1
|
Elkhashab M, Barreto G, Fauconnier M, Le Bourlout Y, Creemers LB, Nieminen HJ, Howard KA. ADAMTS5-specific gapmer release from an albumin biomolecular assembly and cartilage internalization triggered by ultrasound. Drug Deliv 2025; 32:2464921. [PMID: 39967271 PMCID: PMC11841101 DOI: 10.1080/10717544.2025.2464921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE Antisense oligonucleotides (ASOs) have reached the clinic; however, they lack tissue specificity. Albumin is a plasma-abundant macromolecule that has been shown to accumulate in inflamed tissues. In this work, we have designed a recombinant human albumin (rHA)-based biomolecular assembly incorporating a DNase-resistant phosphorothioate-based complementary oligonucleotide (cODN) and an anti-ADAMTS5 ASO for potential delivery to inflamed sites. Ultrasound (US) was used to trigger ASO release from the assembly and enhance internalization into articular cartilage. METHODS A phosphorothioate cODN was conjugated to rHA through a maleimide cross-linker after which, a therapeutic ADAMTS5-specific gapmer ASO was annealed to the cODN. ASO release was assessed after exposing the biomolecular assembly to different US conditions using an US-actuated medical needle operating at 32.2 kHz. Gene silencing efficiency of US-treated anti-ADAMTS5 ASO was assessed in human primary chondrocytes isolated from osteoarthritic patients. US-mediated ASO penetration into articular cartilage was assessed on ex vivo bovine articular cartilage. RESULTS ASO release was observed after exposure to US waves in continuous mode conditions that did not compromise ASO gene silencing efficiency in human chondrocytes. Furthermore, US increased ASO internalization into bovine articular cartilage after 30 min of application without detrimental effects on chondrocyte viability. CONCLUSION A medical needle driven by continuous US waves at 32.2 kHz has the capability of disassembling a duplex oligonucleotide and enhancing released ASOs internalization into articular cartilage. This work offers the potential delivery and the local triggered release of ASOs at the surface of articular cartilage providing potential benefits for the treatment of diverse cartilage pathologies.
Collapse
Affiliation(s)
- Marwa Elkhashab
- Interdisciplinary Nanoscience Center (iNANO) and Department of Molecular Biology, Aarhus University, Aarhus C, Denmark
| | - Goncalo Barreto
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Orton Orthopedic Hospital, Helsinki, Finland
| | - Maxime Fauconnier
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Yohann Le Bourlout
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Laura B. Creemers
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Heikki J. Nieminen
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Kenneth A. Howard
- Interdisciplinary Nanoscience Center (iNANO) and Department of Molecular Biology, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
2
|
Qiao Q, Wan X, Li J, Chen W, Li E, Qiu L, Tu H. Amphiphilic Celecoxib-Polysaccharide Delivery System for Enhanced Colon-Targeted Colitis Therapy. Pharmaceutics 2025; 17:511. [PMID: 40284506 PMCID: PMC12030041 DOI: 10.3390/pharmaceutics17040511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Ulcerative colitis (UC), a subtype of chronic inflammatory bowel disease (IBD), is primarily treated with oral medications to reduce inflammation and alleviate symptoms. Celecoxib (CXB) is an attractive candidate for UC; however, its limited solubility and low bioavailability pose significant challenges to its clinical application. Methods: We reported a novel chondroitin sulfate A-Celecoxib (CSA-CXB) polymeric nanoprodrug to address the limited solubility and low bioavailability of CXB. CXB was conjugated to chondroitin sulfate A (CSA) via succinic anhydride (SA) and ethylenediamine to prepare CSA-CXB polymers, which can self-assemble into nanoparticle structural prodrugs in aqueous condition. We investigated the stability, blood compatibility, and responsiveness of the nanoparticles. The ability of the nanoparticles to treat UC in vitro and in vivo was then evaluated. Results: The CSA-CXB nanoprodrug was spherical with a mean particle size of 188.4 ± 2.2 nm, a zeta potential of -22.9 ± 0.1 mV, and sustained drug release behavior. Furthermore, CSA-CXB exhibited remarkable antioxidant and anti-inflammatory effects, as it can significantly increase the free radical scavenging rate and reduce the expression level of ROS, TNF-α, IL-6, nitric oxide (NO), and COX-2 protein in vitro. In vivo results demonstrated that CSA-CXB targeted the mice's colon efficiently mitigate UC symptoms by inhibiting the expression of inflammatory cytokine. Conclusions: The CSA-CXB nanoprodrug can improve the therapeutic impact of CXB, and has potential as a new preparation for a clinical UC treatment nanoprodrug.
Collapse
Affiliation(s)
- Qiao Qiao
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi 214122, China;
| | - Xian Wan
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (X.W.); (J.L.); (W.C.)
| | - Jie Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (X.W.); (J.L.); (W.C.)
| | - Weijun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (X.W.); (J.L.); (W.C.)
| | - Enxuan Li
- The Second Clinical Medical School, Nanjing Medical University, Nanjing 210029, China
| | - Lipeng Qiu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (X.W.); (J.L.); (W.C.)
| | - Huiming Tu
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi 214122, China;
| |
Collapse
|
3
|
Toader C, Radoi MP, Covlea CA, Covache-Busuioc RA, Ilie MM, Glavan LA, Corlatescu AD, Costin HP, Gica MD, Dobrin N. Cerebral Aneurysm: Filling the Gap Between Pathophysiology and Nanocarriers. Int J Mol Sci 2024; 25:11874. [PMID: 39595942 PMCID: PMC11593836 DOI: 10.3390/ijms252211874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Intracranial aneurysms, characterized by abnormal dilations of cerebral arteries, pose significant health risks due to their potential to rupture, leading to subarachnoid hemorrhage with high mortality and morbidity rates. This paper aim is to explore the innovative application of nanoparticles in treating intracranial aneurysms, offering a promising avenue for enhancing current therapeutic strategies. We took into consideration the pathophysiology of cerebral aneurysms, focusing on the role of hemodynamic stress, endothelial dysfunction, and inflammation in their development and progression. By comparing cerebral aneurysms with other types, such as aortic aneurysms, we identify pathophysiological similarities and differences that could guide the adaptation of treatment approaches. The review highlights the potential of nanoparticles to improve drug delivery, targeting, and efficacy while minimizing side effects. We discuss various nanocarriers, including liposomes and polymeric nanoparticles, and their roles in overcoming biological barriers and enhancing therapeutic outcomes. Additionally, we discuss the potential of specific compounds, such as Edaravone and Tanshinone IIA, when used in conjunction with nanocarriers, to provide neuroprotective and anti-inflammatory benefits. By extrapolating insights from studies on aortic aneurysms, new research directions and therapeutic strategies for cerebral aneurysms are proposed. This interdisciplinary approach underscores the potential of nanoparticles to positively influence the management of intracranial aneurysms, paving the way for personalized treatment options that could significantly improve patient outcomes.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Mugurel Petrinel Radoi
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Christian-Adelin Covlea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Milena Monica Ilie
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Luca-Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Antonio-Daniel Corlatescu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Horia-Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Maria-Daria Gica
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | | |
Collapse
|
4
|
Singh H, Dan A, Kumawat MK, Pawar V, Chauhan DS, Kaushik A, Bhatia D, Srivastava R, Dhanka M. Pathophysiology to advanced intra-articular drug delivery strategies: Unravelling rheumatoid arthritis. Biomaterials 2023; 303:122390. [PMID: 37984246 DOI: 10.1016/j.biomaterials.2023.122390] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 10/29/2023] [Accepted: 11/04/2023] [Indexed: 11/22/2023]
Abstract
Rheumatoid arthritis (RA) is one of the most prevalent life-long autoimmune diseases with an unknown genesis. It primarily causes chronic inflammation, pain, and synovial joint-associated cartilage and bone degradation. Unfortunately, limited information is available regarding the etiology and pathogenesis of this chronic joint disorder. In the last few decades, an improved understanding of RA pathophysiology about key immune cells, antibodies, and cytokines has inspired the development of several anti-rheumatic drugs and biopharmaceuticals to act on RA-affected joints. However, life-long frequent systemic high doses of commercially available drugs are currently a limiting factor in the efficient management of RA. To address this issue, various single and double-barrier intra-articular drug delivery systems (IA-DDSs) such as nanocarriers, microparticles, hydrogels, and particles-hybrid hydrogel composite have been developed which can exclusively target the RA-affected joint cavity and release the precisely controlled therapeutic drug concentration for prolonged time whilst avoiding the systemic toxicity. This review provides a comprehensive overview of the pathogenesis of RA and discusses the rational design and development of biomaterials-based novel IA-DDs, ranging from conventional to advanced systems, for improved treatment of RA. Therefore, this review aims to unravel the pathophysiology of rheumatoid arthritis and explore cutting-edge IA-DD strategies exploiting biomaterials. It offers researchers a consolidated and up-to-date resource platform to analyze existing knowledge, identify research gaps, and contribute to the scientific literature.
Collapse
Affiliation(s)
- Hemant Singh
- Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, 382055, Gujarat, India; Department of Biology, Khalifa University, Main Campus, Abu Dhabi, 127788, United Arab Emirates
| | - Aniruddha Dan
- Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, 382055, Gujarat, India
| | - Mukesh Kumar Kumawat
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Vaishali Pawar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Deepak S Chauhan
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, H3C 3J7, Canada
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL- 33805, USA
| | - Dhiraj Bhatia
- Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, 382055, Gujarat, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Mukesh Dhanka
- Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, 382055, Gujarat, India.
| |
Collapse
|
5
|
Marques SS, Cant DJH, Minelli C, Segundo MA. Combining orthogonal measurements to unveil diclofenac encapsulation into polymeric and lipid nanocarriers. Anal Chim Acta 2023; 1262:341234. [PMID: 37179055 DOI: 10.1016/j.aca.2023.341234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/06/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023]
Abstract
The quantification of the drug associated to nanoparticle carriers, often expressed in terms of encapsulation efficiency, is a regulatory requirement. The establishment of independent methods to evaluate this parameter provides a means for measurement validation, which is critical in providing confidence in the methods and enabling the robust characterization of nanomedicines. Chromatography is traditionally used to measure drug encapsulation into nanoparticles. Here, we describe an additional independent strategy based on analytical centrifugation. The encapsulation of diclofenac into nanocarriers was quantified based on the mass difference between placebo (i.e. unloaded) and loaded nanoparticles. This difference was estimated using particle densities measured by differential centrifugal sedimentation (DCS) and size and concentration values measured by particle tracking analysis (PTA). The proposed strategy was applied to two types of formulations, namely poly(lactic-co-glycolic acid) (PLGA) nanoparticles and nanostructured lipid carriers, which were analysed by DCS operated in sedimentation and flotation modes, respectively. The results were compared to those from high performance liquid chromatography (HPLC) measurements. Additionally, X-ray photoelectron spectroscopy analysis was used to elucidate the surface chemical composition of the placebo and loaded nanoparticles. The proposed approach enables the monitoring of batch-to-batch consistency and the quantification of diclofenac association to PLGA nanoparticles from 0.7 ng to 5 ng of drug per 1 μg of PLGA, with good linear correlation between DCS and HPLC results (R2 = 0.975). Using the same approach, similar quantification in lipid nanocarriers was possible for a loading of diclofenac ≥1.1 ng per 1 μg of lipids, with results in agreement with the HPLC method (R2 = 0.971). Hence, the strategy proposed here expands the analytical tools available for evaluating nanoparticles encapsulation efficiency, being thus significant for increasing the robustness of drug-delivery nanocarriers characterization.
Collapse
Affiliation(s)
- Sara S Marques
- LAQV, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal; National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, United Kingdom
| | - David J H Cant
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, United Kingdom
| | - Caterina Minelli
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, United Kingdom.
| | - Marcela A Segundo
- LAQV, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
6
|
Markovic M, Ben-Shabat S, Nagendra Manda J, Abramov-Harpaz K, Regev C, Miller Y, Aponick A, Zimmermann EM, Dahan A. PLA 2-Triggered Activation of Cyclosporine-Phospholipid Prodrug as a Drug Targeting Approach in Inflammatory Bowel Disease Therapy. Pharmaceutics 2022; 14:pharmaceutics14030675. [PMID: 35336048 PMCID: PMC8950246 DOI: 10.3390/pharmaceutics14030675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Oral medication with activity specifically at the inflamed sites throughout the gastrointestinal tract and limited systemic exposure would be a major advance in our therapeutic approach to inflammatory bowel disease (IBD). For this purpose, we have designed a prodrug by linking active drug moiety to phospholipid (PL), the substrate of phospholipase A2 (PLA2). PLA2 expression and activity is significantly elevated in the inflamed intestinal tissues of IBD patients. Since PLA2 enzyme specifically hydrolyses the sn-2 bond within PLs, in our PL-based prodrug approach, the sn-2 positioned FA is replaced with cyclosporine, so that PLA2 may be exploited as the prodrug-activating enzyme, releasing the free drug from the PL-complex. Owing to the enzyme overexpression, this may effectively target free cyclosporine to the sites of inflammation. Four PL-cyclosporine prodrugs were synthesized, differing by their linker length between the PL and the drug moiety. To study the prodrug activation, a novel enzymatically enriched model was developed, the colonic brush border membrane vesicles (cBBMVs); in this model, tissue vesicles were produced from colitis-induced (vs. healthy) rat colons. PLA2 overexpression (3.4-fold) was demonstrated in diseased vs. healthy cBBMVs. Indeed, while healthy cBBMVs induced only marginal activation, substantial prodrug activation was evident by colitis-derived cBBMVs. Together with the PLA2 overexpression, these data validate our drug targeting strategy. In the diseased cBBMVs, quick and complete activation of the entire dose was obtained for the 12-carbon linker prodrug, while slow and marginal activation was obtained for the 6/8-carbon linkers. The potential to target the actual sites of inflammation and treat any localizations throughout the GIT, together with the extended therapeutic index, makes this orally delivered prodrug approach an exciting new therapeutic strategy for IBD treatment.
Collapse
Affiliation(s)
- Milica Markovic
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | - Shimon Ben-Shabat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | | | - Karina Abramov-Harpaz
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.); (Y.M.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Clil Regev
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.); (Y.M.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Yifat Miller
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.); (Y.M.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Aaron Aponick
- Department of Chemistry, University of Florida, Gainesville, FL 32603, USA; (J.N.M.); (A.A.)
| | - Ellen M. Zimmermann
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL 32610, USA;
| | - Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
- Correspondence:
| |
Collapse
|
7
|
Enjavi Y, Sedghamiz MA, Rahimpour MR. Application of nanofluids in drug delivery and disease treatment. NANOFLUIDS AND MASS TRANSFER 2022:449-465. [DOI: 10.1016/b978-0-12-823996-4.00012-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
8
|
|
9
|
Rocha S, Lucas M, Ribeiro D, Corvo ML, Fernandes E, Freitas M. Nano-based drug delivery systems used as vehicles to enhance polyphenols therapeutic effect for diabetes mellitus treatment. Pharmacol Res 2021; 169:105604. [PMID: 33845125 DOI: 10.1016/j.phrs.2021.105604] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/25/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus is one of the biggest health emergencies of the 21st century worldwide, characterized by deficiency in insulin secretion and/or action, leading to hyperglycemia. Despite the currently available antidiabetic therapeutic options, 4.2 million people died in 2019 due to diabetes. Thus, new effective interventions are required. Polyphenols are plant secondary metabolites and have been recognized for their vast number of biological activities, including potential antidiabetic effects. However, the poor bioavailability and high metabolization of polyphenols restrict their biological effects in vivo. Nanotechnology is a promising area of research to improve the therapeutic effect of several compounds. Therefore, this review provides an overview of the literature about the utility of nano-based drug delivery systems as vehicles of polyphenols in diabetes treatment. It was possible to conclude that, in general, nano-based drug delivery systems can potentiate the beneficial antidiabetic properties of polyphenols, when compared with the free compounds, opening a new field of research in diabetology.
Collapse
Affiliation(s)
- Sónia Rocha
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Mariana Lucas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - M Luísa Corvo
- Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
10
|
Song S, Xia H, Guo M, Wang S, Zhang S, Ma P, Jin Y. Role of macrophage in nanomedicine-based disease treatment. Drug Deliv 2021; 28:752-766. [PMID: 33860719 PMCID: PMC8079019 DOI: 10.1080/10717544.2021.1909175] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Macrophages are a major component of the immunoresponse. Diversity and plasticity are two of the hallmarks of macrophages, which allow them to act as proinflammatory, anti-inflammatory, and homeostatic agents. Research has found that cancer and many inflammatory or autoimmune disorders are correlated with activation and tissue infiltration of macrophages. Recent developments in macrophage nanomedicine-based disease treatment are proving to be timely owing to the increasing inadequacy of traditional treatment. Here, we review the role of macrophages in nanomedicine-based disease treatment. First, we present a brief background on macrophages and nanomedicine. Then, we delve into applications of macrophages as a target for disease treatment and delivery systems and summarize the applications of macrophage-derived extracellular vesicles. Finally, we provide an outlook on the clinical utility of macrophages in nanomedicine-based disease treatment.
Collapse
Affiliation(s)
- Siwei Song
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Xia
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sufei Wang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujing Zhang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Deng Z, Liu S. Inflammation-responsive delivery systems for the treatment of chronic inflammatory diseases. Drug Deliv Transl Res 2021; 11:1475-1497. [PMID: 33860447 PMCID: PMC8048351 DOI: 10.1007/s13346-021-00977-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2021] [Indexed: 12/30/2022]
Abstract
Inflammation is the biological response of immune system to protect living organisms from injurious factors. However, excessive and uncontrolled inflammation is implicated in a variety of devastating chronic diseases including atherosclerosis, inflammatory bowel disease (IBD), and rheumatoid arthritis (RA). Improved understanding of inflammatory response has unveiled a rich assortment of anti-inflammatory therapeutics for the treatment and management of relevant chronic diseases. Notwithstanding these successes, clinical outcomes are variable among patients and serious adverse effects are often observed. Moreover, there exist some limitations for clinical anti-inflammatory therapeutics such as aqueous insolubility, low bioavailability, off-target effects, and poor accessibility to subcellular compartments. To address these challenges, the rational design of inflammation-specific drug delivery systems (DDSs) holds significant promise. Moreover, as compared to normal tissues, inflamed tissue-associated pathological milieu (e.g., oxidative stress, acidic pH, and overexpressed enzymes) provides vital biochemical stimuli for triggered delivery of anti-inflammatory agents in a spatiotemporally controlled manner. In this review, we summarize recent advances in the development of anti-inflammatory DDSs with built-in pathological inflammation-specific responsiveness for the treatment of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Zhengyu Deng
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Sciences At the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, Anhui Province, China
| | - Shiyong Liu
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Sciences At the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, Anhui Province, China.
| |
Collapse
|
12
|
Ferreira-Silva M, Faria-Silva C, Viana Baptista P, Fernandes E, Ramos Fernandes A, Corvo ML. Liposomal Nanosystems in Rheumatoid Arthritis. Pharmaceutics 2021; 13:pharmaceutics13040454. [PMID: 33801603 PMCID: PMC8065723 DOI: 10.3390/pharmaceutics13040454] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that affects the joints and results in reduced patient quality of life due to its chronic nature and several comorbidities. RA is also associated with a high socioeconomic burden. Currently, several available therapies minimize symptoms and prevent disease progression. However, more effective treatments are needed due to current therapies' severe side-effects, especially under long-term use. Drug delivery systems have demonstrated their clinical importance-with several nanocarriers present in the market-due to their capacity to improve therapeutic drug index, for instance, by enabling passive or active targeting. The first to achieve market authorization were liposomes that still represent a considerable part of approved delivery systems. In this manuscript, we review the role of liposomes in RA treatment, address preclinical studies and clinical trials, and discuss factors that could hamper a successful clinical translation. We also suggest some alterations that could potentially improve their progression to the market.
Collapse
Affiliation(s)
- Margarida Ferreira-Silva
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (M.F.-S.); (C.F.-S.)
| | - Catarina Faria-Silva
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (M.F.-S.); (C.F.-S.)
| | - Pedro Viana Baptista
- Unidade de Ciências Biomoleculares Aplicadas UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal;
| | - Eduarda Fernandes
- Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (LAQV, REQUIMTE), Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal;
| | - Alexandra Ramos Fernandes
- Unidade de Ciências Biomoleculares Aplicadas UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal;
- Correspondence: (A.R.F.); (M.L.C.)
| | - Maria Luísa Corvo
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (M.F.-S.); (C.F.-S.)
- Correspondence: (A.R.F.); (M.L.C.)
| |
Collapse
|
13
|
Ferreira-Silva M, Faria-Silva C, Baptista PV, Fernandes E, Fernandes AR, Corvo ML. Drug delivery nanosystems targeted to hepatic ischemia and reperfusion injury. Drug Deliv Transl Res 2021; 11:397-410. [PMID: 33660214 DOI: 10.1007/s13346-021-00915-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Hepatic ischemia and reperfusion injury (IRI) is an acute inflammatory process that results from surgical interventions, such as liver resection surgery or transplantation, or hemorrhagic shock. This pathology has become a severe clinical issue, due to the increasing incidence of hepatic cancer and the high number of liver transplants. So far, an effective treatment has not been implemented in the clinic. Despite its importance, hepatic IRI has not attracted much interest as an inflammatory disease, and only a few reviews addressed it from a therapeutic perspective with drug delivery nanosystems. In the last decades, drug delivery nanosystems have proved to be a major asset in therapy because of their ability to optimize drug delivery, either by passive or active targeting. Passive targeting is achieved through the enhanced permeability and retention (EPR) effect, a main feature in inflammation that allows the accumulation of the nanocarriers in inflammation sites, enabling a higher efficacy of treatment than conventional therapies. These systems also can be actively targeted to specific compounds, such as inflammatory markers and overexpressed receptors in immune system intermediaries, allowing an even more specialized therapy that have already showed encouraging results. In this manuscript, we review drug delivery nanosystems designed for hepatic IRI treatment, addressing their current state in clinical trials, discussing the main hurdles that hinder their successful translation to the market and providing some suggestions that could potentially advance their clinical translation.
Collapse
Affiliation(s)
- Margarida Ferreira-Silva
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Catarina Faria-Silva
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Pedro Viana Baptista
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516, Caparica, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Alexandra Ramos Fernandes
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516, Caparica, Portugal
| | - Maria Luísa Corvo
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
14
|
Nanoparticle-siRNA: A potential strategy for rheumatoid arthritis therapy? J Control Release 2020; 325:380-393. [PMID: 32653501 DOI: 10.1016/j.jconrel.2020.07.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/05/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis (RA) is a common clinical inflammatory disease of the autoimmune system manifested by persistent synovitis, cartilage damage and even deformities. Despite significant progress in the clinical treatment of RA, long-term administration of anti-rheumatic drugs can cause a series of problems, including infections, gastrointestinal reactions, and abnormal liver and kidney functions. The emergence of RNA interference (RNAi) drugs has brought new hope for the treatment of RA. Designing a reasonable vector for RNAi drugs will greatly expand the application prospects of RNAi. Nanoparticles as a promising drug carrier provide reliable support for RNAi drugs. The review summarizes the pathogenesis of RA as a possible target for small interference RNA (siRNA) design. At the same time, the review also analyzes the nanoparticles used in siRNA carriers in recent years, laying the foundation and prospect for the next step in the development of intelligent nanocarriers.
Collapse
|
15
|
Dawulieti J, Sun M, Zhao Y, Shao D, Yan H, Lao YH, Hu H, Cui L, Lv X, Liu F, Chi CW, Zhang Y, Li M, Zhang M, Tian H, Chen X, Leong KW, Chen L. Treatment of severe sepsis with nanoparticulate cell-free DNA scavengers. SCIENCE ADVANCES 2020; 6:eaay7148. [PMID: 32523983 PMCID: PMC7259927 DOI: 10.1126/sciadv.aay7148] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 03/25/2020] [Indexed: 05/20/2023]
Abstract
Severe sepsis represents a common, expensive, and deadly health care issue with limited therapeutic options. Gaining insights into the inflammatory dysregulation that causes sepsis would help develop new therapeutic strategies against severe sepsis. In this study, we identified the crucial role of cell-free DNA (cfDNA) in the regulation of the Toll-like receptor 9-mediated proinflammatory pathway in severe sepsis progression. Hypothesizing that removing cfDNA would be beneficial for sepsis treatment, we used polyethylenimine (PEI) and synthesized PEI-functionalized, biodegradable mesoporous silica nanoparticles with different charge densities as cfDNA scavengers. These nucleic acid-binding nanoparticles (NABNs) showed superior performance compared with their nucleic acid-binding polymer counterparts on inhibition of cfDNA-induced inflammation and subsequent multiple organ injury caused by severe sepsis. Furthermore, NABNs exhibited enhanced accumulation and retention in the inflamed cecum, along with a more desirable in vivo safety profile. Together, our results revealed a key contribution of cfDNA in severe sepsis and shed a light on the development of NABN-based therapeutics for sepsis therapy, which currently remains intractable.
