Journal of Clinical Pathology and Laboratory Medicine

All submissions of the EM system will be redirected to Online Manuscript Submission System. Authors are requested to submit articles directly to Online Manuscript Submission System of respective journal.
Reach Us +1 (202) 780-3397

Editorial - Journal of Clinical Pathology and Laboratory Medicine (2024) Volume 6, Issue 5

Integrating Molecular Tools in Surgical Pathology for Improved Cancer Diagnosis

Elizabeth Brown *

Department of Anatomical Dissection and Donation, Medical University of Lodz, Poland

*Corresponding Author:
Elizabeth Brown
Department of Anatomical Dissection and Donation, Medical University of Lodz, Poland
E-mail: elizabeth.brown@umed.lodz.pl

Received: 2-Oct-2024, Manuscript No. aacplm-25-157648; Editor assigned: 4-Oct-2024, PreQC No. aacplm-25-157648 (PQ) Reviewed:18-Oct-2024, QC No. aacplm-25-157648 Revised:25-Oct-2024, Manuscript No. aacplm-25-157648; Published:30-Oct-2024, DOI: 10.35841/ aacplm - 6.5.234

Citation: Brown E. Integrating molecular tools in surgical pathology for improved cancer diagnosis. J Clin Path Lab Med. 2024;6(5):234

Introduction

Surgical pathology remains the cornerstone of cancer diagnosis, providing vital information on tumor histology, grading, and staging. However, traditional histopathological techniques often have limitations in distinguishing subtle molecular differences between cancer subtypes. In recent years, the integration of molecular tools into surgical pathology has dramatically enhanced the ability to diagnose, classify, and prognosticate cancer with higher precision. Molecular techniques such as next-generation sequencing (NGS), polymerase chain reaction (PCR), fluorescence in situ hybridization (FISH), and immunohistochemistry (IHC) have become indispensable in providing detailed genetic and molecular profiles of tumors, offering crucial insights into tumor biology. This article explores the integration of molecular tools in surgical pathology and how they are improving cancer diagnosis and patient care [1].

Surgical pathology has long been the gold standard for cancer diagnosis, relying on tissue biopsies obtained during surgery. Histological examination of the tumor tissue allows pathologists to identify cancer type, grade, and stage, providing essential information for treatment planning. However, traditional methods are often limited by their inability to detect molecular alterations, which can play a crucial role in determining prognosis and therapeutic options. While histopathology remains vital, the integration of molecular tools adds a new dimension to cancer diagnosis, improving accuracy and therapeutic decision-making [2].

Molecular diagnostics, which assess genetic mutations, gene expression patterns, and chromosomal abnormalities, are transforming the landscape of cancer diagnosis. Traditional histology classifies tumors based on morphology, but molecular tools allow for a more refined classification by identifying genetic alterations that drive tumorigenesis. For instance, in breast cancer, the identification of HER2 amplification using FISH or PCR helps classify patients as HER2-positive, guiding the use of targeted therapies such as trastuzumab (Herceptin). Similarly, molecular profiling of non-small cell lung cancer (NSCLC) identifies mutations in the EGFR gene, allowing for the use of tyrosine kinase inhibitors (TKIs) for targeted treatment [3].

NGS has revolutionized cancer diagnostics by enabling the simultaneous analysis of multiple genes in a single test. This technique allows pathologists to identify mutations, fusions, and amplifications in tumor DNA, providing a comprehensive genetic profile of the cancer. NGS is particularly useful in cancers with complex molecular landscapes, such as lung cancer, colon cancer, and melanoma. By identifying driver mutations and predicting potential therapeutic targets, NGS provides valuable insights that guide treatment decisions. Furthermore, NGS enables the detection of minimal residual disease (MRD) and the identification of rare mutations that may not be evident using conventional techniques [4].

IHC is a widely used technique in surgical pathology that utilizes antibodies to detect specific antigens or proteins in tissue samples. In cancer diagnostics, IHC plays a crucial role in identifying tumor markers, determining the cellular origin of tumors, and assessing the expression of specific proteins that can inform prognosis and treatment. For example, the detection of PD-L1 expression in tumors through IHC helps identify patients who may benefit from immune checkpoint inhibitors like pembrolizumab or nivolumab. IHC can also be used to assess estrogen receptor (ER), progesterone receptor (PR), and HER2 status in breast cancer, guiding the selection of appropriate therapies [5].

FISH is a powerful technique used to detect specific chromosomal abnormalities, such as gene amplifications, translocations, and deletions. In cancer diagnostics, FISH is particularly useful for detecting genetic rearrangements that may not be apparent using conventional histology. For example, FISH is used to detect HER2 gene amplification in breast cancer, the BCR-ABL fusion gene in chronic myelogenous leukemia (CML), and the EML4-ALK fusion in NSCLC. These genetic alterations are critical for determining the prognosis and selecting targeted therapies, making FISH an essential tool in modern cancer diagnostics [6].

The integration of molecular diagnostics into surgical pathology has paved the way for personalized medicine, where treatments are tailored to the individual genetic profile of the tumor. For instance, the identification of mutations in the EGFR gene in NSCLC patients can guide the use of EGFR inhibitors such as erlotinib or gefitinib. Similarly, mutations in the KRAS gene in colorectal cancer predict resistance to EGFR-targeted therapies. By incorporating molecular testing into the diagnostic workflow, clinicians can identify patients who are most likely to benefit from specific therapies, improving treatment efficacy and minimizing unnecessary side effects [7].

Liquid biopsy, a non-invasive molecular diagnostic technique, is emerging as a complementary tool to traditional tissue biopsy in cancer diagnosis. Liquid biopsy involves the analysis of circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), or exosomes in blood samples. This approach allows for real-time monitoring of tumor dynamics, including the detection of genetic mutations, MRD, and therapy resistance. Liquid biopsy has been particularly useful in cancers like lung cancer, where it can detect EGFR mutations and other genetic alterations without the need for invasive tissue biopsies. As the technology advances, liquid biopsy may become a routine part of cancer diagnostics, offering a less invasive and more accessible alternative to tissue biopsy [8].

The tumor microenvironment (TME) plays a crucial role in cancer progression and response to therapy. Molecular tools are increasingly being used to profile the TME and assess immune cell infiltration, cytokine expression, and immune checkpoint activity. Techniques such as multiplex IHC and RNA sequencing allow for the characterization of immune cell populations within tumors, providing insights into how the immune system interacts with the tumor. Profiling the TME can help identify patients who may benefit from immunotherapy, such as immune checkpoint inhibitors or adoptive T cell therapies, offering a more personalized approach to cancer treatment [9].

Despite the promise of molecular tools in cancer diagnosis, there are challenges associated with their integration into routine surgical pathology practice. These include the high cost of molecular tests, the need for specialized expertise, and the complexities involved in interpreting complex molecular data. Additionally, molecular testing can sometimes yield results that are difficult to integrate with traditional histopathological findings, requiring careful collaboration between pathologists, oncologists, and geneticists. Standardizing molecular testing protocols and ensuring access to these advanced technologies in diverse healthcare settings are essential steps toward broader adoption [10].

Conclusion

The integration of molecular tools into surgical pathology has significantly improved cancer diagnosis, offering more accurate classification, prognostication, and personalized treatment options. Techniques such as NGS, IHC, FISH, and liquid biopsy provide deeper insights into tumor biology, allowing clinicians to tailor therapies to the genetic profile of the cancer. While challenges remain in integrating these technologies into routine practice, the future of cancer diagnostics is poised to be increasingly molecular, driving more effective and personalized patient care.

References

  1. Rieckher M, Garinis GA, Schumacher B. Molecular pathology of rare progeroid diseases. Trends Mol Med. 2021;27 (9):907-22.
  2. Indexed atGoogle ScholarCross Ref

  3. Coppedè F, Migliore L. DNA repair in premature aging disorders and neurodegeneration. Curr Aging Sci. 2010;3 (1):3-19.
  4. Indexed atGoogle ScholarCross Ref

  5. Kyng KJ, Bohr VA. Gene expression and DNA repair in progeroid syndromes and human aging. Ageing Res Rev. 2005;4 (4):579-602.
  6. Indexed atGoogle ScholarCross Ref

  7. Puzianowska-Kuznicka M, Kuznicki J. Genetic alterations in accelerated ageing syndromes: Do they play a role in natural ageing?. Int J Biochem Cell Biol. 2005;37(5):947-60.
  8. Indexed atGoogle ScholarCross Ref

  9. Chen P, ZhiLei G, Zhou B. Insight into the role of adult hippocampal neurogenesis in aging and Alzheimer's disease. Ageing Res Rev. 2022:101828.
  10. Indexed atGoogle ScholarCross Ref

  11. Das Gupta A, Krawczynska N, Nelson ER. Extracellular vesicles—the next frontier in endocrinology. J Endocrinol. 2021;162(9).
  12. Indexed atGoogle ScholarCross Ref

  13. Plotegher N, Duchen MR. Mitochondrial dysfunction and neurodegeneration in lysosomal storage disorders. Trends Mol Med. 2017;23(2):116-34.
  14. Indexed atGoogle ScholarCross Ref

  15. Lv J, Li Y, Shi S, et al. Frontier and hotspot evolution in cardiorenal syndrome: A bibliometric analysis from 2003 to 2022. Curr Probl Cardiol. 2022:101238.
  16. Indexed atGoogle ScholarCross Ref

  17. Barone M, D’Amico F, Rampelli S. Age-related diseases, therapies and gut microbiome: A new frontier for healthy aging. Mech Ageing Dev. 2022;206:111711.
  18. Indexed atGoogle ScholarCross Ref

  19. Dutta D, Khan N, Wu J. Extracellular vesicles as an emerging frontier in spinal cord injury pathobiology and therapy. Trends Neurosci. 2021;44(6):492-506.
  20. Indexed atGoogle ScholarCross Ref

Get the App