scholar, Crossref, Indexed at

  9. Sage, Andrew P. and Ziad Mallat. "Multiple potential roles for B cells in atherosclerosis." Ann Med 46 (2014): 297-303.
  10. Google Scholar, Crossref,  Indexed at

  11. Luke, Brian, Andrea Panza, Sophie Redon and Nahid Iglesias, et al. "The Rat1p 5′ to 3′ exonuclease degrades telomeric repeat-containing RNA and promotes telomere elongation in Saccharomyces cerevisiae." Mol cell 32 (2008): 465-477.
  12. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 3968

Cancer Science & Therapy received 3968 citations as per Google Scholar report

Cancer Science & Therapy peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward