Cargando…

Poly ε-Caprolactone Nanoparticles for Sustained Intra-Articular Immune Modulation in Adjuvant-Induced Arthritis Rodent Model

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disorder with synovitis and articular pathology as its primary expositions. Leflunomide (Lfd) is an anti-rheumatic drug that is effective in the treatment of RA, but displays severe side effects upon prolonged systemic administration. Lo...

Descripción completa

Detalles Bibliográficos
Autores principales: Singh, Ekta, Osmani, Riyaz Ali M., Banerjee, Rinti, Abu Lila, Amr Selim, Moin, Afrasim, Almansour, Khaled, Arab, Hany H., Alotaibi, Hadil Faris, Khafagy, El-Sayed
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8953799/
https://www.ncbi.nlm.nih.gov/pubmed/35335895
http://dx.doi.org/10.3390/pharmaceutics14030519
_version_ 1784675938741518336
author Singh, Ekta
Osmani, Riyaz Ali M.
Banerjee, Rinti
Abu Lila, Amr Selim
Moin, Afrasim
Almansour, Khaled
Arab, Hany H.
Alotaibi, Hadil Faris
Khafagy, El-Sayed
author_facet Singh, Ekta
Osmani, Riyaz Ali M.
Banerjee, Rinti
Abu Lila, Amr Selim
Moin, Afrasim
Almansour, Khaled
Arab, Hany H.
Alotaibi, Hadil Faris
Khafagy, El-Sayed
author_sort Singh, Ekta
collection PubMed
description Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disorder with synovitis and articular pathology as its primary expositions. Leflunomide (Lfd) is an anti-rheumatic drug that is effective in the treatment of RA, but displays severe side effects upon prolonged systemic administration. Local therapy might represent a promising strategy to treat rheumatoid arthritis without eliciting systemic adverse effects. In this study, leflunomide-loaded poly(ε-caprolactone) nanoparticles (Lfd-NPs) were prepared and assessed as a local drug delivery system capable of alleviating RA-associated inflammation. Lfd-NPs were optimized using the Quality by Design (QbD) approach, applying a 3(2) full factorial design. In vitro drug release from NPs was examined in simulated synovial fluid. In addition, the in vivo efficacy of Lfd-NPs was evaluated in the Adjuvant Induced Arthritis (AIA) rodent model. Sustained drug release in simulated synovial fluid was observed for up to 168 h. A gradual reduction in paw volume and knee diameter was observed over the course of treatment, indicating the regression of the disease. In addition, significant reductions in serum proinflammatory markers and cytokines, including the C-reactive protein (CRP), rheumatoid factor (RF), TNF-α, IL1-β, and IL-6, were verified upon treatment with Lfd-NPs, suggesting the modulation of immune responses at the pathological site. Most importantly, no remarkable signs of toxicity were observed in Lfd-NP-treated animals. Collectively, intra-articularly administered Lfd-NPs might represent a potential therapeutic alternative to systemically administered drugs for the treatment of rheumatoid arthritis, without eliciting systemic adverse effects.
format Online
Article
Text
id pubmed-8953799
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-89537992022-03-26 Poly ε-Caprolactone Nanoparticles for Sustained Intra-Articular Immune Modulation in Adjuvant-Induced Arthritis Rodent Model Singh, Ekta Osmani, Riyaz Ali M. Banerjee, Rinti Abu Lila, Amr Selim Moin, Afrasim Almansour, Khaled Arab, Hany H. Alotaibi, Hadil Faris Khafagy, El-Sayed Pharmaceutics Article Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disorder with synovitis and articular pathology as its primary expositions. Leflunomide (Lfd) is an anti-rheumatic drug that is effective in the treatment of RA, but displays severe side effects upon prolonged systemic administration. Local therapy might represent a promising strategy to treat rheumatoid arthritis without eliciting systemic adverse effects. In this study, leflunomide-loaded poly(ε-caprolactone) nanoparticles (Lfd-NPs) were prepared and assessed as a local drug delivery system capable of alleviating RA-associated inflammation. Lfd-NPs were optimized using the Quality by Design (QbD) approach, applying a 3(2) full factorial design. In vitro drug release from NPs was examined in simulated synovial fluid. In addition, the in vivo efficacy of Lfd-NPs was evaluated in the Adjuvant Induced Arthritis (AIA) rodent model. Sustained drug release in simulated synovial fluid was observed for up to 168 h. A gradual reduction in paw volume and knee diameter was observed over the course of treatment, indicating the regression of the disease. In addition, significant reductions in serum proinflammatory markers and cytokines, including the C-reactive protein (CRP), rheumatoid factor (RF), TNF-α, IL1-β, and IL-6, were verified upon treatment with Lfd-NPs, suggesting the modulation of immune responses at the pathological site. Most importantly, no remarkable signs of toxicity were observed in Lfd-NP-treated animals. Collectively, intra-articularly administered Lfd-NPs might represent a potential therapeutic alternative to systemically administered drugs for the treatment of rheumatoid arthritis, without eliciting systemic adverse effects. MDPI 2022-02-26 /pmc/articles/PMC8953799/ /pubmed/35335895 http://dx.doi.org/10.3390/pharmaceutics14030519 Text en © 2022 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Singh, Ekta
Osmani, Riyaz Ali M.
Banerjee, Rinti
Abu Lila, Amr Selim
Moin, Afrasim
Almansour, Khaled
Arab, Hany H.
Alotaibi, Hadil Faris
Khafagy, El-Sayed
Poly ε-Caprolactone Nanoparticles for Sustained Intra-Articular Immune Modulation in Adjuvant-Induced Arthritis Rodent Model
title Poly ε-Caprolactone Nanoparticles for Sustained Intra-Articular Immune Modulation in Adjuvant-Induced Arthritis Rodent Model
title_full Poly ε-Caprolactone Nanoparticles for Sustained Intra-Articular Immune Modulation in Adjuvant-Induced Arthritis Rodent Model
title_fullStr Poly ε-Caprolactone Nanoparticles for Sustained Intra-Articular Immune Modulation in Adjuvant-Induced Arthritis Rodent Model
title_full_unstemmed Poly ε-Caprolactone Nanoparticles for Sustained Intra-Articular Immune Modulation in Adjuvant-Induced Arthritis Rodent Model
title_short Poly ε-Caprolactone Nanoparticles for Sustained Intra-Articular Immune Modulation in Adjuvant-Induced Arthritis Rodent Model
title_sort poly ε-caprolactone nanoparticles for sustained intra-articular immune modulation in adjuvant-induced arthritis rodent model
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8953799/
https://www.ncbi.nlm.nih.gov/pubmed/35335895
http://dx.doi.org/10.3390/pharmaceutics14030519
work_keys_str_mv AT singhekta polyecaprolactonenanoparticlesforsustainedintraarticularimmunemodulationinadjuvantinducedarthritisrodentmodel
AT osmaniriyazalim polyecaprolactonenanoparticlesforsustainedintraarticularimmunemodulationinadjuvantinducedarthritisrodentmodel
AT banerjeerinti polyecaprolactonenanoparticlesforsustainedintraarticularimmunemodulationinadjuvantinducedarthritisrodentmodel
AT abulilaamrselim polyecaprolactonenanoparticlesforsustainedintraarticularimmunemodulationinadjuvantinducedarthritisrodentmodel
AT moinafrasim polyecaprolactonenanoparticlesforsustainedintraarticularimmunemodulationinadjuvantinducedarthritisrodentmodel
AT almansourkhaled polyecaprolactonenanoparticlesforsustainedintraarticularimmunemodulationinadjuvantinducedarthritisrodentmodel
AT arabhanyh polyecaprolactonenanoparticlesforsustainedintraarticularimmunemodulationinadjuvantinducedarthritisrodentmodel
AT alotaibihadilfaris polyecaprolactonenanoparticlesforsustainedintraarticularimmunemodulationinadjuvantinducedarthritisrodentmodel
AT khafagyelsayed polyecaprolactonenanoparticlesforsustainedintraarticularimmunemodulationinadjuvantinducedarthritisrodentmodel