GET THE APP

Targeting Epigenetic Modifications in Cancer Therapy Challenges and Opportunities
..

Journal of Biomedical and Pharmaceutical Sciences

ISSN: 2952-8100

Open Access

Mini Review - (2024) Volume 7, Issue 2

Targeting Epigenetic Modifications in Cancer Therapy Challenges and Opportunities

Mara Sinclair*
*Correspondence: Mara Sinclair, Department of Oncology, University of the Philippines Manila, Manila, Philippines, Email:
Department of Oncology, University of the Philippines Manila, Manila, Philippines

Received: 04-Mar-2024, Manuscript No. jbps-24-134836; Editor assigned: 05-Jan-2024, Pre QC No. P-134836; Reviewed: 18-Mar-2024, QC No. Q-134836; Revised: 23-Mar-2024, Manuscript No. R-134836; Published: 30-Mar-2024 , DOI: 10.37421/2952-8100.2024.07.456
Citation: Sinclair, Mara. “Targeting Epigenetic Modifications in Cancer Therapy Challenges and Opportunities.” J Biomed Pharm Sci 7 (2024): 456.
Copyright: © 2024 Sinclair M. 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

Epigenetic modifications play a critical role in the development and progression of cancer by regulating gene expression patterns, chromatin structure, and cellular phenotypes. Targeting epigenetic abnormalities has emerged as a promising therapeutic strategy for cancer treatment, offering opportunities to reverse aberrant gene silencing, restore tumor suppressor function, and inhibit oncogenic signaling pathways. In this review, we discuss the challenges and opportunities of targeting epigenetic modifications in cancer therapy. We explore the mechanisms of epigenetic dysregulation in cancer, the therapeutic agents targeting epigenetic enzymes and readers, the clinical applications of epigenetic therapies, and the challenges associated with drug resistance, toxicity, and patient selection. Despite challenges, epigenetic therapies hold promise for improving cancer treatment outcomes and overcoming therapeutic resistance through personalized and precision medicine approaches.

Keywords

Epigenetics • Cancer therapy • Epigenetic modifications • Histone modifications • DNA methylation • Chromatin remodelling • Targeted therapy • Precision medicine

Introduction

Cancer is a complex and heterogeneous disease characterized by uncontrolled cell growth, invasion, and metastasis, driven by genetic and epigenetic alterations. Epigenetic modifications, including DNA methylation, histone modifications, chromatin remodeling, and non-coding RNA regulation, play a crucial role in regulating gene expression patterns, chromatin structure, and cellular phenotypes. Aberrant epigenetic changes contribute to the initiation, progression, and metastasis of cancer by silencing tumor suppressor genes, activating oncogenes, and promoting genomic instability [1]. Targeting epigenetic abnormalities has emerged as a promising therapeutic strategy for cancer treatment, offering opportunities to reverse epigenetic dysregulation, restore normal gene expression patterns, and inhibit oncogenic signaling pathways.

Epigenetic modifications in cancer involve alterations in DNA methylation patterns, histone post-translational modifications, and chromatin structure, which contribute to the dysregulation of gene expression and cellular functions. DNA methylation, the addition of methyl groups to cytosine residues within CpG dinucleotides, is a key epigenetic mechanism involved in gene silencing and transcriptional repression. Hypermethylation of CpG islands within gene promoter regions can lead to silencing of tumor suppressor genes, such as CDKN2A, MLH1, and BRCA1, while hypomethylation of gene body regions can activate oncogenes, such as MYC and RAS, promoting tumor growth and metastasis.

Literature Review

Histone modifications, including acetylation, methylation, phosphorylation, and ubiquitination, regulate chromatin structure and gene expression by altering histone-DNA interactions and recruiting chromatin-modifying enzymes and transcriptional regulators [2]. Histone acetylation, catalyzed by histone acetyltransferases (HATs) and reversed by histone deacetylases (HDACs), is associated with transcriptional activation and open chromatin conformation, while histone methylation, catalyzed by histone methyltransferases (HMTs) and demethylases (HDMs), can have both activating and repressive effects on gene expression depending on the specific histone residues and methylation states.

Chromatin remodeling complexes, such as SWI/SNF, ISWI, and Polycomb repressive complex (PRC), regulate chromatin accessibility and nucleosome positioning by sliding, ejecting, or restructuring nucleosomes in response to signaling cues and transcriptional programs. Dysregulation of chromatin remodeling complexes in cancer can lead to aberrant gene expression patterns, altered DNA repair mechanisms, and resistance to chemotherapy and targeted therapies.

Discussion

Targeting epigenetic modifications in cancer therapy involves the development of small-molecule inhibitors, monoclonal antibodies, and nucleic acid-based therapeutics that selectively modulate epigenetic enzymes, readers, and chromatin regulators. Histone deacetylase inhibitors (HDACIs), such as vorinostat, romidepsin, and panobinostat, have been approved for the treatment of hematological malignancies, such as cutaneous T-cell lymphoma and multiple myeloma, by restoring acetylation levels, reactivating tumor suppressor genes, and inducing cell cycle arrest and apoptosis.

DNA methyltransferase inhibitors (DNMTIs), such as azacitidine and decitabine, have been approved for the treatment of myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) by inhibiting DNA methylation, reactivating silenced genes, and promoting cellular differentiation and apoptosis [3]. However, DNMTIs have limited efficacy in solid tumors and are associated with toxicity and drug resistance mechanisms, highlighting the need for alternative strategies and combination therapies to enhance their clinical utility.

Histone methyltransferase inhibitors (HMTIs), histone demethylase inhibitors (HDMIs), and histone acetyltransferase inhibitors (HATIs) are emerging as novel therapeutic agents for targeting histone modifications in cancer therapy. These inhibitors selectively target specific epigenetic enzymes and disrupt aberrant chromatin modifications, leading to reactivation of tumor suppressor genes, inhibition of oncogenic signaling pathways, and suppression of tumor growth and metastasis.

Non-coding RNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), play critical roles in epigenetic regulation of gene expression and cancer pathogenesis. Dysregulation of non-coding RNAs in cancer can lead to aberrant epigenetic modifications, altered chromatin structure, and perturbed signaling pathways [4]. Therapeutic targeting of non-coding RNAs using antisense oligonucleotides, small interfering RNAs (siRNAs), and RNA-based therapeutics offers opportunities for modulating gene expression patterns, restoring cellular homeostasis, and inhibiting tumor progression.

Despite the promise of epigenetic therapies in cancer treatment, several challenges remain in clinical translation, patient selection, and therapeutic resistance. Epigenetic therapies can be associated with off-target effects, toxicity, and adverse events, such as myelosuppression, gastrointestinal toxicity, and cardiac toxicity, which can limit their clinical utility and patient compliance [5,6]. Additionally, acquired resistance mechanisms, such as mutations in epigenetic enzymes, activation of compensatory pathways, and tumor heterogeneity, can reduce the efficacy of epigenetic therapies and lead to treatment failure. Furthermore, patient selection criteria, biomarker identification, and predictive modeling are needed to optimize treatment strategies, personalize therapy, and improve patient outcomes in cancer patients.

Conclusion

In conclusion, targeting epigenetic modifications in cancer therapy offers promising opportunities for reversing aberrant gene expression patterns, restoring tumor suppressor function, and inhibiting oncogenic signaling pathways. Epigenetic therapies, including small-molecule inhibitors, monoclonal antibodies, and nucleic acid-based therapeutics, enable precise modulation of epigenetic enzymes, readers, and chromatin regulators, leading to reactivation of silenced genes, inhibition of oncogenic pathways, and suppression of tumor growth and metastasis. Despite challenges in clinical translation, drug resistance, and patient selection, epigenetic therapies hold promise for improving cancer treatment outcomes and overcoming therapeutic resistance through personalized and precision medicine approaches. Ongoing research efforts and collaborative initiatives are needed to further elucidate the mechanisms of epigenetic dysregulation in cancer, identify novel therapeutic targets, and develop innovative strategies for epigenetic-based cancer therapy.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Ahuja, Nita, Anup R. Sharma, and Stephen B. Baylin. "Epigenetic therapeutics: a new weapon in the war against cancer."Annu Rev Med 67 (2016): 73-89.

    Google Scholar, Crossref, Indexed at

  2. Li, Ning, Lu Yang, Xue-Kang Qi, and Yu-Xin Lin, et al. "BET bromodomain inhibitor JQ1 preferentially suppresses EBV-positive nasopharyngeal carcinoma cells partially through repressing c-Myc."Cell Death Dis 9 (2018): 761.

    Google Scholar, Crossref, Indexed at

  3. Chen, Hong Ping, Yu Tina Zhao, and Ting C. Zhao. "Histone deacetylases and mechanisms of regulation of gene expression."Crit Rev Oncog 20 (2015).

    Google Scholar, Crossref, Indexed at

  4. Poulin, Myles B., Jessica L. Schneck, Rosalie E. Matico, and Wangfang Hou, et al. "Nucleosome binding alters the substrate bonding environment of histone H3 lysine 36 methyltransferase NSD2."J Am Chem Soc 138 (2016): 6699-6702.

    Google Scholar, Crossref, Indexed at

  5. Milite, Ciro, Alessandra Feoli, John R. Horton, and Donatella Rescigno, o et al. "Discovery of a novel chemotype of histone lysine methyltransferase EHMT1/2 (GLP/G9a) inhibitors: rational design, synthesis, biological evaluation, and co-crystal structure."J Med Chem 62 (2019): 2666-2689.

    Google Scholar, Crossref, Indexed at

  6. Nikfarjam, Azadeh, Abeed Sarker, Karen O’connor and Rachel Ginn, et al. "Pharmacovigilance from social media: mining adverse drug reaction mentions using sequence labeling with word embedding cluster features."J Am Med Inform Assoc 22 (2015): 671-681.

    Google Scholar, Crossref, Indexed at

arrow_upward arrow_upward