GET THE APP

Comprehensive Genomic Profiling in Lung Cancer: Enhancing Surgical Pathology Diagnosis and Treatment
..

Journal of Surgical Pathology and Diagnosis

ISSN: 2684-4575

Open Access

Opinion - (2024) Volume 6, Issue 2

Comprehensive Genomic Profiling in Lung Cancer: Enhancing Surgical Pathology Diagnosis and Treatment

Steven Bracey*
*Correspondence: Steven Bracey, Department of Pathology, Princeton University, Princeton, NJ 08544,, USA, Email:
Department of Pathology, Princeton University, Princeton, NJ 08544,, USA

Received: 02-Apr-2024, Manuscript No. jspd-24-144721; Editor assigned: 04-Apr-2024, Pre QC No. P-144721; Reviewed: 16-Apr-2024, QC No. Q-144721; Revised: 22-Apr-2024, Manuscript No. R-144721; Published: 29-Apr-2024 , DOI: 10.37421/2684-4575.2024.6.194
Citation: Bracey, Steven. “Comprehensive Genomic Profiling in Lung Cancer: Enhancing Surgical Pathology Diagnosis and Treatment.” J Surg Path Diag 6 (2024): 194.
Copyright: © 2024 Bracey S. 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

Comprehensive genomic profiling (CGP) has become an integral part of cancer diagnostics, offering detailed insights into the genetic landscape of tumors. In lung cancer, CGP provides critical information that enhances surgical pathology diagnosis and informs treatment strategies. This article reviews the role of CGP in lung cancer, examining its impact on diagnosis, prognosis and personalized treatment. We discuss the methodologies used in CGP, the implications for surgical pathology and future directions in integrating genomic data into clinical practice.

Lung cancer remains one of the leading causes of cancer-related deaths worldwide, with complex molecular underpinnings that challenge traditional diagnostic and therapeutic approaches. Comprehensive genomic profiling (CGP) has emerged as a transformative tool in the management of lung cancer, providing a detailed map of genetic alterations that drive tumorigenesis. By integrating CGP into surgical pathology, clinicians can achieve more accurate diagnoses, predict patient outcomes and tailor treatments to individual genetic profiles. This article explores how CGP enhances lung cancer diagnosis and treatment, highlighting its role in surgical pathology [1].

Description

Methodologies in comprehensive genomic profiling

  1. Next-generation sequencing (NGS)
    • Overview: NGS technologies enable the simultaneous analysis of multiple genes and genomic regions, providing a comprehensive view of genetic alterations. This method is widely used for CGP in lung cancer to identify mutations, copy number variations and other genomic features.
    • Applications: NGS helps in identifying driver mutations such as EGFR, ALK, ROS1 and BRAF, which are critical for selecting targeted therapies. It also detects rare and novel mutations that may influence treatment decisions [2].
  1. Polymerase chain reaction (PCR) and droplet digital PCR (ddPCR)
    • Overview: PCR and ddPCR are techniques used to detect specific genetic mutations and quantify the levels of circulating tumor DNA (ctDNA). These methods complement NGS by providing high sensitivity for known mutations.
    • Applications: PCR and ddPCR are employed to monitor treatment response and detect minimal residual disease (MRD) or recurrence, offering real-time insights into tumor dynamics.
  1. Fluorescence in situ hybridization (FISH)
    • Overview: FISH is used to detect chromosomal abnormalities, such as gene rearrangements and amplifications, by visualizing specific DNA sequences within the tumor cells.
    • Applications: FISH is particularly useful for identifying gene fusions, such as those involving ALK and ROS1, which are relevant for targeted therapy decisions [3].

Implications for surgical pathology

  1. Enhanced diagnostic accuracy
    • Genetic subtyping: CGP allows for precise classification of lung cancer subtypes based on genetic alterations. This refinement in classification can lead to more accurate diagnosis and better prediction of disease behavior.
    • Identification of rare mutations: By uncovering rare or atypical mutations, CGP helps in diagnosing less common forms of lung cancer that may not be detected through standard histopathological methods.
  1. Prognostic insights
    • Risk stratification: Genomic profiling provides information on prognostic markers that can stratify patients based on their risk of disease progression and survival. This allows for more tailored and effective treatment plans.
    • Predictive biomarkers: CGP identifies biomarkers associated with treatment response, such as those predicting sensitivity to specific targeted therapies or resistance mechanisms.
  1. Personalized treatment
    • Targeted therapies: Identification of actionable mutations through CGP enables the use of targeted therapies, such as tyrosine kinase inhibitors (TKIs) for EGFR mutations or ALK inhibitors for ALK rearrangements. This personalized approach improves treatment efficacy and minimizes unnecessary side effects [4].
    • Immunotherapy: Genomic profiling can also inform the use of immune checkpoint inhibitors by identifying tumors with high mutational burdens or specific molecular features associated with response to immunotherapy.
  1. Monitoring and follow-up
    • Minimal residual disease (MRD): CGP aids in monitoring MRD by analyzing ctDNA levels, helping in the early detection of recurrence and guiding subsequent treatment decisions.
    • Dynamic monitoring: Regular genomic profiling allows for tracking changes in the tumor genome over time, providing insights into treatment response and emerging resistance mechanisms.

Challenges and limitations

  1. Technical and interpretative complexity
    • Data interpretation: The vast amount of data generated by CGP requires sophisticated bioinformatics tools and expertise to interpret. Accurate interpretation is crucial for deriving actionable clinical insights.
    • Standardization: Variability in CGP methodologies and reporting standards can affect the consistency and comparability of results across different laboratories and clinical settings.
  1. Cost and accessibility
    • Financial barriers: CGP can be costly, potentially limiting its accessibility in some healthcare settings. Efforts to reduce costs and improve access are essential for broader implementation.
    • Insurance and reimbursement: Coverage for CGP may vary, affecting its availability and integration into routine clinical practice [5].
  1. Ethical and Privacy Concerns
    • Genetic information: The collection and handling of genetic data raise concerns about patient privacy and consent. Ensuring secure data management and addressing ethical considerations are important for maintaining patient trust.

Future directions

  1. Integration of genomic data into clinical practice
    • Guideline development: Establishing clinical guidelines for the use of CGP in lung cancer will help standardize its application and improve patient care.
    • Multidisciplinary approaches: Collaboration between pathologists, oncologists and genetic counselors will enhance the effective use of CGP in personalized treatment planning.
  1. Advancements in technology
    • Improved techniques: Ongoing advancements in sequencing technologies and bioinformatics are expected to enhance the accuracy, speed and affordability of CGP.
    • Liquid biopsies: Combining CGP with liquid biopsy technologies may further improve monitoring and early detection of disease recurrence.
  1. Broader applications
    • Expanding beyond lung cancer: Research into the application of CGP for other cancers and conditions will contribute to the overall advancement of personalized medicine and precision oncology.

Conclusion

Comprehensive genomic profiling represents a significant advancement in the diagnosis and treatment of lung cancer, offering detailed insights into the tumor genome that enhance surgical pathology and inform personalized treatment strategies. While there are challenges to overcome, including technical, financial and ethical considerations, the potential benefits of CGP in improving patient outcomes are substantial. Continued research and technological advancements will further integrate CGP into routine clinical practice, transforming the management of lung cancer and other malignancies.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Ong, Choon Kiat, Chutima Subimerb, Chawalit Pairojkul and Sopit Wongkham, et al. "Exome sequencing of liver fluke–associated cholangiocarcinoma."Nat Genet 44 (2012): 690-693.

    Google Scholar, Crossref, Indexed at

  2. Montal, Robert, Daniela Sia, Carla Montironi and Wei Q. Leow, et al. "Molecular classification and therapeutic targets in extrahepatic cholangiocarcinoma."J Hepatol 73 (2020): 315-327.

    Google Scholar, Crossref, Indexed at

  3. Silver, Robert M. and D. Ware Branch. "Placenta accreta spectrum."N Engl J Med378 (2018): 1529-1536.

    Google Scholar, Crossref, Indexed at

  4. Wu, Serena, Masha Kocherginsky and Judith U. Hibbard. "Abnormal placentation: Twenty-year analysis."Am J Obstet Gynecol 192 (2005): 1458-1461.

    Google Scholar, Crossref, Indexed at

  5. Conturie, Charlotte L. and Deirdre J. Lyell. "Prenatal diagnosis of placenta accreta spectrum."Curr Opin Obstet Gynecol342022): 90-99.

    Google Scholar, Crossref, Indexed at

arrow_upward arrow_upward