GET THE APP

RNA-based Approaches to Cancer Treatment: In Silico Development and Assessment of ASOs for Targeted Exon Skipping
..

Molecular and Genetic Medicine

ISSN: 1747-0862

Open Access

Mini Review - (2023) Volume 17, Issue 6

RNA-based Approaches to Cancer Treatment: In Silico Development and Assessment of ASOs for Targeted Exon Skipping

Logan Thorne*
*Correspondence: Logan Thorne, Department of Medical Genetics and Genomics, University of Warwick, Coventry, UK, Email:
Department of Medical Genetics and Genomics, University of Warwick, Coventry, UK

Received: 03-Nov-2023, Manuscript No. jmgm-24-125760; Editor assigned: 06-Nov-2023, Pre QC No. P-125760; Reviewed: 17-Nov-2023, QC No. Q-125760; Revised: 22-Nov-2023, Manuscript No. R-125760; Published: 29-Nov-2023 , DOI: 10.37421/1747-0862.2023.17.638
Citation: Thorne, Logan. “RNA-based Approaches to Cancer Treatment: In Silico Development and Assessment of ASOs for Targeted Exon Skipping.” J Mol Genet Med 17 (2023): 638.
Copyright: © 2023 Thorne L. 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

RNA-based approaches have emerged as promising strategies for cancer treatment, providing innovative solutions to target specific genetic abnormalities associated with tumorigenesis. Among these approaches, the development and assessment of Antisense Oligonucleotides (ASOs) for targeted exon skipping have gained significant attention. In this article, we delve into the intricacies of RNA-based cancer treatment, focusing on the in silico methods employed in the design and evaluation of ASOs for exon skipping. We explore the molecular underpinnings of cancer, the role of RNA in disease progression, and the potential of ASOs to modulate gene expression with precision. Through an in-depth analysis of computational tools and techniques, we evaluate the effectiveness of ASOs in silico, paving the way for enhanced therapeutic outcomes in the realm of cancer treatment.

Keywords

RNA-based therapies • Cancer treatment • Antisense Oligonucleotides (ASOs) • Precision medicine

Introduction

Cancer remains a formidable challenge in the field of medicine, with its complex and heterogeneous nature necessitating innovative therapeutic approaches. Traditional cancer treatments, such as chemotherapy and radiation, often exhibit limited specificity and can cause collateral damage to healthy tissues. The advent of RNA-based therapies has opened new avenues for targeted and personalized cancer treatment strategies. Among these approaches, the use of Antisense Oligonucleotides (ASOs) for targeted exon skipping has gained prominence for its potential to address specific genetic aberrations associated with cancer. This article provides a comprehensive overview of RNA-based approaches to cancer treatment, with a specific focus on the in silico development and assessment of ASOs for targeted exon skipping. By leveraging computational tools and techniques, researchers are able to design ASOs with enhanced precision, offering a glimpse into the future of personalized medicine in oncology [1].

Literature Review

Understanding the molecular basis of cancer is crucial for developing targeted therapies. Cancer arises from the accumulation of genetic mutations that disrupt the normal regulatory mechanisms controlling cell growth and proliferation. Aberrant activation of oncogenes and inactivation of tumor suppressor genes are common events in cancer development. RNA, as an intermediary between DNA and protein, plays a central role in the expression of these genes, making it a prime target for therapeutic intervention. RNA-based therapies encompass a diverse range of approaches aimed at modulating gene expression for therapeutic purposes. These strategies leverage the versatility of RNA molecules, including messenger RNA (mRNA), microRNA (miRNA), small interfering RNA (siRNA), and Antisense Oligonucleotides (ASOs). Each of these RNA species presents unique opportunities for targeted intervention, with ASOs standing out for their ability to induce targeted exon skipping. ASOs are short, synthetic oligonucleotides designed to bind to specific RNA sequences through Watson-Crick base pairing. By targeting pre-mRNA, ASOs can modulate splicing patterns, leading to the exclusion of specific exons during mRNA maturation. In the context of cancer, ASOs offer a precise means to correct aberrant splicing events associated with oncogenic mutations [2].

Discussion

The in silico development of ASOs involves the use of computational tools and algorithms to design oligonucleotides with optimal binding affinity and specificity. Various parameters, such as sequence complementarity, secondary structure, and off-target effects, are considered during the design phase. Additionally, advancements in bioinformatics have enabled the prediction of ASO efficacy and safety, streamlining the experimental validation process. Several computational tools have been developed to aid in the design and assessment of ASOs for targeted exon skipping. Tools such as ExonSkipFinder and SpliceAid-F provide insights into splice site recognition, facilitating the identification of target exons for skipping. RNA secondary structure prediction tools, including Mfold and RNAfold, assist in evaluating potential off-target effects and optimizing ASO binding [3].

In silico evaluation of ASOs is a critical step in determining their potential efficacy and safety before experimental validation. Computational methods can predict the binding affinity of ASOs to target RNA sequences and assess the likelihood of off-target effects. Additionally, the prediction of RNA secondary structures aids in understanding the stability of ASO-RNA interactions, providing valuable information for optimizing ASO design. While in silico approaches offer significant advantages in the development of ASOs, several challenges and considerations must be addressed. Off-target effects, immune system activation, and delivery methods are among the key challenges that researchers face in translating in silico-designed ASOs into effective cancer therapies. Overcoming these hurdles requires interdisciplinary collaboration and a holistic approach to therapeutic development. The translation of in silico-designed ASOs from the laboratory to clinical settings holds immense promise for the future of cancer treatment. Clinical trials exploring the safety and efficacy of ASOs in various cancer types are underway, with preliminary results demonstrating encouraging outcomes [4].

As technology continues to advance, the integration of artificial intelligence and machine learning into ASO design may further enhance the precision and efficiency of RNA-based cancer therapies. The development and deployment of RNA-based therapies, including ASOs, raise ethical and regulatory considerations. Issues such as patient consent, data privacy, and equitable access to emerging therapies require careful consideration. Regulatory bodies play a pivotal role in ensuring the safety and efficacy of novel treatments, necessitating a balance between innovation and patient protection. The landscape of RNA-based cancer therapies is continuously evolving, with ongoing research exploring new avenues and emerging technologies. One such area of interest is the integration of CRISPR-based technologies with ASOs, allowing for precise and targeted genome editing. The combination of CRISPR-Cas systems with ASOs offers the potential not only to skip specific exons but also to correct genetic mutations at the DNA level. This convergence of technologies holds promise for addressing a broader spectrum of genetic abnormalities underlying cancer [5].

Additionally, the advent of single-cell RNA sequencing (scRNA-seq) has provided unprecedented insights into the heterogeneity of cancer at the cellular level. Integrating scRNA-seq data into in silico ASO design could enhance the specificity of therapeutic interventions by accounting for intra-tumoral diversity. This personalized approach may be particularly valuable in addressing challenges posed by tumor heterogeneity, enabling the development of tailored RNA-based therapies for individual patients. Advancements in RNA-based cancer treatment benefit greatly from global collaborations and knowledge sharing. International consortia and research networks focused on RNA therapeutics facilitate the exchange of data, methodologies, and best practices. Collaborative efforts foster a synergistic approach to overcoming challenges and accelerating the translation of innovative therapies from bench to bedside. Initiatives such as the Global Alliance for Genomics and Health (GA4GH) play a pivotal role in promoting data interoperability and ethical standards for global genomic research [6].

Conclusion

RNA-based approaches, particularly the development of ASOs for targeted exon skipping, represent a paradigm shift in cancer treatment. In silico methods have accelerated the design and evaluation of ASOs, offering a rational and systematic approach to therapeutic development. As the field continues to evolve, the integration of computational tools, interdisciplinary collaboration, and robust regulatory frameworks will be essential for realizing the full potential of RNA-based therapies in the fight against cancer. As RNA-based therapies progress through preclinical and clinical development, regulatory agencies play a pivotal role in establishing guidelines and frameworks for their evaluation and approval. Regulatory bodies must adapt to the rapidly evolving landscape of RNA therapeutics, ensuring a balance between facilitating innovation and safeguarding patient safety. International collaboration among regulatory agencies is essential for harmonizing standards and expediting the regulatory review process for novel RNA-based cancer therapies. RNA-based approaches, particularly the in silico development and assessment of ASOs for targeted exon skipping, represent a groundbreaking frontier in cancer treatment. The convergence of computational biology, molecular biology, and clinical research has paved the way for precision medicine tailored to the genetic intricacies of individual tumors. As we navigate the challenges and opportunities in this field, ongoing research, global collaborations, and regulatory frameworks will be pivotal in realizing the full potential of RNAbased therapies for the benefit of cancer patients worldwide.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Tate, John G., Sally Bamford, Harry C. Jubb and Zbyslaw Sondka, s et al. "COSMIC: The catalogue of somatic mutations in cancer." Nucleic Acid Res 47 (2019): D941-D947.
  2. Google Scholar, Crossref, Indexed at

  3. Priestley, Peter, Jonathan Baber, Martijn P. Lolkema and Neeltje Steeghs, et al. "Pan-cancer whole-genome analyses of metastatic solid tumours." Nat 575 (2019): 210-216.
  4. Google Scholar, Crossref, Indexed at

  5. Scotti, Marina M. and Maurice S. Swanson. "RNA mis-splicing in disease." Nat Rev Genet 17 (2016): 19-32.
  6. Google Scholar, Crossref, Indexed at

  7. Carter, John C., Daniel W. Sheehan, Andre Prochoroff and David J. Birnkrant. "Muscular dystrophies." Clin Chest Med 39 (2018): 377-389.
  8. Google Scholar, Crossref, Indexed at

  9. Scharnhorst, Volkher, Alex J. van der Eb and Aart G. Jochemsen. "WT1 proteins: Functions in growth and differentiation." Gene 273 (2001): 141-161.
  10. Google Scholar, Crossref, Indexed at

  11. Mann, Christopher J., Kaite Honeyman, Graham McClorey and Sue Fletcher, et al. "Improved antisense oligonucleotide induced exon skipping in the mdx mouse model of muscular dystrophy." J Gene Med: A cross‐disciplinary journal for research on the science of gene transfer and its clinical applications 4 (2002): 644-654.
  12. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 3919

Molecular and Genetic Medicine received 3919 citations as per Google Scholar report

Molecular and Genetic Medicine peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward