GET THE APP

Genomic Profiling and Personalized Medicine in Pulmonary Cancer Treatment
..

Pulmonary & Respiratory Medicine

ISSN: 2161-105X

Open Access

Mini Review - (2023) Volume 13, Issue 4

Genomic Profiling and Personalized Medicine in Pulmonary Cancer Treatment

Terry Hardy*
*Correspondence: Terry Hardy, Department of Clinical Studies, University of Guelph, Guelph, Canada, Email:
Department of Clinical Studies, University of Guelph, Guelph, Canada

Received: 01-Aug-2023, Manuscript No. jprm-23-111767; Editor assigned: 03-Aug-2023, Pre QC No. P-111767; Reviewed: 15-Aug-2023, QC No. Q-111767; Revised: 21-Aug-2023, Manuscript No. R-111767; Published: 28-Aug-2023 , DOI: 10.37421/2161-105X.2023.13.646
Citation: Hardy, Terry. “Genomic Profiling and Personalized Medicine in Pulmonary Cancer Treatment.” J Pulm Respir Med 13 (2023): 646.
Copyright: © 2023 Hardy T. 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

Lung cancer remains one of the most prevalent and deadliest malignancies worldwide, contributing significantly to cancer-related morbidity and mortality. Traditional treatment approaches have primarily focused on generalized therapies that may not be optimally effective for all patients due to the inherent heterogeneity of lung cancer. However, advancements in genomic profiling and personalized medicine have opened new avenues for tailoring treatment strategies to individual patients, enhancing treatment outcomes and potentially revolutionizing the landscape of pulmonary cancer treatment.

Keywords

Genetic • Public health • Lung cancer

Introduction

Genomic profiling involves the comprehensive analysis of a patient's genetic material to identify alterations, mutations, and variations in their DNA. In the context of lung cancer, genomic profiling primarily targets somatic mutations that are unique to the tumor cells. These genetic alterations can provide crucial insights into the underlying molecular mechanisms driving tumor growth, progression, and resistance to treatment. One of the ground breaking discoveries that emerged from genomic profiling is the identification of oncogenic driver mutations. These mutations play a pivotal role in initiating and sustaining tumor growth. For instance, mutations in genes such as Epidermal Growth Factor Receptor (EGFR), Anaplastic Lymphoma Kinase (ALK), ROS1, and BRAF have been identified as key drivers in certain subsets of lung cancer patients. Identification of these mutations through genomic profiling allows clinicians to select targeted therapies that specifically inhibit the activity of these mutant proteins, leading to improved treatment responses and reduced adverse effects compared to traditional chemotherapy.

Literature Review

The advent of personalized medicine has transformed the way lung cancer is managed. Instead of employing a one-size-fits-all approach, personalized medicine considers the unique genetic makeup of each patient's tumor and tailors treatment strategies accordingly. This approach has the potential to maximize therapeutic efficacy while minimizing unnecessary treatments and associated toxicities. Targeted therapies constitute a cornerstone of personalized medicine in lung cancer treatment. These therapies exploit the specific vulnerabilities created by oncogenic driver mutations. For instance, Tyrosine Kinase Inhibitors (TKIs) have revolutionized the treatment of EGFRmutant lung cancers. Drugs like erlotinib, Gefitinib, and Osimertinib selectively block the aberrant signaling pathways activated by mutant EGFR, resulting in remarkable responses in patients with these mutations [1].

Similarly, ALK and ROS1 rearrangements have paved the way for ALK and ROS1 inhibitors, such as crizotinib, alectinib, and entrectinib, which have demonstrated impressive efficacy in patients harboring these genetic alterations. The ability to match the right patients with the right targeted therapies showcases the potential of personalized medicine to significantly extend survival and improve the quality of life for lung cancer patients [2].

Discussion

While genomic profiling and personalized medicine offer promising prospects, several challenges and limitations need to be addressed. The most significant challenge is the identification of actionable mutations in the context of tumor heterogeneity. Tumors can harbor multiple sub clones with distinct genetic alterations, making it essential to capture the entire genomic landscape to make informed treatment decisions. Additionally, the accessibility and affordability of comprehensive genomic profiling remain barriers, hindering the widespread integration of these approaches into routine clinical practice. Another limitation is the emergence of acquired resistance to targeted therapies. Although initial responses to targeted agents can be remarkable, tumors often develop resistance mechanisms over time, leading to disease progression. Research efforts are underway to decipher the molecular mechanisms underlying resistance and to develop strategies to overcome or delay its onset [3].

Immunotherapy has emerged as a revolutionary approach in cancer treatment, including lung cancer. Genomic profiling has contributed to our understanding of the complex interactions between tumors and the immune system. Certain genetic alterations, such as high tumor mutational burden and microsatellite instability, have been associated with increased responsiveness to Immune Checkpoint Inhibitors (ICIs), a class of immunotherapeutic agents.

Programmed Death-Ligand 1 (PD-L1) expression, determined through genomic profiling, serves as a predictive biomarker for patient responses to ICIs. Tumors with higher PD-L1 expression tend to exhibit greater susceptibility to these agents. However, the relationship between PD-L1 expression and treatment response is not absolute, highlighting the need for a multifaceted approach that considers both genomic and immune factors. Genomic profiling and personalized medicine have revolutionized the landscape of lung cancer treatment, offering unprecedented opportunities to tailor interventions to the genetic makeup of individual tumors. As technology continues to advance, comprehensive genomic profiling will likely become more accessible, enabling clinicians to make informed treatment decisions based on a broader genetic landscape.

Combination therapies that target multiple vulnerabilities simultaneously, including both oncogenic drivers and immune-related factors, represent an exciting frontier. Clinical trials are investigating the synergistic effects of combining targeted therapies with immunotherapies to enhance treatment responses and extend patient survival. genomic profiling and personalized medicine have ushered in a new era of hope for pulmonary cancer treatment. By deciphering the intricate genetic alterations that drive tumor growth, clinicians can make informed decisions about the most appropriate targeted therapies or immunotherapies for each patient. As research advances and technology becomes more refined, the integration of these approaches into routine clinical practice has the potential to transform lung cancer from a deadly disease to a manageable chronic condition, improving patient outcomes and quality of life [4-6].

Conclusion

Ongoing advancements in our understanding of the molecular mechanisms underlying lung cancer are likely to uncover new and previously unforeseen therapeutic targets. As we delve deeper into the genomics of lung cancer, more precise treatment options will emerge, allowing for even greater personalization of therapies. it is important to acknowledge that the successful integration of genomic profiling and personalized medicine into routine clinical practice requires a multidisciplinary approach. Collaboration between oncologists, pathologists, geneticists, and researchers is essential to ensure accurate diagnosis, effective treatment selection, and continuous monitoring of treatment responses. Ethical considerations surrounding genomic profiling and personalized medicine should not be overlooked. Patient privacy, data security, and the potential for unintended consequences resulting from genetic testing must be carefully addressed as these approaches become more widespread. The synergy between genomic profiling and personalized medicine has revolutionized the treatment landscape for pulmonary cancer. The ability to unravel the genetic intricacies of tumors and tailor treatment strategies accordingly has redefined the possibilities for patient care. While challenges and limitations exist, the potential to provide more effective, targeted, and less toxic treatments makes this approach a beacon of hope for patients and healthcare providers alike. As research and technology continue to progress, the vision of truly individualized lung cancer treatment draws closer, offering the promise of prolonged survival, improved quality of life, and a brighter future for those affected by this devastating disease.

Acknowledgement

None.

Conflict of Interest

The authors declare that there is no conflict of interest associated with this manuscript.

References

  1. Schachter, Jacob, Antoni Ribas, Georgina V. Long and Ana Arance, et al. "Pembrolizumab vs. ipilimumab for advanced melanoma: Final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006)." The Lancet 390 (2017): 1853-1862.
  2. Google Scholar, Crossref, Indexed at

  3. Napolitano, S., G. Brancaccio, G. Argenziano and E. Martinelli, et al. "It is finally time for adjuvant therapy in melanoma." Cancer Treat Rev 69 (2018): 101-111.
  4. Google Scholar, Crossref, Indexed at

  5. Wang, Daniel Y., Joe-Elie Salem, Justine V. Cohen and Sunandana Chandra, et al. "Fatal toxic effects associated with immune checkpoint inhibitors: A systematic review and meta-analysis." JAMA Oncol 4 (2018): 1721-1728.
  6. Google Scholar, Crossref, Indexed at

  7. Kumar, Vivek, Neha Chaudhary, Mohit Garg and Charalampos S. Floudas, et al. "Current diagnosis and management of immune related Adverse Events (irAEs) induced by immune checkpoint inhibitor therapy." Front Pharmacol 8 (2017): 49.
  8. Google Scholar, Crossref, Indexed at

  9. Sosa, Aaron, Esther Lopez Cadena, Cristina Simon Olive and Niki Karachaliou, et al. "Clinical assessment of immune-related adverse events." Ther Adv Med Oncol 10 (2018): 1758835918764628.
  10. Google Scholar, Crossref, Indexed at

  11. Howell, Matthew, Rebecca Lee, Samantha Bowyer and Alberto Fusi, et al. "Optimal management of immune-related toxicities associated with checkpoint inhibitors in lung cancer." Lung Cancer 88 (2015): 117-123.
  12. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 1690

Pulmonary & Respiratory Medicine received 1690 citations as per Google Scholar report

Pulmonary & Respiratory Medicine peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward