Journal of Cancer Immunology & Therapy

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

Rapid Communication - Journal of Cancer Immunology & Therapy (2024) Volume 7, Issue 2

Advancements in personalized cancer treatment: A new era of precision oncology.

p> Youra Gyga *

 

Department of Pathology, Dalhousie University, Canada

*Corresponding Author:
Youra Gyga
Department of Pathology, Dalhousie University, Canada
E-mail: gygi.y@dal.ca

Received: 02-Apr -2024, Manuscript No. AAJCIT-24-132921; Editor assigned: 03-Apr-2024, PreQC No. AAJCIT-24-132921 (PQ); Reviewed:17-Apr-2024, QC No. AAJCIT-24-132921; Revised:22-Apr-2024, Manuscript No. AAJCIT-24-132921(R); Published:29-Apr-2024, DOI:10.35841/aara-7.2.201

Citation: Gyga Y., Advancements in personalized cancer treatment: A new era of precision oncology. J Cancer Immunol Ther. 2024;7(2):201

Visit for more related articles at Journal of Cancer Immunology & Therapy

Introduction

Cancer, once considered a monolithic disease, is now recognized as a complex constellation of disorders characterized by diverse molecular and genetic alterations. This heterogeneity poses significant challenges to traditional one-size-fits-all treatment approaches. However, the advent of personalized cancer treatment, also known as precision oncology, has ushered in a new era of tailored therapies that aim to match treatments to the specific genetic and molecular profiles of individual patients' tumors. In this article, we explore the advancements in personalized cancer treatment and their profound impact on improving outcomes for cancer patients [1].

Precision oncology encompasses a multifaceted approach to cancer treatment that integrates advanced genomic sequencing, molecular profiling, and computational analysis to identify targetable alterations driving tumor growth [2].

By elucidating the unique genetic mutations, gene expressions, and signaling pathways driving cancer progression, precision oncology enables clinicians to select therapies with greater likelihood of efficacy and minimal toxicity for each patient [3].

Central to personalized cancer treatment is the comprehensive genomic profiling of tumors to identify actionable alterations and potential therapeutic targets. High-throughput sequencing technologies, such as next-generation sequencing (NGS), facilitate the rapid and cost-effective analysis of tumor genomes, enabling the identification of driver mutations, oncogenic fusions, and other genomic aberrations. Concurrently, biomarker discovery efforts aim to identify predictive biomarkers, such as mutations in genes like EGFR, ALK, and BRAF in lung cancer, to guide treatment selection and optimize patient outcomes [4].

Targeted therapies constitute a cornerstone of personalized cancer treatment, designed to specifically inhibit molecular targets implicated in cancer pathogenesis. These therapies include small molecule inhibitors and monoclonal antibodies that selectively target oncogenic proteins or signaling pathways essential for tumor survival and growth. Examples of targeted therapies include EGFR inhibitors in lung cancer, BRAF inhibitors in melanoma, and HER2-targeted therapies in breast cancer [5].

By precisely targeting the vulnerabilities of cancer cells, targeted therapies offer the potential for improved efficacy and reduced off-target toxicity compared to conventional chemotherapy. Another groundbreaking advancement in personalized cancer treatment is the advent of immunotherapy, which harnesses the body's immune system to recognize and eliminate cancer cells [6].

Immune checkpoint inhibitors, such as anti-PD-1 and anti-CTLA-4 antibodies, have demonstrated remarkable efficacy across various cancer types, including melanoma, lung cancer, and bladder cancer. Additionally, adoptive cell therapies, such as chimeric antigen receptor (CAR) T-cell therapy, are being explored as personalized treatments for hematologic malignancies by genetically engineering patients' T cells to target specific tumor antigens [7].

Liquid biopsies, which analyze circulating tumor DNA (ctDNA) or other biomarkers in blood samples, have emerged as valuable tools for non-invasive monitoring of disease progression and treatment response in cancer patients [8].

By detecting genetic mutations, copy number alterations, and other molecular changes in real-time, liquid biopsies enable clinicians to track tumor dynamics, detect treatment resistance, and adjust therapeutic strategies accordingly. Moreover, liquid biopsies hold promise for early detection of cancer recurrence and monitoring minimal residual disease following surgery or other treatments [9].

Despite the remarkable progress in personalized cancer treatment, several challenges remain to be addressed. These include the need for improved biomarker discovery, validation of predictive biomarkers, overcoming tumor heterogeneity and evolution, and expanding access to genomic profiling and targeted therapies for all patients. Additionally, the integration of multi-omics data, including genomics, transcriptomics, proteomics, and metabolomics, holds the potential to further refine treatment algorithms and identify novel therapeutic targets [10].

conclusion

Personalized cancer treatment represents a paradigm shift in oncology, moving towards tailored therapies that capitalize on the unique molecular characteristics of individual tumors. By leveraging genomic insights and targeted interventions, precision oncology offers the promise of improved outcomes, enhanced quality of life, and ultimately, personalized approaches to cancer care. As technology continues to advance and our understanding of cancer biology deepens, the future of personalized cancer treatment holds tremendous potential for transforming the landscape of cancer care and ushering in an era of precision medicine.

References

  1. A Baudino T. Targeted cancer therapy: the next generation of cancer treatment. Curr Drug Discov Technol. 2015;12(1):3-20.
  2. Indexed at, Goggle Scholar, Cross ref

  3. Efferth T, Li PC, Konkimalla VS, et al., From traditional Chinese medicine to rational cancer therapy. Trends Mol Med. 2007;13(8):353-61.
  4. Indexed at, Goggle Scholar

  5. Moslehi JJ. Cardiovascular toxic effects of targeted cancer therapies. New England Journal of Medicine. 2016;375(15):1457-67.
  6. Goggle Scholar

  7. Sullivan R, Smith JE, Rowan NJ. Medicinal mushrooms and cancer therapy: translating a traditional practice into Western medicine. Perspect Biol Med. 2006;49(2):159-70.
  8. Indexed at, Goggle Scholar

  9. Zhang H, Berezov A, Wang Q, et al., ErbB receptors: from oncogenes to targeted cancer therapies. J Clin Investig. 2007;117(8):2051-8.
  10. Indexed at, Goggle Scholar

  11. Perera PK. Traditional medicine-based therapies for cancer management. Sys Rev Pharm. 2019;10(1):90-2.
  12. Indexed at, Goggle Scholar, Cross ref

  13. Ouyang L, Luo Y, Tian M, et al., Plant natural products: from traditional compounds to new emerging drugs in cancer therapy. Cell Prolif. 2014;47(6):506-15.
  14. Indexed at, Goggle Scholar, Cross ref

  15. Ky B, Vejpongsa P, Yeh ET, et al., Emerging paradigms in cardiomyopathies associated with cancer therapies. Circ Res.2013;113(6):754-64.
  16. Indexed at, Goggle Scholar, Cross ref

  17. Herrmann J. Adverse cardiac effects of cancer therapies: cardiotoxicity and arrhythmia. Nat Rev Cardiol. 2020;17(8):474-502.
  18. Indexed at, Goggle Scholar, Cross ref

  19. Chen H, Zhou X, Gao Y, et al., Recent progress in development of new sonosensitizers for sonodynamic cancer therapy. Drug Discov Today. 2014;19(4):502-9.
  20. Indexed at, Goggle Scholar, Cross ref

Get the App