GET THE APP

Unveiling Regorafenib Responsiveness and Molecular Mechanisms in Recurrent Glioblastoma Tumors
..

Molecular Biomarkers & Diagnosis

ISSN: 2155-9929

Open Access

Perspective - (2024) Volume 15, Issue 1

Unveiling Regorafenib Responsiveness and Molecular Mechanisms in Recurrent Glioblastoma Tumors

Joanne Sara*
*Correspondence: Joanne Sara, Department of Analytical Chemistry, Ankara University, Ankara, Turkey, Email:
Department of Analytical Chemistry, Ankara University, Ankara, Turkey

Received: 27-Jan-2024, Manuscript No. jmbd-24-130655; Editor assigned: 30-Jan-2024, Pre QC No. P-130655; Reviewed: 13-Feb-2024, QC No. Q-130655; Revised: 19-Feb-2024, Manuscript No. R-130655; Published: 29-Feb-2024 , DOI: 10.37421/2155-9929.2024.15.628
Citation: Sara, Joanne. “Unveiling Regorafenib Responsiveness and Molecular Mechanisms in Recurrent Glioblastoma Tumors.” J Mol Biomar Diagn 15 (2024): 628.
Copyright: © 2024 Sara J. 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

Glioblastoma (GBM) remains one of the most challenging cancers to treat due to its aggressive nature and high recurrence rates. Despite advancements in therapy, the prognosis for recurrent GBM remains dismal. Regorafenib, a multi-targeted tyrosine kinase inhibitor, has shown promise in various cancers, including GBM. This article explores the responsiveness of regorafenib in recurrent GBM and delves into the underlying molecular mechanisms that govern its efficacy. Glioblastoma is the most common and aggressive primary brain tumor in adults, characterized by its infiltrative growth, high proliferation rate and resistance to therapy. Despite aggressive treatment strategies involving surgery, radiation and chemotherapy, GBM almost invariably recurs, leading to poor patient outcomes. Recurrent GBM poses a significant therapeutic challenge due to acquired resistance mechanisms and limited treatment options. Novel therapeutic approaches are urgently needed to improve outcomes for patients with recurrent GBM.

Regorafenib is an oral multi-kinase inhibitor that targets various receptor tyrosine kinases involved in tumor angiogenesis, oncogenesis and the tumor microenvironment. Preclinical studies have demonstrated promising antitumor activity of regorafenib in GBM models, leading to its evaluation in clinical trials. However, the responsiveness of regorafenib in recurrent GBM and the underlying molecular mechanisms of its action remain incompletely understood [1].

Description

Clinical trials evaluating the efficacy of regorafenib in recurrent GBM have shown encouraging results, albeit with modest improvements in Progression- Free Survival (PFS) and Overall Survival (OS). A phase II trial demonstrated that regorafenib monotherapy achieved a disease control rate of 38% and a median PFS of 3.7 months in patients with recurrent GBM, indicating its potential as a treatment option in this setting. Subsequent studies have explored the combination of regorafenib with other agents or radiation therapy to enhance its therapeutic efficacy. The modest clinical benefit observed with regorafenib in recurrent GBM highlights the need for a deeper understanding of its molecular mechanisms of action and identification of predictive biomarkers to select patients who are most likely to benefit from treatment [2].

Regorafenib exerts its anti-tumor effects through the inhibition of multiple signaling pathways involved in tumor growth, angiogenesis and immune evasion. Key targets of regorafenib include Vascular Endothelial Growth Factor Receptor (VEGFR), Platelet-Derived Growth Factor Receptor (PDGFR), Fibroblast Growth Factor Receptor (FGFR) and oncogenic kinases such as RAF, RET and KIT [3]. In GBM, dysregulated signaling pathways contribute to tumor progression and therapy resistance. Regorafenib targets these pathways, leading to inhibition of tumor growth, angiogenesis and invasion. Preclinical studies have demonstrated that regorafenib inhibits GBM cell proliferation, induces apoptosis and suppresses tumor angiogenesis through blockade of VEGFR and PDGFR signaling. Furthermore, regorafenib modulates the tumor immune microenvironment by inhibiting immunosuppressive pathways and enhancing anti-tumor immune responses. By targeting immunosuppressive Myeloid-Derived Suppressor Cells (MDSCs) and regulatory T cells regorafenib promotes the activation of Cytotoxic T Lymphocytes (CTLs) and Natural Killer (NK) cells, leading to enhanced anti-tumor immunity [4,5].

Conclusion

Regorafenib holds promise as a therapeutic option for recurrent GBM, offering modest clinical benefit and manageable toxicity. However, further research is needed to optimize its use in this setting and improve patient outcomes. Future studies should focus on identifying predictive biomarkers of regorafenib responsiveness, elucidating resistance mechanisms and exploring rational combination strategies to enhance its efficacy. Moreover, advancements in molecular profiling techniques, such as next-generation sequencing and single-cell analysis, will facilitate the identification of patient subgroups most likely to benefit from regorafenib therapy. Personalized treatment approaches based on molecular signatures and tumor microenvironment characteristics may maximize the clinical utility of regorafenib in recurrent GBM. In summary, regorafenib represents a valuable addition to the armamentarium against recurrent GBM, offering a novel therapeutic strategy with the potential to improve patient outcomes. Continued research efforts aimed at unraveling its molecular mechanisms of action and optimizing its clinical use are warranted to address the unmet medical need in recurrent GBM management.

Acknowledgement

None.

Conflict of Interest

There are no conflicts of interest by author.

References

  1. Ostrom, Quinn T., Nirav Patil, Gino Cioffi and Kristin Waite, et al. "CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2013–2017." Neuro Oncol 22 (2020): iv1-iv96.

    Google Scholar, Crossref, Indexed at

  2. Stupp, Roger, Warren P. Mason, Martin J. Van Den Bent and Michael Weller, et al. "Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma." N Eng J Med 352 (2005): 987-996.

    Google Scholar, Crossref, Indexed at

  3. Rønning, Pål A., Eirik Helseth, Torstein R. Meling and Tom B. Johannesen. "A population-based study on the effect of temozolomide in the treatment of glioblastoma multiforme." Neuro Oncol 14 (2012): 1178-1184.

    Google Scholar, Crossref, Indexed at

  4. Batchelor, Tracy T., Elizabeth R. Gerstner, Xiaobu Ye and Serena Desideri, et al. "Feasibility, phase I and phase II studies of tandutinib, an oral platelet-derived growth factor receptor-β tyrosine kinase inhibitor, in patients with recurrent glioblastoma." Neuro Oncol 19 (2017): 567-575.

    Google Scholar, Crossref, Indexed at

  5. Bruix, Jordi, Shukui Qin, Philippe Merle and Alessandro Granito, et al. "Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): A randomised, double-blind, placebo-controlled, phase 3 trial." Lancet 389 (2017): 56-66.

    Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 2054

Molecular Biomarkers & Diagnosis received 2054 citations as per Google Scholar report

Molecular Biomarkers & Diagnosis peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward