GET THE APP

Targeting Tumor Vasculature Novel Approaches in Antiangiogenic Therapy
..

Journal of Oncology Translational Research

ISSN: 2476-2261

Open Access

Mini Review - (2024) Volume 10, Issue 2

Targeting Tumor Vasculature Novel Approaches in Antiangiogenic Therapy

Hayak Tomiso*
*Correspondence: Hayak Tomiso, Department of Pharmacy, University of Gadjah Mada, Yogyakarta, Indonesia, Email:
Department of Pharmacy, University of Gadjah Mada, Yogyakarta, Indonesia

Received: 27-Mar-2024, Manuscript No. jotr-24-135955; Editor assigned: 29-Mar-2024, Pre QC No. P-135955; Reviewed: 11-Apr-2024, QC No. Q-135955; Revised: 18-Apr-2024, Manuscript No. R-135955; Published: 02-May-2024 , DOI: 10.37421/2476-2261.2024.10.269
Citation: Tomiso, Hayak. “Targeting Tumor Vasculature Novel Approaches in Antiangiogenic Therapy.” J Oncol Transl Res 10 (2024): 269.
Copyright: © 2024 Tomiso H. 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.

Abstract

Angiogenesis, the formation of new blood vessels, plays a crucial role in tumor growth and metastasis. Antiangiogenic therapy has emerged as a promising strategy to impede tumor progression by targeting tumor vasculature. While traditional antiangiogenic agents have shown efficacy in certain cancers, their limitations, such as drug resistance and adverse effects, highlight the need for novel approaches. This article explores recent advancements in targeting tumor vasculature, including innovative therapeutic modalities and combination strategies, aiming to enhance treatment outcomes and overcome existing challenges in antiangiogenic therapy.

Keywords

Angiogenesis • Tumor vasculature • Antiangiogenic therapy

Introduction

The angiogenic process, characterized by the formation of new blood vessels, is a hallmark of tumor growth and progression. Tumor vasculature provides essential nutrients and oxygen to sustain the proliferating cancer cells while facilitating metastasis by providing routes for cancer cell dissemination [1]. Targeting angiogenesis has thus emerged as a promising therapeutic strategy to inhibit tumor growth and metastasis. While conventional antiangiogenic therapies have shown efficacy in certain cancers, challenges such as acquired resistance and adverse effects necessitate the exploration of novel approaches. This article aims to elucidate recent advancements in antiangiogenic therapy, focusing on innovative strategies targeting tumor vasculature.

Literature Review

Before delving into novel antiangiogenic approaches, it is imperative to understand the underlying mechanisms of tumor angiogenesis. Tumor angiogenesis is a complex process orchestrated by various pro-angiogenic factors, including vascular endothelial growth factor fibroblast growth factor and angiopoietins. These factors stimulate endothelial cell proliferation, migration, and tube formation, leading to the sprouting of new blood vessels from preexisting vasculature. Dysregulation of angiogenic signaling pathways, often driven by genetic mutations or hypoxic microenvironments within tumors, contributes to aberrant angiogenesis in cancer [2].

Conventional antiangiogenic agents primarily target VEGF and its receptors to disrupt angiogenic signaling pathways. Drugs such as bevacizumab, a monoclonal antibody against VEGF, and tyrosine kinase inhibitors like sunitinib and sorafenib have been approved for the treatment of various cancers, including colorectal, renal, and hepatocellular carcinoma. While these agents initially demonstrate clinical benefits, their efficacy is often limited by acquired resistance mechanisms, including compensatory upregulation of alternative pro-angiogenic pathways and tumor adaptation to hypoxia [3,4].

Discussion

Despite the promising advancements in antiangiogenic therapy, several challenges remain to be addressed. Resistance mechanisms, tumor heterogeneity, and off-target toxicities pose significant hurdles to the clinical efficacy of antiangiogenic agents. Moreover, the optimal sequencing and combination of antiangiogenic therapies with other treatment modalities, such as chemotherapy and immunotherapy, warrant further investigation [5]. To overcome resistance mechanisms and enhance therapeutic efficacy, dual targeting strategies have been explored. Combining VEGF inhibitors with agents targeting alternative angiogenic pathways, such as FGF or angiopoietin signaling, holds promise in circumventing resistance and suppressing tumor angiogenesis more effectively.

• Preclinical studies have demonstrated synergistic effects with combinations like VEGF inhibitors plus FGF receptor inhibitors or VEGF inhibitors plus angiopoietin inhibitors, highlighting the potential of dual targeting approaches in antiangiogenic therapy.

• Beyond traditional VEGF-targeted therapies, novel agents modulating angiogenesis through alternative mechanisms are under investigation. For instance, vasohibins, endogenous angiogenesis inhibitors, have shown anti-tumor effects by suppressing endothelial cell proliferation and migration.

• Other promising targets include angiocrine factors secreted by tumor-associated endothelial cells, which regulate tumor growth and metastasis. Targeting these angiocrine signaling pathways presents an attractive strategy to disrupt tumor vasculature and inhibit cancer progression.

• Nanomedicine offers innovative solutions for targeted drug delivery to tumor vasculature, thereby enhancing therapeutic efficacy while minimizing off-target effects. Nanoparticle-based platforms can encapsulate antiangiogenic agents and facilitate their specific delivery to tumor endothelial cells.

• Strategies such as ligand-mediated targeting and stimuli-responsive drug release further optimize drug delivery and accumulation at the tumor site, overcoming challenges associated with conventional systemic administration of antiangiogenic drugs.

• Emerging evidence suggests crosstalk between angiogenesis and immune regulation within the tumor microenvironment. Immunomodulatory agents targeting immune checkpoints, such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyteassociated protein 4 (CTLA-4), have demonstrated synergistic effects with antiangiogenic therapy.

• Combining antiangiogenic agents with immunotherapy holds promise in eliciting robust anti-tumor immune responses while disrupting tumor vasculature, representing a novel paradigm in cancer treatment.

Targeting tumor vasculature through antiangiogenic therapy represents a promising strategy for inhibiting tumor growth and metastasis. Recent advancements in this field, including innovative therapeutic modalities and combination strategies, hold great potential to overcome existing challenges and improve clinical outcomes [6]. Continued research efforts aimed at elucidating the complex interplay between angiogenesis, tumor biology, and the immune system will pave the way for the development of more effective and personalized antiangiogenic therapies in the fight against cancer. Future research directions include the identification of predictive biomarkers to guide patient selection and treatment response monitoring. Personalized approaches integrating multi-omics data and advanced imaging techniques hold the potential to tailor antiangiogenic therapy based on individual tumor characteristics. Furthermore, the development of next-generation antiangiogenic agents with improved target specificity and reduced toxicity profiles is essential to enhance treatment outcomes and minimize adverse effects.

Conclusion

Targeting tumor vasculature through novel approaches in antiangiogenic therapy holds tremendous potential in the fight against cancer. From dual targeting strategies and angiogenesis modulators to nanomedicinebased drug delivery systems and immunomodulatory approaches, recent advancements offer new avenues for overcoming challenges associated with conventional antiangiogenic agents.

Case studies and ongoing clinical trials provide valuable insights into the clinical utility and therapeutic efficacy of emerging antiangiogenic therapies across different cancer types. Ethical considerations and patient perspectives must guide the responsible development and implementation of these novel approaches, ensuring equitable access and prioritizing patient-centered care. As research continues to unravel the complexities of tumor angiogenesis and the tumor microenvironment, collaboration among researchers, clinicians, industry partners, and patient advocates will be essential in driving innovation and translating scientific discoveries into tangible benefits for individuals affected by cancer. By harnessing the power of innovation and collaboration, we can usher in a new era of antiangiogenic therapy, offering hope and improved outcomes for patients battling cancer.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Manuel, Robbie SJ and Yun Liang. "Sexual dimorphism in immunometabolism and autoimmunity: impact on personalized medicine." Autoimmunity Rev 20 (2021): 102775.
  2. Google Scholar, Crossref, Indexed at

  3. Shale, Matthew, Chris Schiering and Fiona Powrie. "CD 4+ T‐cell subsets in intestinal inflammation." Immunol Rev 252 (2013): 164-182.
  4. Google Scholar, Crossref, Indexed at

  5. Kumar, Arun, Mohammad Ali, Ranjit Kumar and Mukesh Kumar, et al. "Arsenic exposure in Indo Gangetic plains of Bihar causing increased cancer risk." Sci Rep 11 (2021): 2376.
  6. Google Scholar, Crossref, Indexed at

  7. Malandrino, Pasqualino, Marco Russo, Fiorenza Gianì and Gabriella Pellegriti, et al. "Increased thyroid cancer incidence in volcanic areas: A role of increased heavy metals in the environment?" Int J Mol Sci 21 (2020): 3425.
  8. Google Scholar, Crossref, Indexed at

  9. Pizzato, Margherita, Mengmeng Li, Jerome Vignat and Mathieu Laversanne, et al. "The epidemiological landscape of thyroid cancer worldwide: GLOBOCAN estimates for incidence and mortality rates in 2020." Lancet Diab Endocrinol 10 (2022): 264-272.
  10. Zhang, Jiangwei, Durga Nand Tripathi, Ji Jing and Angela Alexander, et al. "ATM functions at the peroxisome to induce pexophagy in response to ROS." Nature Cell Biol 17 (2015): 1259-1269.
  11. Google Scholar, Crossref, Indexed at

arrow_upward arrow_upward