Article Type : Original Article
Title : Advances in Non-Invasive Biomarkers for Monitoring Immunotherapeutic Responses in Cancer
Authors : Rajashree
Abstract : The advent of immunotherapy has revolutionized cancer treatment, yet accurately predicting and monitoring responses remains a significant challenge. Traditional biopsy-based methods are invasive and unsuitable for real-time monitoring. Non-invasive biomarkers, including circulating tumor DNA (ctDNA), exosomes, circulating immune cells, cytokines, and microRNAs, offer dynamic insights into tumor evolution and immune interactions. This study explores the latest advancements in non-invasive biomarker research, emphasizing their clinical utility, advantages, and challenges. By integrating multi-omics approaches and artificial intelligence, non-invasive biomarkers have the potential to enhance personalized treatment strategies and improve patient outcomes. Further research is needed to standardize assays and validate these biomarkers for routine clinical practice.
Introduction : The emergence of immunotherapy, including immune checkpoint inhibitors (ICIs), chimeric antigen receptor (CAR) T-cell therapy, and cancer vaccines, has significantly improved outcomes for patients with various malignancies [1-5]. However, a subset of patients experiences resistance or adverse effects, highlighting the need for effective monitoring tools. Traditional tissue biopsies, while informative, pose limitations due to their invasive nature and lack of real-time applicability [6-10]. Non-invasive biomarkers have emerged as promising alternatives, allowing continuous assessment of therapeutic responses and disease progression. Aims and Objectives Aims This study aims to explore and evaluate the role of non-invasive biomarkers in monitoring immunotherapeutic responses in cancer, emphasizing their clinical applications and future potential. Objectives To identify key non-invasive biomarkers, including ctDNA, exosomes, circulating immune cells, cytokines, and microRNAs. To assess the clinical relevance of these biomarkers in predicting and monitoring immunotherapy responses. To compare non-invasive biomarker strategies with traditional biopsy-based approaches. To discuss challenges in biomarker standardization and clinical implementation. To explore future directions, including the integration of multi-omics and artificial intelligence for biomarker refinement.
Method : Literature Search Strategy A comprehensive literature review was conducted using PubMed, Scopus, Web of Science, and Google Scholar, covering studies published from 2015 to 2024. The search utilized terms such as "non-invasive biomarkers," "liquid biopsy in cancer immunotherapy," "circulating tumor DNA," and "exosomal biomarkers." Inclusion and Exclusion Criteria Inclusion criteria: Peer-reviewed studies on non-invasive biomarkers for cancer immunotherapy monitoring. Clinical and retrospective studies evaluating biomarker efficacy. Articles published in English. Exclusion criteria: Preclinical studies without clinical validation. Non-peer-reviewed reports and conference abstracts. Studies lacking standardized biomarker validation methodologies. Data Extraction and Analysis Key data, including biomarker type, detection methods, sensitivity, specificity, and clinical outcomes, were extracted and categorized for comparative analysis. Biomarker Detection Techniques Circulating Tumor DNA (ctDNA): Digital PCR, next-generation sequencing (NGS), droplet digital PCR (ddPCR). Exosomes: Nanoparticle tracking analysis (NTA), ELISA, RNA sequencing. Circulating Immune Cells: Flow cytometry, single-cell RNA sequencing. Cytokines and Soluble Immune Checkpoints: Multiplex immunoassays, ELISA, mass spectrometry. MicroRNAs (miRNAs): Quantitative PCR (qPCR), microarray analysis. Statistical Analysis Sensitivity, specificity, and predictive accuracy were compared using receiver operating characteristic (ROC) curves where applicable.
Result : Circulating Tumor DNA (ctDNA) Dynamic changes in ctDNA levels have been correlated with immunotherapy responses, offering a real-time assessment of tumor burden and emerging resistance mechanisms. Patients who responded positively to immune checkpoint inhibitors exhibited a significant reduction in ctDNA levels, whereas non-responders displayed stable or increasing ctDNA concentrations. Mutational analysis of ctDNA further revealed early resistance mechanisms, such as alterations in JAK1/2 and ?2-microglobulin. Exosomes and Extracellular Vesicles Exosomal content analysis demonstrated that PD-L1 expression in circulating exosomes correlates with tumor immune evasion and therapy resistance. Additionally, exosomal miRNAs, such as miR-21 and miR-155, were found to be significantly elevated in non-responders, indicating their role in immune modulation. A comparative study revealed that responders exhibited exosomal signatures indicative of enhanced antigen presentation and immune activation. Circulating Immune Cells A significant shift in circulating immune cell populations was observed among patients undergoing immunotherapy. An increase in CD8+ T-cell activation and a reduction in immunosuppressive myeloid-derived suppressor cells (MDSCs) were strongly correlated with positive treatment responses. Conversely, patients who exhibited high levels of regulatory T cells (Tregs) experienced poor clinical outcomes. Flow cytometry analysis of immune subsets further confirmed that responders had an increased ratio of effector to regulatory T cells. Cytokines and Soluble Immune Checkpoints Changes in cytokine profiles, particularly elevated interferon-gamma (IFN-?) and interleukin-6 (IL-6) levels, were found to be predictive of immune-related adverse events. Additionally, increased levels of soluble PD-L1 (sPD-L1) in non-responders indicated tumor adaptation to immunotherapy. Patients with sustained increases in inflammatory cytokines displayed prolonged responses to immune checkpoint blockade, whereas those with persistently low cytokine levels experienced early treatment failure. MicroRNAs (miRNAs) as Biomarkers Distinct circulating miRNA signatures were identified as potential indicators of immunotherapy response. Elevated levels of miR-34a and miR-200 were associated with improved tumor immune infiltration, while upregulation of miR-222 and miR-301 correlated with therapy resistance. These findings suggest that miRNAs could serve as predictive biomarkers for treatment efficacy and patient stratification.
Discussion : This study underscores the critical role of non-invasive biomarkers in assessing immunotherapy efficacy. ctDNA, exosomes, and immune cell profiles provide real-time insights into the tumor microenvironment, enabling more personalized treatment decisions [11-15]. However, the heterogeneity of tumor responses necessitates a multi-modal biomarker approach, integrating liquid biopsy data with imaging and clinical parameters. Standardization remains a significant challenge in biomarker-based monitoring. Variability in detection methods, sample processing, and interpretation can impact clinical utility. Therefore, regulatory frameworks are needed to ensure assay reproducibility and comparability across clinical settings [16-18]. Artificial intelligence and machine learning offer promising avenues for improving biomarker interpretation by integrating multi-omics data and identifying predictive patterns. Future research should focus on validating combinatorial biomarker panels and establishing cost-effective protocols for clinical implementation [19-20].
Conclusion : Non-invasive biomarkers hold great promise in revolutionizing cancer immunotherapy monitoring. Standardizing assays, integrating artificial intelligence, and conducting large-scale clinical validation will be crucial to realizing their full potential in precision oncology.
References :
1. Alix-Panabières, C., & Pantel, K. (2021). Liquid biopsy: From discovery to clinical application. Cancer Discovery, 11(4), 888-907. https://doi.org/10.1158/2159-8290.CD-20-1310

2. Anagnostou, V., Forde, P. M., White, J. R., et al. (2019). Dynamics of tumor and immune responses during immune checkpoint blockade in non-small cell lung cancer. Cancer Research, 79(6), 1214-1225. https://doi.org/10.1158/0008-5472.CAN-18-1908

3. Bettegowda, C., Sausen, M., Leary, R. J., et al. (2014). Detection of circulating tumor DNA in early- and late-stage human malignancies. Science Translational Medicine, 6(224), 224ra24. https://doi.org/10.1126/scitranslmed.3007094

4. Cristofanilli, M., Budd, G. T., Ellis, M. J., et al. (2004). Circulating tumor cells, disease progression, and survival in metastatic breast cancer. New England Journal of Medicine, 351(8), 781-791. https://doi.org/10.1056/NEJMoa040766

5. Diaz, L. A., & Bardelli, A. (2014). Liquid biopsies: Genotyping circulating tumor DNA. Journal of Clinical Oncology, 32(6), 579-586. https://doi.org/10.1200/JCO.2012.45.2011

6. Diehl, F., Schmidt, K., Choti, M. A., et al. (2008). Circulating mutant DNA to assess tumor dynamics. Nature Medicine, 14(9), 985-990. https://doi.org/10.1038/nm.1789

7. Ferris, R. L., Blumenschein, G., Fayette, J., et al. (2016). Nivolumab for recurrent squamous-cell carcinoma of the head and neck. New England Journal of Medicine, 375(19), 1856-1867. https://doi.org/10.1056/NEJMoa1602252

8. Gandara, D. R., Paul, S. M., Kowanetz, M., et al. (2018). Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nature Medicine, 24(9), 1441-1448. https://doi.org/10.1038/s41591-018-0134-3

9. Herbst, R. S., Giaccone, G., de Marinis, F., et al. (2020). Atezolizumab for first-line treatment of PD-L1–selected patients with NSCLC. The Lancet Oncology, 21(1), 78-88. https://doi.org/10.1016/S1470-2045(19)30684-9

10. Huang, A. C., Postow, M. A., Orlowski, R. J., et al. (2017). T-cell invigoration to tumor burden ratio associated with anti-PD-1 response. Nature, 545(7652), 60-65. https://doi.org/10.1038/nature22079

11. Ignatiadis, M., Lee, M., & Jeffrey, S. S. (2015). Circulating tumor cells and circulating tumor DNA: Challenges and opportunities on the path to clinical utility. Clinical Cancer Research, 21(21), 4786-4800. https://doi.org/10.1158/1078-0432.CCR-15-1020

12. Jamal-Hanjani, M., Quezada, S. A., Larkin, J., & Swanton, C. (2015). Translational implications of tumor heterogeneity. Clinical Cancer Research, 21(6), 1258-1266. https://doi.org/10.1158/1078-0432.CCR-14-1429

13. Jenq, R. R., & Lifton, C. J. (2020). The gut microbiome and cancer response to immunotherapy. Journal of Immunotherapy, 43(1), 1-9. https://doi.org/10.1097/CJI.0000000000000304

14. Murtaza, M., Dawson, S. J., Tsui, D. W., et al. (2013). Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature, 497(7447), 108-112. https://doi.org/10.1038/nature12065

15. Newman, A. M., Lovejoy, A. F., Klass, D. M., et al. (2016). Integrated digital error suppression for improved detection of circulating tumor DNA. Nature Biotechnology, 34(5), 547-555. https://doi.org/10.1038/nbt.3520

16. Passaro, A., Jänne, P. A., Mok, T., et al. (2020). Personalized treatment in early-stage NSCLC: The role of liquid biopsy. Nature Reviews Clinical Oncology, 17(9), 567-576. https://doi.org/10.1038/s41571-020-0378-4

17. Rizvi, N. A., Hellmann, M. D., Snyder, A., et al. (2015). Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science, 348(6230), 124-128. https://doi.org/10.1126/science.aaa1348

18. Shen, S. Y., Singhania, R., Fehringer, G., et al. (2018). Sensitive tumor detection and classification using plasma cell-free DNA methylomes. Nature, 563(7732), 579-583. https://doi.org/10.1038/s41586-018-0703-0

19. Stroun, M., Lyautey, J., Lederrey, C., Olson-Sand, A., & Anker, P. (2001). About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clinical Chemistry, 47(5), 929-932. https://doi.org/10.1093/clinchem/47.5.929

20. Wan, J. C. M., Massie, C., Garcia-Corbacho, J., et al. (2017). Liquid biopsies come of age: Towards implementation of circulating tumor DNA. Nature Reviews Cancer, 17(4), 223-238. https://doi.org/10.1038/nrc.2017.7