Opinion Article - Journal of Cancer Immunology & Therapy (2024) Volume 7, Issue 6
Emerging Biomarkers in Tumor Immunology for Personalized Cancer Treatment
Alane Gouldy *
Department of Oncology, Johns Hopkins University, United States
- *Corresponding Author:
- Alane Gouldy
> Department of Oncology, Johns Hopkins University, United States
E-mail: alanegol@jhu.edu
Received: 03-Dec -2024, Manuscript No. AAJCIT-24-155294; Editor assigned: 04-Dec-2024, PreQC No. AAJCIT-24-155294 (PQ); Reviewed:18-Dec-2024, QC No. AAJCIT-24-155294; Revised:23-Dec-2024, Manuscript No. AAJCIT-24-155294 (R); Published:28-Dec-2024, DOI:10.35841/aara-7.6.241
Citation: Gouldy A. Emerging biomarkers in tumor immunology for personalized cancer treatment. J Cancer Immunol Ther. 2024;7(6):241
Introduction
The era of personalized medicine has brought significant advancements in cancer treatment, with tumor immunology playing a pivotal role. Biomarkers that can predict response to immunotherapies are essential for tailoring treatments to individual patients, improving efficacy while minimizing unnecessary side effects. This article explores the emerging biomarkers in tumor immunology and their implications for personalized cancer treatment [1].
Tumor mutational burden, the total number of mutations in a tumor’s DNA, has emerged as a critical biomarker. Tumors with high TMB are more likely to produce neoantigens that the immune system can recognize, making them more responsive to immune checkpoint inhibitors (ICIs) such as anti-PD-1 and anti-CTLA-4 therapies. While TMB is not universally predictive across all cancers, its utility is particularly evident in melanoma and non-small cell lung cancer (NSCLC) [2].
Programmed death-ligand 1 (PD-L1) expression on tumor and immune cells is a widely studied biomarker for predicting responses to PD-1/PD-L1 inhibitors. High levels of PD-L1 often correlate with better responses to these therapies. However, PD-L1 testing has limitations, including variability in detection methods and expression heterogeneity within tumors [3].
MSI and dMMR are genetic features that lead to the accumulation of mutations, generating neoantigens that can activate the immune system. These biomarkers have proven predictive of immunotherapy efficacy, particularly in colorectal cancer and other MSI-high tumors. The FDA has approved pembrolizumab, a PD-1 inhibitor, for treating MSI-high or dMMR cancers, regardless of tumor origin [4].
The presence and activity of tumor-infiltrating lymphocytes are strong indicators of an ongoing immune response. High levels of cytotoxic CD8+ TILs are associated with better outcomes in various cancers and improved responses to immunotherapies. Advances in imaging and single-cell analysis are enhancing our ability to quantify and characterize TILs, enabling more precise predictions of treatment response [5].
Neoantigens, derived from tumor-specific mutations, are pivotal in eliciting an anti-tumor immune response. A high neoantigen load is associated with better responses to ICIs. Personalized neoantigen vaccines, which stimulate the immune system against these unique targets, are currently under clinical investigation [6].
Gene expression profiles can reveal the immune landscape of tumors. For example, an “immune-hot” tumor characterized by high expression of immune activation genes often responds better to ICIs compared to “immune-cold” tumors with low immune activity. Tools like RNA sequencing and multiplex immunohistochemistry are facilitating the development of immune gene signatures for clinical use [7].
These immune checkpoints are gaining attention as targets for next-generation ICIs. Biomarkers assessing their expression could predict responses to therapies targeting these pathways. The gut microbiome’s influence on systemic immunity has revealed its potential as a biomarker. Specific bacterial profiles are associated with better immunotherapy outcomes, and interventions to modulate the microbiome are being explored [8].
Liquid biopsies detecting ctDNA can provide real-time insights into tumor mutational profiles and monitor treatment responses dynamically. While the identification of biomarkers holds great promise, challenges remain: Tumor heterogeneity complicates biomarker assessment and may lead to inconsistent predictions [9].
Lack of standardized assays and methodologies for biomarker detection hinders their widespread adoption. High costs and limited availability of advanced diagnostic tools can restrict access to biomarker-driven therapies. Ongoing research aims to address these challenges by developing robust, reproducible, and cost-effective biomarker tests. Integration of multi-omics data combining genomic, transcriptomic, and proteomic analysesis expected to provide a holistic understanding of tumor-immune interactions [10].
Conclusion
Emerging biomarkers in tumor immunology are transforming personalized cancer treatment, offering the potential to optimize therapeutic strategies and improve patient outcomes. By identifying the right patients for the right therapies, these biomarkers are paving the way for more effective and precise cancer care. As research advances, the integration of novel biomarkers into clinical practice will continue to enhance the impact of immunotherapy on cancer treatment.
References
- Hortobagyi GN, de la Garza Salazar J, Pritchard K, et al. ABREAST Investigators The global breast cancer burden: variations in epidemiology and survival. Clin Breast Can. 2005;6(5):391–401.
- Parkin DM, Fernández LM. Use of statistics to assess the global burden of breast cancer.Breast J. 2006;12(s1):S70–S80.
- Mavaddat N, Pharoah PD, Michailidou K, et al. Prediction of breast cancer risk based on profiling with common genetic variants. JNCI: J Natl Cancer Inst. 2015;107(5):djv036.
- Khakbazan Z, Taghipour A, Latifnejad Roudsari R, et al. Help seeking behavior of women with self-discovered breast cancer symptoms: a meta-ethnographic synthesis of patient delay.PLoS One. 2014;9(12):e110262.
- Li L, Zhong Y, Zhang H, et al. Association between oral contraceptive use as a risk factor and triple-negative breast cancer: A systematic review and meta-analysis. Mol Clin Oncol. 2017;7(1):76–80.
- Carethers JM, Braun J, Sands BE. Genetics, genetic testing, and biomarkers of digestive diseases.2015;149:1131–3.
- Cheng J, Wang W, Sun C, et al. Meta-analysis of the prognostic and diagnostic significance of serum/plasma osteopontin in hepatocellular carcinoma.J Clin Gastroenterol.2014;48:806–14.
- Chimparlee N, Chuaypen N, Khlaiphuengsin A, et al. Diagnostic and prognostic roles of serum osteopontin and osteopontin promoter polymorphisms in hepatitis B-related hepatocellular carcinoma.Asian Pac J Cancer Prev.2015;16:7211–7.
- Lanier LL. Up on the tightrope: Natural killer cell activation and inhibition.Nat Immunol. 2008;9(5):495–502.
- Clavien PA, Lesurtel M, Bossuyt PM, et al. Recommendations for liver transplantation for hepatocellular carcinoma:an international consensus conference report.Lancet Oncol.2012;13:e11–22.
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at,Google Scholar, Cross Ref
Indexed at,Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref