CGE_2024v12n2

Cancer Genetics and Epigenetics 2024, Vol.12, No.2, 97-105 http://medscipublisher.com/index.php/cge 102 genetic landscape. Additionally, the genetic diversity in prostate cancer underscores the need for personalized approaches to screening and treatment, as a one-size-fits-all strategy may not be effective (Cózar et al., 2018). 6.2 Ethical considerations The use of genetic markers in early screening for prostate cancer raises several ethical concerns. One major issue is the potential for genetic discrimination, where individuals may face discrimination based on their genetic predisposition to prostate cancer. This could affect their employment, insurance, and social standing. Furthermore, the psychological impact of genetic testing cannot be overlooked. The knowledge of carrying a genetic risk for prostate cancer can cause significant anxiety and stress for patients and their families. There is also the ethical dilemma of informed consent, where patients must fully understand the implications of genetic testing, including the potential for false positives or negatives, and the limitations of current genetic markers in providing definitive prognostic information (Aly et al., 2011). Ensuring that patients are adequately informed and supported throughout the genetic testing process is crucial to addressing these ethical challenges. 6.3 Accessibility and cost-effectiveness The accessibility and cost-effectiveness of genetic testing for prostate cancer screening are significant barriers to its widespread adoption. Genetic tests can be expensive, and their costs may not be covered by insurance, making them inaccessible to many patients, particularly those from lower socioeconomic backgrounds (Boström et al., 2015). Additionally, the implementation of genetic testing in clinical practice requires substantial investment in infrastructure, training, and resources, which may not be feasible for all healthcare settings (Cucchiara et al., 2018). The cost utility of these tests is still under evaluation, and large-scale, multi-institutional studies are needed to validate their efficacy and cost-effectiveness. Moreover, the availability of genetic testing may be limited in certain regions, further exacerbating disparities in access to early screening and personalized treatment options (Nevo et al., 2020). Addressing these issues is essential to ensure that the benefits of genetic markers in prostate cancer screening are equitably distributed across all patient populations. 7 Future Directions 7.1 Emerging genetic markers The landscape of genetic markers for prostate cancer (PCa) is rapidly evolving, with several promising candidates on the horizon. Recent studies have identified numerous single nucleotide polymorphisms (SNPs) associated with PCa risk, with 35 SNPs already validated and an additional 50~75 expected to be identified soon (Aly et al., 2011). These markers, although modest in their individual effect, collectively explain a significant portion of familial risk. Moreover, emerging markers such as the rs6983561 genotype have shown potential in improving predictive accuracy for early-onset PCa, particularly among high-risk groups like African American men (Hughes et al., 2012). The integration of these new genetic markers into clinical practice could significantly enhance early detection and risk stratification. 7.2 Integration with other diagnostic modalities The future of PCa diagnosis lies in the integration of genetic markers with other diagnostic modalities. Combining genetic information with traditional biomarkers such as prostate-specific antigen (PSA) and advanced imaging techniques could provide a more comprehensive assessment of PCa risk. For instance, the combination of serum PSA, urinary PCA3, and TMPRSS2-ERG fusion has demonstrated superior sensitivity and specificity compared to traditional diagnostic approaches (McGrath et al., 2016). Additionally, urine-based assays like SelectMDx and Mi-Prostate Score are emerging as valuable tools for identifying patients who may benefit from prostate biopsy (Cucchiara et al., 2018). This multimodal approach could reduce unnecessary biopsies and improve the accuracy of PCa diagnosis. 7.3 Personalized medicine approaches Personalized medicine is poised to revolutionize the management of PCa by tailoring treatment strategies based on individual genetic profiles. Genomic biomarkers such as AR-V7 expression and mutations in DNA mismatch repair genes are already being used to guide treatment decisions in castration-resistant PCa 3. Furthermore, commercially available gene panels like Prolaris, Oncotype DX, and Decipher are being utilized to estimate

RkJQdWJsaXNoZXIy MjQ4ODYzNQ==