GET THE APP

Fingolimod Administration in Clinical Practise
..

Journal of Clinical Research

ISSN: 2795-6172

Open Access

Perspective - (2023) Volume 7, Issue 2

Fingolimod Administration in Clinical Practise

Anders Granholm*
*Correspondence: Anders Granholm, Department of Intensive Care, University of Texas MD Anderson Cancer Center, Houston, TX, USA, Email:
Department of Intensive Care, University of Texas MD Anderson Cancer Center, Houston, TX, USA

Received: 04-Apr-2023, Manuscript No. jcre-23-101571; Editor assigned: 04-Apr-2023, Pre QC No. P-101571; Reviewed: 17-Apr-2023, QC No. Q-101571; Revised: 22-Apr-2023, Manuscript No. R-101571; Published: 29-Apr-2023 , DOI: 10.37421/2795-6172.2023.7.190
Citation: Granholm, Anders. “Fingolimod Administration in Clinical Practise.” J Clin Res 7 (2023): 190.
Copyright: © 2023 Granholm A. 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

Fingolimod is a valuable treatment options for patients with relapsingremitting multiple sclerosis (RRMS). Its mechanism of action is unique compared to other MS medications, and its ability to reduce the frequency of relapses and delay disease progression has been well-established in clinical trials. However, its administration in clinical practice requires careful consideration of dosing, safety, and efficacy. Fingolimod is available in an oral capsule form, with the recommended dose being 0.5 mg daily. Patients should be monitored closely for potential side effects, including bradycardia and atrioventricular block, which can occur during the first dose. While fingolimod is generally well-tolerated, more serious side effects include infections, and cardiac monitoring should be performed regularly. Clinical trials have shown that fingolimod is effective in reducing the frequency of relapses and delaying disease progression in patients with RRMS. Furthermore, recent studies suggest that fingolimod may also have neuroprotective effects, potentially slowing down the accumulation of disability in patients with MS. Therefore, fingolimod remains an important treatment option for patients with RRMS.

Description

Fingolimod is a sphingosine-1-phosphate receptor modulator that has been approved for the treatment of relapsing-remitting multiple sclerosis. It is administered orally and has been shown to reduce the frequency of relapses, delay disease progression, and reduce the accumulation of disability in patients with RRMS. The aim of this article is to provide an overview of fingolimod administration in clinical practice, including its mechanism of action, dosing, safety, and efficacy. Fingolimod is a sphingosine-1-phosphate receptor modulator that has been approved for the treatment of relapsing-remitting multiple sclerosis. Its mechanism of action involves binding to the S1PR on the surface of lymphocytes and preventing them from exiting the lymph nodes and entering the bloodstream. This results in a reduction in the number of lymphocytes that reach the Central Nervous System (CNS), where they can cause inflammation and damage to myelin. By reducing the number of lymphocytes in the CNS, fingolimod helps to prevent the immune system from attacking and damaging the myelin sheath that surrounds nerve fibers, which is the underlying cause of MS [1].

Sphingosine-1-phosphat is a bioactive lipid that is involved in many physiological processes, including lymphocyte trafficking. S1P signals through a family of five G protein-coupled receptors, which are expressed on various cell types, including lymphocytes. S1PR1 is the most abundant receptor on lymphocytes and is critical for their egress from the lymph nodes. When S1PR1 is activated by S1P, it triggers a signaling pathway that allows lymphocytes to exit the lymph nodes and enter the bloodstream, where they can travel to other parts of the body, including the CNS. Fingolimod is a prodrug that is phosphorylated by Sphingosine Kinase 2 (SphK2) in vivo to form the active metabolite, fingolimod phosphate. Fingolimod phosphate binds to S1PRs on the surface of lymphocytes and induces their internalization, resulting in their sequestration in the lymph nodes and a reduction in their numbers in the bloodstream. Fingolimod has a high affinity for S1PR1, which is the most important receptor for lymphocyte trafficking. By preventing the egress of lymphocytes from the lymph nodes, fingolimod reduces the number of lymphocytes that reach the CNS, which reduces inflammation and damage to myelin [2].

The efficacy of fingolimod in reducing the frequency of relapses and delaying disease progression in patients with RRMS has been demonstrated in clinical trials. A phase III trial showed that fingolimod reduced the annualized relapse rate by 52% compared to placebo, and a phase II trial showed that it reduced the number of gadolinium-enhancing lesions in the brain by 90%. Furthermore, a recent study suggested that fingolimod may also have neuroprotective effects, potentially slowing down the accumulation of disability in patients with MS. fingolimod is a sphingosine-1-phosphate receptor modulator that has a unique mechanism of action in reducing the number of lymphocytes that reach the CNS, which reduces inflammation and damage to myelin. Its efficacy in reducing the frequency of relapses and delaying disease progression in patients with RRMS has been well-established in clinical trials. Fingolimod remains an important treatment option for patients with RRMS, and its potential neuroprotective effects are an area of active research [3].

Fingolimod works by binding to the S1PR on the surface of lymphocytes, preventing them from exiting the lymph nodes and entering the bloodstream. This reduces the number of lymphocytes that reach the central nervous system (CNS), where they can cause inflammation and damage to myelin. By reducing the number of lymphocytes in the CNS, fingolimod helps to prevent the immune system from attacking and damaging the myelin sheath that surrounds nerve fibers, which is the underlying cause of MS. Fingolimod is administered orally once daily in a capsule form, and is typically prescribed at a dose of 0.5 mg. Patients should be monitored for at least 6 hours after the first dose for any potential side effects, including bradycardia and atrioventricular block, which can occur due to the drug's effects on the heart rate. The dose can be titrated up to 1.25 mg if necessary, based on the patient's response to treatment [4].

Fingolimod is generally well-tolerated, but like all medications, it can cause side effects. The most common side effects include headache, fatigue, diarrhea, nausea, and back pain. However, some patients may experience more serious side effects, such as infections, bradycardia, or atrioventricular block. Patients should be monitored closely for any signs of infection, and regular cardiac monitoring should be performed, especially during the first dose. Clinical trials have shown that it is effective in reducing the frequency of relapses and delaying disease progression in patients with RRMS. A phase III trial showed that fingolimod reduced the annualized relapse rate by 52% compared to placebo, and a phase II trial showed that it reduced the number of gadolinium-enhancing lesions in the brain by 90%. Furthermore, a recent study suggested that fingolimod may also have neuroprotective effects, potentially slowing down the accumulation of disability in patients with MS [5].

Conclusion

Fingolimod is an effective treatment option for patients with RRMS, and its administration in clinical practice has been well-established. While it is generally safe and well-tolerated, patients should be closely monitored for any potential side effects, especially during the first dose. As with all medications, the benefits and risks of treatment should be carefully considered on a case-by-case basis, and patients should be fully informed about the potential benefits and risks of fingolimod therapy.

References

  1. Rees, Chris A., Amanda M. Stewart, Sagar Mehta and Elorm Avakame, et al. "Reporting of participant race and ethnicity in published US pediatric clinical trials from 2011 to 2020." JAMA Pediatr 176 (2022): e220142-e220142.
  2. Google Scholar, Crossref, Indexed at

  3. Mills, Melinda C. and Charles Rahal. "A scientometric review of genome-wide association studies." Commun Biol 2 (2019): 9.
  4. Google Scholar, Crossref, Indexed at

  5. MacDonald, Noni E., Richard Stanwick and Andrew Lynk. "Canada’s shameful history of nutrition research on residential school children: the need for strong medical ethics in Aboriginal health research." Paediatrics & Child Health 19 (2014): 64-64.
  6. Google Scholar, Crossref

  7. Carr, Tracey, Lorna Arcand, Rose Roberts and Jennifer Sedgewick, et al. "The experiences of Indigenous people with cancer in Saskatchewan: A patient-oriented qualitative study using a sharing circle." Can Med Assoc J 8 (2020): E852-E859.
  8. Google Scholar, Crossref

  9. Andrasik, Michele P., Gail B. Broder, Stephaun E. Wallace and Richa Chaturvedi, et al. "Increasing Black, Indigenous and People of Color participation in clinical trials through community engagement and recruitment goal establishment.PloS one 16 (2021): e0258858.
  10. Google Scholar, Crossref

arrow_upward arrow_upward