GET THE APP

The Clinical Implications of Tumor Suppressor Inactivation in Cancer Therapy
..

Reports in Thyroid Research

ISSN: 2684-4273

Open Access

Mini Review - (2023) Volume 7, Issue 3

The Clinical Implications of Tumor Suppressor Inactivation in Cancer Therapy

Feraco Angela*
*Correspondence: Feraco Angela, Department of Integrative Biology, University of California, Berkeley, USA, Email:
Department of Integrative Biology, University of California, Berkeley, USA

Received: 22-Aug-2023, Manuscript No. rtr-23-116754; Editor assigned: 24-Aug-2023, Pre QC No. P-116754; Reviewed: 07-Sep-2023, QC No. Q-116754; Revised: 12-Sep-2023, Manuscript No. R-116754; Published: 19-Sep-2023 , DOI: 10.37421/2684-4273.2023.7.51
Citation: Angela, Feraco. “The Clinical Implications of Tumor Suppressor Inactivation in Cancer Therapy” Rep Thyroid Res 7 (2023): 51.
Copyright: © 2023 Angela F. 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

Tumor suppressor genes are critical components of the cellular machinery that regulates cell growth and prevents the development of cancer. Inactivation or mutation of these genes is a hallmark of many cancer types and plays a central role in tumorigenesis. This article reviews the clinical implications of tumor suppressor inactivation in cancer therapy, focusing on the therapeutic strategies that have emerged to target tumor suppressor-deficient tumors. We discuss the impact of inactivated tumor suppressors on cancer prognosis, treatment resistance, and potential therapeutic vulnerabilities. Furthermore, we explore various therapeutic modalities, including synthetic lethality, immune checkpoint inhibitors, and gene therapy, designed to exploit the consequences of tumor suppressor inactivation. The evolving landscape of precision medicine and personalized therapy in the context of tumor suppressor status is also examined. As our understanding of tumor suppressors and their inactivation deepens, the development of novel therapeutic approaches and the optimization of existing treatments offer new hope for cancer patients.

Keywords

Tumor suppressor • Tumors • Proliferation

Introduction

Tumor suppressor genes, such as p53, BRCA1 and PTEN, are vital components of the cell's defense mechanism against uncontrolled proliferation and malignant transformation. Inactivation or mutation of these genes is a frequent occurrence in many cancers and is often associated with a poor clinical outcome. The loss of tumor suppressor function can disrupt essential cellular processes, making tumors more aggressive and less responsive to traditional cancer therapies. In this article, we delve into the clinical implications of tumor suppressor inactivation in cancer therapy, addressing how these alterations affect patient prognosis and treatment responses [1].

Literature Review

The status of tumor suppressor genes can significantly impact clinical prognosis. Inactivation or loss of these genes is often associated with a more advanced cancer stage, higher tumor grade and increased risk of metastasis. Patients with tumor suppressor-deficient tumors may have a less favorable prognosis and lower overall survival rates. Understanding the genetic landscape of a patient's tumor, particularly the status of tumor suppressor genes, has become pivotal in guiding treatment decisions and predicting clinical outcomes. One of the most challenging aspects of managing tumor suppressor-deficient tumors is their propensity for treatment resistance. The loss of these genes can confer resistance to conventional treatments like chemotherapy and radiation therapy. For instance, p53-deficient tumors often show resistance to DNA-damaging agents due to impaired apoptotic pathways. Similarly, BRCA1/2 mutations can result in resistance to certain DNA repairtargeted therapies [2,3].

In recent years, synthetic lethality has emerged as a promising therapeutic strategy for tumor suppressor-deficient tumors. Synthetic lethal interactions involve targeting a gene or pathway that becomes essential for the survival of cells lacking a specific tumor suppressor. For example, poly (ADP-ribose) polymerase (PARP) inhibitors exploit synthetic lethality in BRCA1/2-mutated cancers, leading to the accumulation of DNA damage and selective cancer cell death. These targeted therapies have shown significant clinical benefit in patients with tumor suppressor-deficient tumors. The tumor microenvironment of tumor suppressor-deficient tumors often exhibits unique immune characteristics. Tumor suppressor inactivation can lead to increased mutational burden, rendering the tumor more immunogenic. This has paved the way for the use of immune checkpoint inhibitors, such as anti-PD-1 and anti-CTLA-4 antibodies, in the treatment of these tumors. Immunotherapy has demonstrated promising results, particularly in some advanced malignancies with tumor suppressor gene alterations [4].

Discussion

Emerging gene therapy approaches aim to restore or compensate for the loss of tumor suppressor function. These therapies include the delivery of functional tumor suppressor genes to tumor cells, gene editing techniques, and the use of viral vectors to correct genetic defects. While these strategies are still in the experimental stage, they hold potential for the treatment of tumor suppressor-deficient tumors and may revolutionize cancer therapy in the future. The advent of precision medicine has led to a paradigm shift in cancer therapy. The ability to profile the genomic and molecular characteristics of tumors, including tumor suppressor gene status, allows for tailored treatment regimens. Targeted therapies designed to exploit specific genetic alterations have demonstrated substantial clinical benefits, especially in patients with tumor suppressor-deficient tumors. The ongoing research in this field continues to refine our understanding of tumor suppressor inactivation in cancer therapy, offering hope for improved patient outcomes and more effective treatments [5,6].

Conclusion

Tumor suppressor inactivation in cancer presents a complex challenge in clinical oncology. It impacts prognosis, treatment resistance, and patient outcomes. However, on-going research is unravelling the intricacies of tumor suppressor function, providing insights into potential therapeutic vulnerabilities and innovative treatment strategies. From synthetic lethality and immunotherapy to gene therapy and the principles of precision medicine, a wide array of therapeutic options is emerging to combat tumor suppressordeficient tumors. As we advance in our understanding of the clinical implications of tumor suppressor inactivation, we are poised to usher in a new era of cancer therapy that is more precise, effective, and patient-centric. The future of cancer treatment is filled with promise, where tailored therapies based on tumor suppressor status may redefine the landscape of oncology and bring renewed hope to patients facing this formidable adversary.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Huang, Franklin W., Eran Hodis, Mary Jue Xu and Gregory V. Kryukov et al. "Highly recurrent TERT promoter mutations in human melanoma." Sci 339 (2013): 957-959.

    Google Scholar, Crossref, Indexed at

  2. Esteller, Manel. "CpG island hypermethylation and tumor suppressor genes: A booming present, a brighter future." Oncogene 21 (2002): 5427-5440.

    Indexed at

  3. Horn, Susanne, Adina Figl, P. Sivaramakrishna Rachakonda and Christine Fischer, et al. "TERT promoter mutations in familial and sporadic melanoma." Sci 339 (2013): 959-961.

    Google Scholar, Crossref, Indexed at

  4. Sawada, Maria IBAC, Guilherme da S Ferreira and Marisa Passarelli. "Cholesterol derivatives and breast cancer: Oxysterols driving tumor growth and metastasis." Biomark Med 14 (2020): 1299-1302.

    Indexed at

  5. Huang, Dong-Sheng, Zhaohui Wang, Xu-Jun He and Bill H. Diplas, et al. "Recurrent TERT promoter mutations identified in a large-scale study of multiple tumour types are associated with increased TERT expression and telomerase activation." Eur J Cancer 51 (2015): 969-976.

    Google Scholar, Crossref, Indexed at

  6. Ahn, Hwa Young, Min Suk Kim, Min Joo Kim and Soo Youn Cho, et al. "Loss of ER β expression in papillary thyroid carcinoma is associated with recurrence in young female." Clin Endocrinol 82 (2015): 300-306.

    Google Scholar, Crossref, Indexed at

arrow_upward arrow_upward