GET THE APP

Biomarkers of Immune Dysregulation in Precision Medicine
Journal of Inflammatory Bowel Diseases & Disorders

Journal of Inflammatory Bowel Diseases & Disorders

ISSN: 2476-1958

Open Access

Opinion - (2025) Volume 10, Issue 2

Biomarkers of Immune Dysregulation in Precision Medicine

Williams Taylor*
*Correspondence: Williams Taylor, Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden, Tel: taylor.williams@gmail.com,
Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden

Received: 01-May-2025, Manuscript No. jibdd-25-165605; Editor assigned: 03-May-2025, Pre QC No. P-165605; Reviewed: 17-May-2025, QC No. Q-165605; Revised: 22-May-2025, Manuscript No. R-165605; Published: 29-May-2025 , DOI: 10.37421/2476-1958.2025.10.236
Citation: Taylor, Williams. “Biomarkers of Immune Dysregulation in Precision Medicine.” J Inflamm Bowel Dis 10 (2025): 236.
Copyright: © 2025 Taylor W. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited.

Introduction

The advent of precision medicine has revolutionized the approach to diagnosing and treating diseases by focusing on the molecular and cellular characteristics unique to each patient. Within this paradigm, biomarkers of immune dysregulation play a pivotal role in identifying disease subtypes, predicting outcomes, and guiding individualized therapies. Immune dysregulation is a hallmark of numerous conditions, including autoimmune diseases, chronic infections, cancer, and emerging syndromes such as post-viral sequelae. By accurately characterizing immune status through specific biomarkers, clinicians and researchers can better tailor therapeutic interventions to achieve optimal efficacy and minimize adverse effects [1].

Immune biomarkers encompass a broad range of measurable indicators, including cytokines, immune cell subsets, gene expression profiles, autoantibodies, and epigenetic signatures. These markers provide insight into the functional state of the immune system, revealing patterns of activation, suppression, or imbalance. For instance, elevated levels of pro-inflammatory cytokines such as IL-6, TNF-α, and IFN-γ are indicative of hyperinflammatory states, commonly seen in autoimmune diseases and certain cancers. Conversely, markers such as PD-1 and CTLA-4, which are upregulated in exhausted T cells, signify immune suppression and are often associated with chronic infections and tumor immune evasion.

Description

In autoimmune diseases like Systemic Lupus Erythematosus (SLE) and rheumatoid arthritis, the detection of specific autoantibodies—such as anti-dsDNA, anti-Smith, and anti-CCP—serves not only as a diagnostic tool but also as a predictor of disease activity and progression. Additionally, gene expression profiling has identified interferon signatures in many autoimmune disorders, correlating with disease severity and guiding the use of interferon-targeted therapies. Such molecular stratification has transformed the clinical management of autoimmune conditions, enabling a shift from generalized immunosuppression to more targeted and tolerable treatment regimens. In oncology, immune biomarkers are critical for selecting patients for immunotherapy [2]. The expression of PD-L1 on tumor cells or infiltrating immune cells, as well as Tumor Mutational Burden (TMB) and Microsatellite Instability (MSI) status, are used to predict responsiveness to immune checkpoint inhibitors. Furthermore, the composition and activity of the tumor immune microenvironment, including the presence of cytotoxic T cells, regulatory T cells, and myeloid-derived suppressor cells, are being incorporated into multiparametric models that refine prognostication and treatment selection. These insights underscore the value of immune profiling in aligning therapeutic strategies with the unique immunological landscape of each tumor [3].

Recent advances in high-throughput technologies such as single-cell RNA sequencing, mass cytometry, and spatial transcriptomics have greatly expanded the capacity to identify and characterize immune dysregulation at an unprecedented resolution. These approaches allow for the discovery of novel biomarkers and the construction of comprehensive immune maps across various disease states. Integration of these datasets with clinical outcomes enables the development of predictive models that can inform treatment decisions in real time, advancing the goals of precision medicine [4]. In the context of infectious diseases and emerging health threats such as long COVID, immune biomarkers are being investigated to differentiate between acute, resolving, and chronic immune responses. Biomarkers such as IL-1β, IL-10, and markers of T cell exhaustion are being evaluated for their role in identifying patients at risk for persistent symptoms or immune complications. These markers may also serve as endpoints in clinical trials testing immunomodulatory therapies, facilitating the rapid translation of findings into clinical practice. The use of immune biomarkers is not without challenges. Biological variability, the influence of comorbidities, and the dynamic nature of the immune system necessitate careful interpretation and validation of biomarkers across diverse populations. Standardization of assays and the development of robust reference ranges are essential to ensure reproducibility and clinical utility. Moreover, ethical considerations regarding data privacy and access to advanced diagnostics must be addressed to ensure equitable implementation of precision medicine [5].

Conclusion

In conclusion, biomarkers of immune dysregulation are central to the advancement of precision medicine, offering critical insights into disease mechanisms, therapeutic targets, and patient stratification. As technologies continue to evolve, the integration of immune biomarkers into clinical workflows promises to enhance the accuracy of diagnoses, the personalization of treatments, and the overall quality of patient care. The continued exploration and validation of these biomarkers across different diseases will be key to realizing the full potential of immunologically informed precision medicine.

Acknowledgment

None.

Conflict of Interest

None.

References

  1. Anderson, Judy E., Lise Lotte Hansen, Frank C. Mooren and Markus Post, et al. "Methods and biomarkers for the diagnosis and prognosis of cancer and other diseases: Towards personalized medicine." Drug Resist Updates 9 (2006): 198-210.

Google Scholar Cross Ref Indexed at 

  1. Collins, C. D., S. Purohit, R. H. Podolsky and H. S. Zhao, et al. "The application of genomic and proteomic technologies in predictive, preventive and personalized medicine." Vasc Pharmacol 45 (2006): 258-267.

Google Scholar Cross Ref Indexed at

  1. Phillips, Kathryn A., Stephanie Van Bebber and Amalia M. Issa. "Diagnostics and biomarker development: Priming the pipeline." Nat Rev Drug Discov 5 (2006): 463-469.

Google Scholar Cross Ref Indexed at

  1. Crimini, Edoardo, Matteo Repetto, Philippe Aftimos and Andrea Botticelli, et al. "Precision medicine in breast cancer: From clinical trials to clinical practice." Cancer Treat Rev 98 (2021): 102223.

Google Scholar Cross Ref Indexed at

  1. Bradbury, Angela R. and Olufunmilayo I. Olopade. "Genetic susceptibility to breast cancer." Rev Endocr Metab Disord 8 (2007): 255-267.

Google Scholar Cross Ref Indexed at

Google Scholar citation report
Citations: 77

Journal of Inflammatory Bowel Diseases & Disorders received 77 citations as per Google Scholar report

Journal of Inflammatory Bowel Diseases & Disorders peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward