GET THE APP

Emerging Therapeutic Targets for the Treatment of Cystic Fibrosis
..

Journal of Lung Diseases & Treatment

ISSN: 2472-1018

Open Access

Opinion - (2023) Volume 9, Issue 5

Emerging Therapeutic Targets for the Treatment of Cystic Fibrosis

Li Jhang*
*Correspondence: Li Jhang, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA, Email:
Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA

Received: 01-Sep-2023, Manuscript No. LDT-23-119291; Editor assigned: 04-Sep-2023, Pre QC No. P-119291; Reviewed: 16-Sep-2023, QC No. Q-119291; Revised: 21-Sep-2023, Manuscript No. R-119291; Published: 28-Sep-2023 , DOI: 10.37421/2472-1018.2023.9.205
Citation: Jhang, Li. “Emerging Therapeutic Targets for the Treatment of Cystic Fibrosis.” J Lung Dis Treat 9 (2023): 205.
Copyright: © 2023 Jhang 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

Cystic Fibrosis (CF) is a genetic disorder that affects the respiratory, digestive and reproductive systems. It is caused by mutations in the CFTR (cystic fibrosis transmembrane conductance regulator) gene, which leads to the production of thick and sticky mucus in the lungs and other organs. Over the years, significant progress has been made in understanding the molecular and cellular mechanisms of CF, leading to the development of several therapies. However, there is still no cure for the disease and patients continue to suffer from its debilitating effects. In recent years, research has identified several promising emerging therapeutic targets for the treatment of cystic fibrosis. This article explores some of these innovative approaches and their potential to improve the lives of CF patients. The most well-known breakthrough in CF treatment came with the development of correctors and potentiators [1].

These drugs aim to address the underlying cause of CF, which is the malfunctioning CFTR protein. Mutations in the CFTR gene can result in a misfolded or dysfunctional CFTR protein, preventing it from reaching the cell surface and functioning properly. Correctors, such as ivacaftor, aim to help the protein fold correctly, while potentiators, like lumacaftor, enhance its activity once it reaches the cell surface. Ivacaftor was the first drug of its kind to be approved by the U.S. Food and Drug Administration (FDA) in 2012. It has shown significant benefits in patients with specific CFTR mutations, such as G551D. Lumacaftor-ivacaftor combination therapy was later approved for patients with the F508del mutation, the most common CFTR mutation. These therapies have been life-changing for many CF patients, but they are only effective for specific mutations. Emerging research is focused on expanding the number of mutations that can be targeted by these drugs, as well as improving their overall efficacy [2].

Description

One of the most promising areas of research for cystic fibrosis treatment is gene editing. Techniques like CRISPR-Cas9 have the potential to directly correct the CFTR gene's mutations. With gene editing, scientists can precisely modify the patient's own genetic material to produce a functional CFTR protein. While gene editing is still in its experimental stages for CF, it holds tremendous promise for future therapies. Challenges include ensuring the safety and specificity of gene-editing techniques, as well as their delivery to the target cells within the patient's body. RNA therapies are another emerging approach for treating cystic fibrosis. These therapies focus on modifying the RNA that is transcribed from the CFTR gene. By altering the RNA, it is possible to influence the expression and function of the CFTR protein. This approach can be especially valuable for patients with certain types of mutations that are difficult to address using traditional gene editing or CFTR modulators [3].

Small interfering RNA (siRNA) and Antisense Oligonucleotides (ASOs) are two types of RNA-based therapies that are being explored for CF. They work by either degrading the mutant RNA or altering its splicing to produce a functional CFTR protein. Clinical trials are underway to assess the safety and efficacy of these therapies in CF patients. CFTR is a complex protein that undergoes various stages of synthesis, folding and degradation within the cell. Proteostasis regulators aim to optimize these processes to ensure the proper functioning of the CFTR protein. This emerging therapeutic target seeks to enhance the production, folding and stability of CFTR, making it more effective in transporting chloride ions across the cell membrane. Several drugs, such as C18 and N6022, have shown promise in preclinical studies as proteostasis regulators. They target different aspects of CFTR processing and have the potential to benefit a broader range of CF patients by addressing the fundamental cellular defects associated with the disease [4].

In addition to addressing the core defect in CFTR function, it is equally important to manage the inflammatory response and infection-related complications in the lungs of CF patients. Chronic inflammation and recurrent lung infections are common in CF and can cause progressive lung damage. New anti-inflammatory agents are being developed to reduce the inflammation associated with the disease and potentially slow its progression. Anti-inflammatory drugs, such as lenabasum and azithromycin, have shown promise in reducing lung inflammation and improving lung function in CF patients. These drugs may complement existing CFTR modulators and antibiotics in the treatment of cystic fibrosis. Mucus buildup in the airways is a hallmark of CF, leading to chronic lung infections and reduced lung function. Mucolytic therapies aim to thin and loosen the mucus, making it easier for CF patients to clear it from their airways [5].

Conclusion

Emerging therapeutic targets for the treatment of cystic fibrosis offer a glimmer of hope for patients and their families. The progress made in recent years has been remarkable and ongoing research continues to push the boundaries of what is possible in CF treatment. With a focus on personalized medicine, innovative technologies and a comprehensive understanding of the underlying biology of the disease, the future holds promise for improved outcomes and, ultimately, a cure for cystic fibrosis. However, it is essential to address the challenges of accessibility, safety and efficacy to ensure that these promising therapies reach the individuals who need them the most. As the field of CF research continues to evolve, we are inching closer to a future where cystic fibrosis is no longer a life-threatening disease but a manageable condition, significantly improving the lives of those affected by it.

Acknowledgement

None.

Conflict of Interest

There are no conflicts of interest by author.

References

  1. Polgreen, Philip M. and Alejandro P. Comellas. "Clinical phenotypes of cystic fibrosis carriers." Annu Rev Med 73 (2022): 563-574.
  2. Google Scholar, Crossref, Indexed at

  3. Mai-Prochnow, Anne, Janice Gee Kay Hui, Staffan Kjelleberg and Jasna Rakonjac, et al. "Big things in small packages: The genetics of filamentous phage and effects on fitness of their host." FEMS Microbiol Rev 39 (2015): 465-487.
  4. Google Scholar, Crossref, Indexed at

  5. Fiedoruk, Krzysztof, Magdalena Zakrzewska, Tamara Daniluk and Ewelina Piktel, et al. "Two lineages of P. aeruginosa filamentous phages: Structural uniformity over integration preferences." Genome Biol Evol 12 (2020): 1765-1781.
  6. Google Scholar, Crossref, Indexed at

  7. Gavric, Damir and Petar Knezevic. "Filamentous pseudomonas phage Pf4 in the context of therapy-inducibility, infectivity, lysogenic conversion and potential application." Viruses 14 (2022): 1261.
  8. Google Scholar, Crossref, Indexed at

  9. Winstanley, Craig, Morgan GI Langille, Joanne L. Fothergill and Irena Kukavica-Ibrulj, et al. "Newly introduced genomic prophage islands are critical determinants of in vivo competitiveness in the liverpool epidemic strain of P. aeruginosa." Genome Res 19 (2059): 12-23.
  10. Google Scholar, Crossref, Indexed at

arrow_upward arrow_upward