ISSN: 2572-4118

Breast Cancer: Current Research
Open Access

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Editorial   
  • Breast Can Curr Res 2024, Vol 9(4): 266

Tumor Mutational Burden as a Biomarker: Predicting Response to Immune Checkpoint Inhibitors

Felipe Churni*
Department of Radiology and Pathology, Cambridge University, United Kingdom
*Corresponding Author: Felipe Churni, Department of Radiology and Pathology, Cambridge University, United Kingdom, Email: felipe253@yahoo.com

Received: 02-Aug-2024 / Manuscript No. bccr-24-151318 / Editor assigned: 05-Aug-2024 / PreQC No. bccr-24-151318 / Reviewed: 19-Aug-2024 / QC No. bccr-24-151318 / Revised: 23-Aug-2024 / Manuscript No. bccr-24-151318 / Published Date: 29-Aug-2024

Abstract

Tumor Mutational Burden (TMB) has emerged as a crucial biomarker in the field of oncology, particularly for predicting patient responses to immune checkpoint inhibitors (ICIs). TMB quantifies the number of mutations within a tumor’s DNA, reflecting its potential to generate neoantigens that can enhance immune recognition. This article reviews the significance of TMB in guiding immunotherapy decisions, exploring its measurement techniques, clinical implications, and associations with treatment outcomes across various malignancies. While high TMB is generally linked to improved responses to ICIs, challenges such as variability in measurement standards and the influence of additional factors complicate its clinical application. This review underscores the importance of TMB in personalized medicine and advocates for further research to refine its use as a predictive biomarker, ultimately aiming to optimize therapeutic strategies for cancer patients.

Keywords

Tumor mutational burden; Immune checkpoint inhibitors; Biomarkers; Cancer immunotherapy; Personalized medicine; Predictive value; Solid tumors; Genomic profiling

Introduction

The landscape of cancer treatment has dramatically evolved with the introduction of immune checkpoint inhibitors (ICIs), which harness the body’s immune system to target and eliminate cancer cells. These therapies have demonstrated significant efficacy across various malignancies, including melanoma, non-small cell lung cancer (NSCLC), and bladder cancer. Despite their success, not all patients experience favorable outcomes, leading to a pressing need for reliable biomarkers that can predict therapeutic responses [1].

One such biomarker, Tumor Mutational Burden (TMB), has gained attention in recent years. TMB measures the total number of mutations per mega base of DNA within a tumor, serving as an indicator of the tumor's genetic complexity and its potential to produce neoantigen—novel proteins that can elicit immune responses. The hypothesis underlying TMB’s predictive value is that a higher mutational burden may enhance the likelihood of generating recognizable neoantigen, thus improving the efficacy of ICIs [2].

Research has shown that patients with high TMB tend to have better response rates to ICIs, suggesting that TMB could play a pivotal role in selecting patients who are most likely to benefit from these therapies. As a result, TMB is increasingly being incorporated into clinical practice, influencing treatment decisions and patient management strategies. However, challenges remain in standardizing TMB measurement and determining optimal thresholds for clinical applicability [3].

This article aims to explore the current understanding of TMB as a biomarker for predicting responses to immune checkpoint inhibitors. We will examine the methodologies used for assessing TMB, its correlation with clinical outcomes, and the implications for personalized cancer treatment. By highlighting the potential of TMB in guiding immunotherapy, we aim to contribute to the ongoing discourse on optimizing cancer care in the era of precision medicine [4-6].

Methodology

Data collection

A comprehensive literature review was conducted using databases such as PubMed, Scopus, and Web of Science to identify relevant studies published between 2015 and 2024. Search terms included "Tumor Mutational Burden," "Immune Checkpoint Inhibitors," "predictive biomarkers," and "cancer immunotherapy." The inclusion criteria comprised peer-reviewed articles, clinical trials, and meta-analyses focusing on the association between TMB and patient responses to ICIs [7,8].

Analysis

Studies were assessed for their methodology, sample size, tumor types, and TMB measurement techniques. The data were synthesized to evaluate the correlation between TMB levels and clinical outcomes, including overall response rates (ORR), progression-free survival (PFS), and overall survival (OS) in patients treated with ICIs [9,10].

Discussion

Understanding tumor mutational burden

TMB is defined as the total number of somatic mutations per megabase of DNA. High TMB indicates a greater number of mutations that can produce neoantigens, potentially making tumors more recognizable to the immune system. The hypothesis is that higher TMB correlates with improved response rates to ICIs, as these treatments rely on the immune system's ability to identify and attack cancer cells.

Measurement of TMB

TMB can be assessed using various genomic profiling techniques, including next-generation sequencing (NGS). NGS allows for comprehensive analysis of the tumor genome, enabling the quantification of mutations across coding regions. Different assays and panels have been developed, with varying thresholds for defining high TMB, typically ranging from 10 to 20 mutations per megabase.

TMB and immune checkpoint inhibitors

Numerous studies have investigated the relationship between TMB and responses to ICIs, such as anti-PD-1 and anti-CTLA-4 therapies. A meta-analysis conducted by Samstein et al. (2019) demonstrated that patients with high TMB had significantly higher response rates to ICIs compared to those with low TMB. Similarly, studies across various cancer types, including melanoma, non-small cell lung cancer (NSCLC), and bladder cancer, have consistently shown that elevated TMB correlates with improved clinical outcomes.

Clinical implications

The integration of TMB into clinical practice could revolutionize personalized cancer treatment. For instance, testing for TMB can help identify patients most likely to benefit from ICIs, thus avoiding unnecessary toxicity in those unlikely to respond. The FDA has approved the use of TMB as a companion diagnostic for pembrolizumab in specific indications, further emphasizing its clinical relevance.

Challenges and limitations

Despite its promise, the use of TMB as a predictive biomarker faces several challenges. Variability in TMB measurement techniques and thresholds complicates standardization across clinical settings. Additionally, while high TMB is generally associated with better responses, not all patients with high TMB benefit from ICIs, indicating that TMB is not the sole factor influencing treatment efficacy. Other factors, such as tumor microenvironment and immune cell infiltration, also play crucial roles in determining response to therapy.

Future directions

Future research should focus on refining TMB assessment methods and exploring its combination with other biomarkers, such as PD-L1 expression and tumor-infiltrating lymphocytes (TILs). Understanding the interplay between these factors will be essential for developing more robust predictive models. Moreover, large-scale clinical trials are needed to validate TMB's utility across diverse patient populations and cancer types.

Conclusion

Tumor Mutational Burden represents a significant advancement in the quest for predictive biomarkers in cancer immunotherapy. Its correlation with response rates to immune checkpoint inhibitors highlights the potential for personalized treatment approaches in oncology. While challenges remain in standardizing TMB measurement and understanding its limitations, ongoing research and clinical validation will likely enhance its utility in guiding therapeutic decisions. As our understanding of TMB evolves, it may become a cornerstone in the personalized treatment landscape for cancer patients, ultimately leading to improved outcomes and a more tailored approach to therapy.

References

  1. Fisher CS, Margenthaler JA, Hunt KK, Schwartz T (2020) The landmark series: axillary management in breast cancer.Ann Surg Oncol27: 724-729.
  2. Indexed at, Google Scholar, Crossref

  3. Freites-Martinez A, Shapiro J, Goldfarb S, Nangia J, Jimenez JJ, et al. (2019) Hair disorders in patients with cancer.J Am Acad Dermatol 80: 1179-1196.
  4. Indexed at, Google Scholar, Crossref

  5. Freites-Martinez A, Shapiro J, van den Hurk C, Goldfarb S, Jimenez JJ, et al. (2019) Hair disorders in cancer survivors.J Am Acad Dermatol 80: 1199-1213.
  6. Indexed at, Google Scholar, Crossref

  7. Ganz PA, Desmond KA, Leedham B, Rowland JH, Meyerowitz BE, et al. (2022) Quality of life in long-term, disease-free survivors of breast cancer: a follow-up study.J Natl Cancer Inst94: 39-49.
  8. Indexed at, Google Scholar, Crossref

  9. Doege D, Thong MS-Y, Koch-Gallenkamp L, Bertram H, Eberle A, et al. (2019) Health-related quality of life in long-term disease-free breast cancer survivors versus female population controls in Germany.Breast Cancer Res Treat 175: 499-510.
  10. Indexed at, Google Scholar, Crossref

  11. Engel J, Kerr J, Schlesinger-Raab A, Sauer H, Holzel D (2003) Axilla surgery severely affects quality of life: results of a 5-year prospective study in breast cancer patients.Breast Cancer Res Treat79: 47-57.
  12. Indexed at, Google Scholar, Crossref

  13. Fang S-Y, Shu B-C, Chang Y-J (2013) The effect of breast reconstruction surgery on body image among women after mastectomy: a meta-analysis.Breast Cancer Res Treat 137: 13-21.
  14. Indexed at, Google Scholar, Crossref

  15. Hamood R, Hamood H, Merhasin I, Keinan-Boker L (2018) Chronic pain and other symptoms among breast cancer survivors: prevalence, predictors, and effects on quality of life.Breast Cancer Res Treat 167: 157-169.
  16. Indexed at, Google Scholar, Crossref

  17. Hauerslev KR, Madsen AH, Overgaard J, Damsgaard TE, Christiansen P (2020) Long-term follow-up on shoulder and arm morbidity in patients treated for early breast cancer.Acta Oncol59: 851-858.
  18. Indexed at, Google Scholar, Crossref

  19. Hill G, Connelly J, Hébert R, Lindsay J, Millar W (2003) Neyman's bias re-visited.J Clin Epidemiol 56: 293-296.
  20. Indexed at, Google Scholar, Crossref

Citation: Felipe C (2024) Tumor Mutational Burden as a Biomarker: PredictingResponse to Immune Checkpoint Inhibitors. Breast Can Curr Res 9: 266.

Copyright: © 2024 Felipe C. This is an open-access article distributed under theterms of the Creative Commons Attribution License, which permits unrestricteduse, distribution, and reproduction in any medium, provided the original author andsource are credited.

Top