GET THE APP

CRISPR/Cas9-Mediated Genome Editing in Cancer Therapy
..

Journal of General Practice

ISSN: 2329-9126

Open Access

Opinion - (2024) Volume 12, Issue 1

CRISPR/Cas9-Mediated Genome Editing in Cancer Therapy

Lerrtgrt Vedrdfer*
*Correspondence: Lerrtgrt Vedrdfer, Department of Medicine, University of California Riverside, Riverside, CA 92521, USA, Email:
Department of Medicine, University of California Riverside, Riverside, CA 92521, USA

Received: 20-Jan-2024, Manuscript No. JGPR-24-129376; Editor assigned: 22-Jan-2024, Pre QC No. P-129376; Reviewed: 06-Feb-2024, QC No. Q-129376; Revised: 12-Feb-2024, Manuscript No. R-129376; Published: 29-Feb-2024 , DOI: 10.37421/2329-9126.2024.12.542
Citation: Vedrdfer, Lerrtgrt. “CRISPR/Cas9-Mediated Genome Editing in Cancer Therapy.” J Gen Pract 12 (2024): 542.
Copyright: © 2024 Vedrdfer 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.

Introduction

Cancer remains one of the most challenging diseases to treat, with its intricate genetic landscape and propensity for mutation. Conventional therapies such as chemotherapy and radiation often come with significant side effects and limited efficacy. However, the emergence of CRISPR/Cas9- mediated genome editing has sparked a new wave of hope in cancer therapy. This revolutionary technology offers precise manipulation of the genome, opening doors to targeted therapies, personalized medicine and potentially even cures. In this article, we delve into the promising applications of CRISPR/ Cas9 in cancer treatment, exploring its mechanisms, current challenges and future prospects [1].

Description

CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) and its associated protein, Cas9, comprise a powerful genome editing tool derived from bacterial immune systems. The system functions by utilizing guide RNA (gRNA) to direct Cas9 to specific DNA sequences, where it induces precise modifications, such as insertions, deletions, or substitutions. This capability allows researchers to edit genes with unprecedented accuracy, making it a versatile tool in various biological applications, including cancer therapy. CRISPR/Cas9 enables targeted disruption of oncogenes, which promote cancer progression and activation of tumor suppressor genes, which inhibit it. By precisely altering these genes, researchers can impede tumor growth and proliferation. For instance, inactivating the oncogene KRAS has shown promise in suppressing pancreatic and colorectal cancers, while restoring the function of tumor suppressors like TP53 can induce apoptosis in cancer cells [2].

Immunotherapy has emerged as a groundbreaking approach in cancer treatment by harnessing the body's immune system to target cancer cells. CRISPR/Cas9 can enhance the efficacy of immunotherapy by modifying immune cells, such as T cells, to recognize and attack cancer cells more effectively. This includes knocking out inhibitory receptors like PD-1 or CTLA- 4 to prevent immune evasion by tumors, as well as engineering Chimeric Antigen Receptor (CAR) T cells to target specific tumor antigens. CRISPR/ Cas9 facilitates the creation of precise cancer models by introducing mutations observed in patient tumors into laboratory cell lines or animal models. These models provide valuable insights into cancer biology, drug resistance mechanisms and the efficacy of potential therapeutics. Moreover, CRISPR screens enable high-throughput identification of genes essential for cancer survival, paving the way for the development of novel targeted therapies [3].

Cas9 may inadvertently edit genomic regions resembling the target sequence, leading to unintended mutations with unpredictable consequences. Efficient delivery of CRISPR components to target cells remains a hurdle, particularly in vivo. Various delivery vehicles, including viral vectors and nanoparticles, are under development to overcome this challenge. Host immune responses to CRISPR components can hinder their efficacy and pose safety concerns, necessitating strategies to evade or mitigate immune reactions. The ethical implications of genome editing, particularly in human germline cells, raise important ethical questions regarding safety, consent and equity. Regulatory frameworks must ensure responsible and equitable deployment of CRISPR technologies in cancer therapy. Despite the obstacles, ongoing research and technological advancements continue to propel CRISPR/Cas9 towards clinical application in cancer therapy [4].

Development of next-generation CRISPR systems with enhanced specificity, efficiency and versatility, such as base editing and prime editing, holds promise for safer and more precise genome editing. Continued optimization of delivery methods, including non-viral vectors and ex vivo approaches, will improve the efficiency and safety of CRISPR-mediated therapies. Transitioning from preclinical studies to clinical trials will be crucial for evaluating the safety and efficacy of CRISPR-based cancer therapies in human patients. Close collaboration between researchers, clinicians, regulators and ethicists is essential to navigate the complex landscape of clinical translation. Combining CRISPR/Cas9-mediated genome editing with existing modalities such as chemotherapy, radiation and immunotherapy may potentiate synergistic effects and overcome treatment resistance in cancer [5].

Conclusion

CRISPR/Cas9-mediated genome editing represents a paradigm shift in cancer therapy, offering unprecedented precision and versatility in targeting the underlying genetic alterations driving cancer progression. While challenges persist, ongoing research and innovation hold the potential to revolutionize cancer treatment, paving the way for personalized, targeted therapies with improved efficacy and reduced toxicity. As we continue to unravel the complexities of cancer biology and refine CRISPR technologies, the prospect of eradicating this devastating disease grows ever closer within reach needs of this vulnerable population and ultimately enhance their quality of life.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Finch Andrew J., Christine Hilcenko, Nicolas Basse and Lesley F. Drynan, et al. "Uncoupling of GTP hydrolysis from eIF6 release on the ribosome causes Shwachman-Diamond syndrome."Genes Dev 25 (2011): 917-929.

    Google Scholar, Crossref, Indexed at

  2. Almåsbak, Hilde, Tanja Aarvak and Mohan C. Vemuri. "CAR T cell therapy: A game changer in cancer treatment."J Immunol Res (2016): 12-25.

    Google Scholar, Crossref, Indexed at

  3. Palucka, A. Karolina and Lisa M. Coussens. "The basis of oncoimmunology."Cell164 (2016): 1233-1247.

    Google Scholar, Crossref, Indexed at

  4. Chu, Fuliang, Jingjing Cao and Sattva S. Neelalpu. "Versatile CAR T-cells for cancer immunotherapy."Wspolczesna Onkol 20182018): 73-80.

    Google Scholar, Crossref, Indexed at

  5. Gomez, Stephanie, Tomasz Tabernacki, Julie Kobyra and Paige Roberts, et al. "Combining epigenetic and immune therapy to overcome cancer resistance." Semin Cancer Biol 65 (2020): 99-113.

    Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 952

Journal of General Practice received 952 citations as per Google Scholar report

Journal of General Practice peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward