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

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

Targeted Therapies and Molecular Diagnostics: The Evolving Role of Molecular Pathology

Raj Ali *

Department of Psychiatry, McGill University, Canada

*Corresponding Author:
Raj Ali
Department of Psychiatry, McGill University, Canada
E-mail: raj.ali@gmail.com

Received: 2-Oct-2024, Manuscript No. aacplm-25-157651; Editor assigned: 4-Oct-2024, PreQC No. aacplm-25-157651 (PQ) Reviewed:18-Oct-2024, QC No. aacplm-25-157651 Revised:25-Oct-2024, Manuscript No. aacplm-25-157651; Published:30-Oct-2024, DOI: 10.35841/ aacplm - 6.5.235

Citation: Ali R. Targeted therapies and molecular diagnostics: The evolving role of molecular pathology. J Clin Path Lab Med. 2024;6(5):235

Introduction

The landscape of cancer treatment has undergone a paradigm shift with the advent of targeted therapies, driven by a deeper understanding of the molecular mechanisms underlying tumorigenesis. Molecular pathology, which integrates genetic and molecular diagnostic tools, plays a pivotal role in the identification of specific tumor alterations that guide the use of these therapies. By focusing on the molecular drivers of cancer, targeted therapies offer a more personalized and precise approach to treatment, often with fewer side effects than traditional chemotherapy. This article explores the evolving role of molecular pathology in the diagnosis and treatment of cancer, with a focus on how molecular diagnostics are reshaping targeted therapies [1].

Traditional cancer treatments, such as surgery, chemotherapy, and radiation therapy, target rapidly dividing cells, often affecting both cancerous and healthy tissues. Targeted therapies, on the other hand, focus on specific molecules involved in cancer cell growth, survival, and spread. These therapies aim to disrupt the molecular pathways that drive tumorigenesis, providing a more effective and less toxic treatment option for patients. The success of targeted therapies, such as tyrosine kinase inhibitors (TKIs) for lung cancer and trastuzumab (Herceptin) for HER2-positive breast cancer, has revolutionized cancer care, offering hope for better outcomes and improved quality of life [2].

Molecular pathology involves the analysis of genetic mutations, protein expression, and other molecular features within tumor tissues. By utilizing techniques such as next-generation sequencing (NGS), polymerase chain reaction (PCR), and fluorescence in situ hybridization (FISH), molecular pathologists can identify specific alterations in tumor DNA and RNA. These molecular changes, such as mutations, amplifications, and translocations, can provide valuable information about the underlying causes of cancer and the most appropriate therapeutic strategies. The role of molecular pathology is essential in determining which patients are most likely to benefit from targeted therapies based on their tumor's molecular profile [3].

NGS has emerged as a revolutionary tool in molecular pathology, enabling the simultaneous sequencing of multiple genes and providing comprehensive genetic information about a tumor. Unlike traditional methods, which focus on individual gene mutations, NGS can identify a wide range of genetic alterations, including point mutations, insertions, deletions, and gene fusions. In cancers such as non-small cell lung cancer (NSCLC) and melanoma, NGS has been instrumental in identifying actionable mutations, such as EGFR, ALK, and BRAF, which can be targeted by specific therapies. NGS allows for a more detailed and personalized approach to cancer treatment, improving patient outcomes [4].

Biomarker testing plays a central role in determining whether a patient is a suitable candidate for targeted therapy. Various molecular markers, including mutations, amplifications, and gene fusions, are assessed through diagnostic tests to predict the likelihood of response to specific treatments. For example, testing for mutations in the EGFR gene in NSCLC patients helps identify individuals who are likely to respond to EGFR inhibitors like erlotinib and gefitinib. Similarly, HER2 amplification testing in breast cancer helps guide the use of HER2-targeted therapies such as trastuzumab. These biomarker-driven approaches ensure that patients receive the most effective therapy based on their tumor’s molecular characteristics [5].

Targeted therapies have shown significant promise in the treatment of solid tumors, offering better precision and fewer side effects compared to traditional treatments. In addition to EGFR inhibitors in NSCLC and HER2-targeted therapies in breast cancer, other targeted therapies are emerging for a variety of solid tumors. For example, the use of BRAF inhibitors in melanoma has led to improved survival outcomes for patients with BRAF V600E mutations. Similarly, inhibitors targeting the PI3K/AKT/mTOR pathway are being explored for cancers such as breast cancer, endometrial cancer, and glioblastoma. The ability to match specific molecular alterations with targeted therapies has revolutionized the treatment landscape for many solid tumors [6].

Molecular diagnostics have also transformed the treatment of hematologic malignancies, such as leukemia, lymphoma, and multiple myeloma. In chronic myelogenous leukemia (CML), the discovery of the BCR-ABL fusion gene has led to the development of tyrosine kinase inhibitors like imatinib, which specifically target the abnormal fusion protein. Similarly, the identification of mutations in the JAK2 gene in myeloproliferative disorders has led to the use of JAK2 inhibitors such as ruxolitinib. Molecular profiling of hematologic cancers has enabled the development of targeted therapies that significantly improve patient survival and quality of life, while minimizing toxicity [7].

One of the challenges in targeted therapy is the development of resistance, which can occur through secondary mutations, bypass pathways, or altered drug metabolism. For instance, resistance to EGFR inhibitors in NSCLC can develop through secondary mutations in the EGFR gene or activation of alternate signaling pathways. To overcome this, molecular pathologists use ongoing genomic testing to monitor changes in the tumor’s molecular profile and identify new mutations associated with resistance. Combination therapies, which target multiple molecular pathways simultaneously, are also being explored to prevent or overcome resistance and improve long-term outcomes [8].

Liquid biopsy is an emerging non-invasive technique that allows for the analysis of tumor-derived genetic material, such as ctDNA, CTCs, and exosomes, in blood or other bodily fluids. Liquid biopsy can be used to monitor the genetic alterations present in tumors, detect minimal residual disease (MRD), and assess resistance mechanisms during treatment. This technique offers a less invasive alternative to traditional tissue biopsies, enabling real-time monitoring of tumor dynamics and providing insights into treatment response and disease progression. Liquid biopsy is particularly useful for tracking mutations in cancers such as lung, colorectal, and breast cancer, where tissue biopsy may be challenging or unavailable [9].

Companion diagnostics are tests developed alongside targeted therapies to identify patients who are most likely to benefit from the treatment. These tests are essential in ensuring that targeted therapies are used appropriately and effectively. For example, the FDA-approved companion diagnostic for the use of pembrolizumab (Keytruda) in cancer immunotherapy tests for PD-L1 expression, identifying patients who are more likely to respond to immune checkpoint inhibitors. Companion diagnostics also play a crucial role in the development of new targeted therapies, as they help define patient populations that are likely to benefit from novel treatments in clinical trials [10].

Conclusion

Molecular pathology has become an indispensable part of modern cancer diagnosis and treatment, driving the success of targeted therapies. By identifying specific molecular alterations in tumors, molecular diagnostics enable clinicians to match patients with the most appropriate and effective therapies, leading to improved outcomes and reduced side effects. As technologies continue to evolve, the role of molecular pathology in personalized cancer care will only grow, offering new opportunities to treat cancer more effectively and with greater precision. Targeted therapies, guided by molecular diagnostics, represent the future of cancer treatment, transforming the way we approach and manage this complex disease.

References

  1. Reifenberger G, Wirsching HG, Knobbe-Thomsen CB. Advances in the molecular genetics of gliomas—implications for classification and therapy. Nat Rev Clin. Oncol. 2017;14(7):434-52.
  2. Indexed atGoogle ScholarCross Ref

  3. Ahn JC, Teng PC, Chen PJ, et al. Detection of circulating tumor cells and their implications as a biomarker for diagnosis, prognostication, and therapeutic monitoring in hepatocellular carcinoma. J Hepatol. 2021;73(1):422-36.
  4. Indexed atGoogle ScholarCross Ref

  5. Weber T, Schmitz R. Molecular subgroups of diffuse large B cell lymphoma: Biology and implications for clinical practice. Curr Oncol Rep. 2022;24(1):13-21.
  6. Indexed atGoogle ScholarCross Ref

  7. Wadmann S. Disease classification: A framework for analysis of contemporary developments in precision medicine. SSM Qual Res Health. 2023:100217.
  8. Indexed atGoogle ScholarCross Ref

  9. Gomes P, Tzouanou F, Skolariki K, et al. Extracellular vesicles and Alzheimer’s disease in the novel era of precision medicine: Implications for disease progression, diagnosis and treatment. Exp Neurol 2022;358:114183.
  10. Indexed atGoogle ScholarCross Re

  11. Li F, Lu ZY, Xue YT,et al. Molecular basis of JAK2 H608Y and H608N mutations in the pathology of acute myeloid leukemia. Int J Biol Macromol. 2023;229:247-59.
  12. Indexed atGoogle ScholarCross Ref

  13. Thiagalingam S. Epigenetic memory in development and disease: Unraveling the mechanism. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2020;1873(2):188349.
  14. Indexed atGoogle Scholar

  15. Roullet M, Bagg A. The basis and rational use of molecular genetic testing in mature B-cell lymphomas. Adv Anat Pathol. 2010;17(5):333-58.
  16. Indexed atGoogle ScholarCross Ref

  17. Luciano M, Krenn PW, Horejs-Hoeck J. The cytokine network in acute myeloid leukemia. Front Immunol. 2022;13.
  18. Indexed atGoogle ScholarCross Ref

  19. Kim Y, Greenleaf WJ, Bendall SC. Systems biology approaches to unravel lymphocyte subsets and function. Curr Opin Immunol. 2023;82:102323.
  20. Indexed atGoogle Scholar

Get the App