Collapse
Affiliation(s)
- Jianati Dawulieti
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences and School of Nursing, Jilin University, Changchun 130021, China
| | - Madi Sun
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences and School of Nursing, Jilin University, Changchun 130021, China
- Institutes of Life Sciences, National Engineering Research Center for Tissue Restoration and Reconstruction, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
| | - Yawei Zhao
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences and School of Nursing, Jilin University, Changchun 130021, China
| | - Dan Shao
- Institutes of Life Sciences, National Engineering Research Center for Tissue Restoration and Reconstruction, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Huize Yan
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Hanze Hu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Lianzhi Cui
- Clinical Laboratory, Jilin Cancer Hospital, Changchun 130012, China
| | - Xiaoyan Lv
- Clinical Laboratory, The Second Hospital of Jilin University, Changchun 130021, China
| | - Feng Liu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Chun-Wei Chi
- Department of Biomedical Engineering CUNY–City College of New York, New York, NY 10031, USA
| | - Yue Zhang
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences and School of Nursing, Jilin University, Changchun 130021, China
| | - Mingqiang Li
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Guangdong Provincial Key Laboratory of Liver Disease, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Ming Zhang
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences and School of Nursing, Jilin University, Changchun 130021, China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Li Chen
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences and School of Nursing, Jilin University, Changchun 130021, China
| |
Collapse
|
16
|
Targeting cancer cells with nanotherapeutics and nanodiagnostics: Current status and future perspectives. Semin Cancer Biol 2020; 69:52-68. [PMID: 32014609 DOI: 10.1016/j.semcancer.2020.01.011] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/23/2020] [Accepted: 01/25/2020] [Indexed: 01/07/2023]
Abstract
Nanotechnology is reshaping health care strategies and is expected to exert a tremendous impact in the coming years offering better healthcare facilities. It has led to not only therapeutic drug delivery feasibility but also to diagnostics. Materials in the size of nano range (1-100 nm) used in the design, fabrication, regulation, and application of therapeutic drugs or devices are classified as medical nanotechnology and nanopharmacology. Delivery of more complex molecules to the specific site of action as well as gene therapy has pushed forward the nanoparticle-based drug delivery to its maximum. Areas that benefit from nano-based drug delivery systems are cancer, diabetes, infectious diseases, neurodegenerative diseases, blood disorders and orthopedic-related ailments. Moreover, development of nanotherapeutics with multi-functionalities has a considerable potential to fill the gaps that exist in the present therapeutic domain. In cancer treatment, nanomedicines have superiority over current therapeutic practices as they can effectively deliver the drug to the affected tissues, thus reducing drug toxicities. Along this line, polymeric conjugates of asparaginase and polymeric micelles of paclitaxel have recently been recommended for the treatment of various types of cancers. Nanotechnology-based therapeutics and diagnostics provide greater effectiveness with less or no toxicity concerns. Similarly, diagnostic imaging holds promising future applications with newer nano-level imaging elements. Advancements in nanotechnology have emerged to a newer direction which use nanorobotics for various applications in healthcare. Accordingly, this review comprehensively highlights the potentialities of various nanocarriers and nanomedicines for multifaceted applications in diagnostics and drug delivery, especially the potentialities of polymeric nanoparticle, nanoemulsion, solid-lipid nanoparticle, nanostructured lipid carrier, self-micellizing anticancer lipids, dendrimer, nanocapsule and nanosponge-based therapeutic approaches in the field of cancer. Furthermore, this article summarizes the most recent literature pertaining to the use of nano-technology in the field of medicine, particularly in treating cancer patients.
Collapse
|
17
|
Nurhidayah D, Maruf A, Zhang X, Liao X, Wu W, Wang G. Advanced drug-delivery systems: mechanoresponsive nanoplatforms applicable in atherosclerosis management. Nanomedicine (Lond) 2019; 14:3105-3122. [PMID: 31823682 DOI: 10.2217/nnm-2019-0172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nanoplatforms have been used extensively as advanced carriers to enhance the effectiveness of drug delivery, mostly through passive aggregation provided by the enhanced permeability and retention effect. Mechanical stimuli provide a robust strategy to bolster drug delivery performance by increasing the accumulation of nanoplatforms at the lesion sites, facilitating on-demand cargo release and providing theranostic aims. In this review, we focus on recent advances of mechanoresponsive nanoplatforms that can accomplish targeted drug delivery, and subsequent drug release, under specific stimuli, either endogenous (shear stress) or exogenous (magnetic field and ultrasound), to synergistically combat atherosclerosis at the molecular level.
Collapse
Affiliation(s)
- Deti Nurhidayah
- Key Laboratory for Biorheological Science & Technology of Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Ali Maruf
- Key Laboratory for Biorheological Science & Technology of Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Xiaojuan Zhang
- Key Laboratory for Biorheological Science & Technology of Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Xiaoling Liao
- Chongqing Engineering Laboratory of Nano/Micro Biological Medicine Detection Technology, Chongqing University of Science & Technology, Chongqing 401331, China
| | - Wei Wu
- Key Laboratory for Biorheological Science & Technology of Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science & Technology of Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| |
Collapse
|
18
|
Gummy gold and silver nanoparticles of apricot (Prunus armeniaca) confer high stability and biological activity. ARAB J CHEM 2019. [DOI: 10.1016/j.arabjc.2016.02.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
19
|
Wang X, Feng Y, Fu J, Wu C, He B, Zhang H, Wang X, Dai W, Sun Y, Zhang Q. A Lipid Micellar System Loaded with Dexamethasone Palmitate Alleviates Rheumatoid Arthritis. AAPS PharmSciTech 2019; 20:316. [PMID: 31602546 DOI: 10.1208/s12249-019-1449-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/01/2019] [Indexed: 12/22/2022] Open
Abstract
Glucocorticoids have been confirmed to be effective in the treatment of a variety of inflammatory diseases. However, their application encounters limitations in terms of tissue distribution and bioavailability in vivo. To address these key issues, we designed and developed a nanopreparation by using egg yolk lecithin/sodium glycocholate (EYL/SGC) and utilize such mixed micelles (MMs) to encapsulate dexamethasone palmitate (DMP) for the treatment of rheumatoid arthritis (RA). The prepared DMP-MMs had an average particle size of 49.18 ± 0.43 nm and were compared with an emulsion-based dexamethasone palmitate. Pharmacokinetic and in vivo fluorescence imaging showed that mixed micelles had higher bioavailability and targeting efficiency in inflammatory sites. An arthritis rat model was established via induction by Complete Freund's Adjuvant (CFA), followed by the efficacy studies by the observations of paw volume, histology, spleen index, pro-inflammatory cytokines, and CT images. It was confirmed that intravenous injection of DMP-MMs exhibited advantages in alleviating joint inflammation compared with the emulsion system. Composed of pharmaceutical adjuvants only, the nanoscale mixed micelles seem a promising carrier system for the RA treatment with lipophilic drugs.
Collapse
|
20
|
Song P, Yang C, Thomsen JS, Dagnæs-Hansen F, Jakobsen M, Brüel A, Deleuran B, Kjems J. Lipidoid-siRNA Nanoparticle-Mediated IL-1β Gene Silencing for Systemic Arthritis Therapy in a Mouse Model. Mol Ther 2019; 27:1424-1435. [PMID: 31153827 DOI: 10.1016/j.ymthe.2019.05.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 11/18/2022] Open
Abstract
Interleukin-1 beta (IL-1β) plays a central role in the induction of rheumatoid arthritis (RA). In the present study, we demonstrated that lipidoid-polymer hybrid nanoparticle (FS14-NP) can efficiently deliver siRNA against IL-1β (siIL-1β) to macrophages and effectively suppress the pathogenesis of experimental arthritis induced by collagen antibody (CAIA mice). FS14-NP/siIL-1β achieved approximately 70% and 90% gene-silencing efficiency in the RAW 264.7 cell line and intraperitoneal macrophages, respectively. Intravenous administration of FS14-NP/siRNA led to rapid accumulation of siRNA in macrophages within the arthritic joints. Furthermore, FS14-NP/siIL-1β treatment lowered the expression of pro-inflammatory cytokines in arthritic joints and dramatically attenuated ankle swelling, bone erosion, and cartilage destruction. These results demonstrate that FS14-NP/siIL-1β may represent an effective therapy for systemic arthritis and other inflammatory disorders.
Collapse
Affiliation(s)
- Ping Song
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, 8000 Aarhus C, Denmark; Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Chuanxu Yang
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, 8000 Aarhus C, Denmark; Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark.
| | | | | | - Maria Jakobsen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Bent Deleuran
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; Department of Rheumatology, Aarhus University Hospital, 8000 Aarhus C, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, 8000 Aarhus C, Denmark; Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
21
|
Chen X, Liu Y, Wen Y, Yu Q, Liu J, Zhao Y, Liu J, Ye G. A photothermal-triggered nitric oxide nanogenerator combined with siRNA for precise therapy of osteoarthritis by suppressing macrophage inflammation. NANOSCALE 2019; 11:6693-6709. [PMID: 30900717 DOI: 10.1039/c8nr10013f] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Although nitric oxide (NO) can be used to treat osteoarthritis (OA) by inhibiting inflammation, a method for the accurately controlled release of NO in inflammatory cells is still elusive. Herein, photothermal-triggered NO nanogenerators NO-Hb@siRNA@PLGA-PEG (NHsPP) were constructed by assembling photothermal-agents and NO molecules within nanoparticles. In the NHsPP nanoparticles the hemoglobin (Hb) nanoparticles can act as a NO carrier which can absorb near-infrared light at 650 nm (0.5 W cm-2) and convert it into heat to trigger the release of NO. Moreover, after loading Notch1-siRNA, precise treatment can be achieved. Furthermore, using the synergistic effect of photothermal therapy, the NHsPP nanoparticles achieved simultaneous treatment with NO, siRNA and PTT. Through this combination therapy, the therapeutic effect of the NHsPP nanoparticles was significantly enhanced compared to the treatment groups using only NO, siRNA or PTT. This combination therapy inhibits the inflammatory response effectively by reducing the level of pro-inflammatory cytokines and the macrophage response. Subsequently, guided by dual-modal imaging, the NHsPP nanoparticles can not only accumulate effectively in OA mice, but can also reduce the inflammatory response and efficiently prevent cartilage erosion, without causing toxic side effects in the major organs. Therefore, this novel photothermal nanoparticle-based NO-releasing system is expected to be a potential alternative for clinical inflammatory disease therapy and may provide image guidance when combined with other nanotherapy systems.
Collapse
Affiliation(s)
- Xu Chen
- Department of Chemistry, Jinan University. Guangzhou, 510632, China.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Yildiz T, Gu R, Zauscher S, Betancourt T. Doxorubicin-loaded protease-activated near-infrared fluorescent polymeric nanoparticles for imaging and therapy of cancer. Int J Nanomedicine 2018; 13:6961-6986. [PMID: 30464453 PMCID: PMC6217908 DOI: 10.2147/ijn.s174068] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Despite significant progress in the field of oncology, cancer remains one of the leading causes of death. Chemotherapy is one of the most common treatment options for cancer patients but is well known to result in off-target toxicity. Theranostic nanomedicines that integrate diagnostic and therapeutic functions within an all-in-one platform can increase tumor selectivity for more effective chemotherapy and aid in diagnosis and monitoring of therapeutic responses. MATERIAL AND METHODS In this work, theranostic nanoparticles were synthesized with commonly used biocompatible and biodegradable polymers and used as cancer contrast and therapeutic agents for optical imaging and treatment of breast cancer. These core-shell nanoparticles were prepared by nanoprecipitation of blends of the biodegradable and biocompatible amphiphilic copolymers poly(lactic-co-glycolic acid)-b-poly-l-lysine and poly(lactic acid)-b-poly(ethylene glycol). Poly-l-lysine in the first copolymer was covalently decorated with near-infrared fluorescent Alexa Fluor 750 molecules. RESULTS The spherical nanoparticles had an average size of 60-80 nm. The chemotherapeutic drug doxorubicin was encapsulated in the core of nanoparticles at a loading of 3% (w:w) and controllably released over a period of 30 days. A 33-fold increase in near-infrared fluorescence, mediated by protease-mediated cleavage of the Alexa Fluor 750-labeled poly-l-lysine on the surface of the nanoparticles, was observed upon interaction with the model protease trypsin. The cytocompatibility of drug-free nanoparticles and growth inhibition of drug-loaded nanoparticles on MDA-MB-231 breast cancer cells were investigated with a luminescence cell-viability assay. Drug-free nanoparticles were found to cause minimal toxicity, even at high concentrations (0.2-2,000 µg/mL), while doxorubicin-loaded nanoparticles significantly reduced cell viability at drug concentrations >10 µM. Finally, the interaction of the nanoparticles with breast cancer cells was studied utilizing fluorescence microscopy, demonstrating the potential of the nanoparticles to act as near-infrared fluorescence optical imaging agents and drug-delivery carriers. CONCLUSION Doxorubicin-loaded, enzymatically activatable nanoparticles of less than 100 nm were prepared successfully by nanoprecipitation of copolymer blends. These nanoparticles were found to be suitable as controlled drug delivery systems and contrast agents for imaging of cancer cells.
Collapse
Affiliation(s)
- Tugba Yildiz
- Materials Science, Engineering, and Commercialization Program, Texas State University, San Marcos, TX,
| | - Renpeng Gu
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC
| | - Stefan Zauscher
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC
| | - Tania Betancourt
- Materials Science, Engineering, and Commercialization Program, Texas State University, San Marcos, TX,
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA,
| |
Collapse
|
23
|
Thakur S, Riyaz B, Patil A, Kaur A, Kapoor B, Mishra V. Novel drug delivery systems for NSAIDs in management of rheumatoid arthritis: An overview. Biomed Pharmacother 2018; 106:1011-1023. [DOI: 10.1016/j.biopha.2018.07.027] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 12/21/2022] Open
|
24
|
Gauthier A, Fisch A, Seuwen K, Baumgarten B, Ruffner H, Aebi A, Rausch M, Kiessling F, Bartneck M, Weiskirchen R, Tacke F, Storm G, Lammers T, Ludwig MG. Glucocorticoid-loaded liposomes induce a pro-resolution phenotype in human primary macrophages to support chronic wound healing. Biomaterials 2018; 178:481-495. [DOI: 10.1016/j.biomaterials.2018.04.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/27/2018] [Accepted: 04/02/2018] [Indexed: 02/07/2023]
|
25
|
Hua S, de Matos MBC, Metselaar JM, Storm G. Current Trends and Challenges in the Clinical Translation of Nanoparticulate Nanomedicines: Pathways for Translational Development and Commercialization. Front Pharmacol 2018; 9:790. [PMID: 30065653 PMCID: PMC6056679 DOI: 10.3389/fphar.2018.00790] [Citation(s) in RCA: 555] [Impact Index Per Article: 79.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/28/2018] [Indexed: 01/02/2023] Open
Abstract
The use of nanotechnology in medicine has the potential to have a major impact on human health for the prevention, diagnosis, and treatment of diseases. One particular aspect of the nanomedicine field which has received a great deal of attention is the design and development of nanoparticulate nanomedicines (NNMs) for drug delivery (i.e., drug-containing nanoparticles). NNMs are intended to deliver drugs via various mechanisms: solubilization, passive targeting, active targeting, and triggered release. The NNM approach aims to increase therapeutic efficacy, decrease the therapeutically effective dose, and/or reduce the risk of systemic side effects. In order to move a NNM from the bench to the bedside, several experimental challenges need to be addressed. This review will discuss the current trends and challenges in the clinical translation of NNMs as well as the potential pathways for translational development and commercialization. Key issues related to the clinical development of NNMs will be covered, including biological challenges, large-scale manufacturing, biocompatibility and safety, intellectual property (IP), government regulations, and overall cost-effectiveness in comparison to current therapies. These factors can impose significant hurdles limiting the appearance of NNMs on the market, irrelevant of whether they are therapeutically beneficial or not.
Collapse
Affiliation(s)
- Susan Hua
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW, Australia
| | - Maria B C de Matos
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Josbert M Metselaar
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Netherlands.,Department of Experimental Molecular Imaging, RWTH University Clinic Aachen, Aachen, Germany
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Netherlands.,Imaging Division, University Medical Centre Utrecht, Utrecht, Netherlands
| |
Collapse
|
26
|
Palmioli A, La Ferla B. Glycofunctionalization of Poly(lactic-co-glycolic acid) Polymers: Building Blocks for the Generation of Defined Sugar-Coated Nanoparticles. Org Lett 2018; 20:3509-3512. [DOI: 10.1021/acs.orglett.8b01287] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Alessandro Palmioli
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milan, Italy
| | - Barbara La Ferla
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milan, Italy
| |
Collapse
|
27
|
Al-Ahmady ZS. Selective drug delivery approaches to lesioned brain through blood brain barrier disruption. Expert Opin Drug Deliv 2018; 15:335-349. [DOI: 10.1080/17425247.2018.1444601] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Zahraa S. Al-Ahmady
- Nanomedicine Lab, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Heath, University of Manchester, UK
| |
Collapse
|
28
|
Horvatova A, Utaipan T, Otto AC, Zhang Y, Gan-Schreier H, Pavek P, Pathil A, Stremmel W, Chamulitrat W. Ursodeoxycholyl lysophosphatidylethanolamide negatively regulates TLR-mediated lipopolysaccharide response in human THP-1-derived macrophages. Eur J Pharmacol 2018; 825:63-74. [PMID: 29475064 DOI: 10.1016/j.ejphar.2018.02.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/15/2018] [Accepted: 02/19/2018] [Indexed: 11/26/2022]
Abstract
The bile acid-phospholipid conjugate ursodeoxycholyl oleoyl-lysophophatidylethanolamide (UDCA-18:1LPE) is an anti-inflammatory and anti-fibrotic agent as previously shown in cultured hepatocytes and hepatic stellate cells as well as in in vivo models of liver injury. We hypothesize that UDCA-18:1LPE may directly inhibit the activation of immune cells. We found that UDCA-18:1LPE was capable of inhibiting the migration of phorbol ester-differentiated human THP-1 cells. We examined anti-inflammatory activity of UDCA-18:1LPE during activation of THP1-derived macrophages. Treatment of these macrophages by bacterial lipopolysaccharide (LPS) for 24 h induced the release of pro-inflammatory cytokines TNF-α, IL-6 and IL-1β. This release was markedly inhibited by pretreatment with UDCA-18:1LPE by ~ 65-90%. Derivatives with a different fatty-acid chain in LPE moiety also exhibited anti-inflammatory property. Western blotting and indirect immunofluorescence analyses revealed that UDCA-18:1LPE attenuated the expression of phosphorylated p38, MKK4/MKK7, JNK1/2, and c-Jun as well as nuclear translocation of NF-κB by ~ 22-86%. After LPS stimulation, the Toll-like receptor adaptor proteins, myeloid differentiation factor 88 and TNF receptor associated factor 6, were recruited into lipid rafts and UDCA-18:1LPE inhibited this recruitment by 22% and 58%, respectively. Moreover, LPS treatment caused a decrease of the known cytoprotective lysophosphatidylcholine species containing polyunsaturated fatty acids by 43%, and UDCA-18:1LPE co-treatment reversed this decrease. In conclusion, UDCA-18:1LPE and derivatives inhibited LPS inflammatory response by interfering with Toll-like receptor signaling in lipid rafts leading to an inhibition of MAPK and NF-κB activation. These conjugates may represent a class of lead compounds for development of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Alzbeta Horvatova
- Department of Internal Medicine IV, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University in Prague, Heyorovskeho 1203, 500 05 Hradec Kralove, Czech Republic
| | - Tanyarath Utaipan
- Department of Pre-Clinic, Faculty of Sciences and Technology, Prince of Songkla University, Pattani Campus, 94000 Pattani, Thailand
| | - Ann-Christin Otto
- Department of Internal Medicine IV, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Yuling Zhang
- Department of Internal Medicine IV, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Hongying Gan-Schreier
- Department of Internal Medicine IV, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Petr Pavek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University in Prague, Heyorovskeho 1203, 500 05 Hradec Kralove, Czech Republic
| | - Anita Pathil
- Department of Internal Medicine IV, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Wolfgang Stremmel
- Department of Internal Medicine IV, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Walee Chamulitrat
- Department of Internal Medicine IV, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| |
Collapse
|
29
|
Abstract
The phenomenal advances in pharmaceutical sciences over the last few decades have led to the development of new therapeutics like peptides, proteins, RNAs, DNAs and highly potent small molecules. Fruitful applications of these therapeutics have been challenged by several anatomical and physiological barriers that limit adequate drug disposition at the site-of-action and by off-target drug distribution to undesired tissues, which together result in the reduced effectiveness and increased side effects of therapeutic agents. As such, the development of drug delivery and targeting systems has been recognised as a cornerstone for future drug development. Research in pharmaceutical sciences is now devoted to tackling delivery challenges through engineering delivery systems that move beyond conventional dosage forms and regimens into state-of-the-art targeted drug delivery tailored toward specific therapeutic needs. Modern drug delivery systems comprise passive and active targeting approaches. While passive targeting relies on the natural course of distribution of drugs or drug carriers in the body, as governed by their physicochemical properties, active targeting often exploits targeting moieties that home preferentially into target tissues. Here, we provide an overview of theories of and approaches to passive and active drug delivery. As the design of drug delivery is dependent on the unique structure of target tissues and organs, we present our discussion in an organ-specific manner with the aim to inspire the development of new strategies for curing disease with high accuracy and efficiency.
Collapse
Affiliation(s)
- Mohammad Alsaggar
- a Department of Pharmaceutical Technology, College of Pharmacy , Jordon University of Science and Technology , Irbid , Jordan
| | - Dexi Liu
- b Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy , University of Georgia , Athens , GA , USA
| |
Collapse
|
30
|
Prasad M, Lambe UP, Brar B, Shah I, J M, Ranjan K, Rao R, Kumar S, Mahant S, Khurana SK, Iqbal HMN, Dhama K, Misri J, Prasad G. Nanotherapeutics: An insight into healthcare and multi-dimensional applications in medical sector of the modern world. Biomed Pharmacother 2018; 97:1521-1537. [PMID: 29793315 DOI: 10.1016/j.biopha.2017.11.026] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/28/2017] [Accepted: 11/03/2017] [Indexed: 02/08/2023] Open
Abstract
In recent years nanotechnology has revolutionized the healthcare strategies and envisioned to have a tremendous impact to offer better health facilities. In this context, medical nanotechnology involves design, fabrication, regulation, and application of therapeutic drugs and devices having a size in nano-range (1-100 nm). Owing to the revolutionary implications in drug delivery and gene therapy, nanotherapeutics has gained increasing research interest in the current medical sector of the modern world. The areas which anticipate benefits from nano-based drug delivery systems are cancer, diabetes, infectious diseases, neurodegenerative diseases, blood disorders and orthopedic problems. The development of nanotherapeutics with multi-functionalities has considerable potential to fill the lacunae existing in the present therapeutic domain. Nanomedicines in the field of cancer management have enhanced permeability and retention of drugs thereby effectively targeting the affected tissues. Polymeric conjugates of asparaginase, polymeric micelles of paclitaxel have been recmended for various types of cancer treatment .The advancement of nano therapeutics and diagnostics can provide the improved effectiveness of the drug with less or no toxicity concerns. Similarly, diagnostic imaging is having potential future applications with newer imaging elements at nano level. The newly emerging field of nanorobotics can provide new directions in the field of healthcare. In this article, an attempt has been made to highlight the novel nanotherapeutic potentialities of polymeric nanoparticles, nanoemulsion, solid lipid nanoparticle, nanostructured lipid carriers, dendrimers, nanocapsules and nanosponges based approaches. The useful applications of these nano-medicines in the field of cancer, nutrition, and health have been discussed in details. Regulatory and safety concerns along with the commercial status of nanosystems have also been presented. In summary, a successful translation of emerging nanotherapeutics into commercial products may lead to an expansion of biomedical science. Towards the end of the review, future perspectives of this important field have been introduced briefly.
Collapse
Affiliation(s)
- Minakshi Prasad
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar, Haryana, 125004, India.
| | - Upendra P Lambe
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar, Haryana, 125004, India
| | - Basanti Brar
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar, Haryana, 125004, India
| | - Ikbal Shah
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar, Haryana, 125004, India
| | - Manimegalai J
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar, Haryana, 125004, India
| | - Koushlesh Ranjan
- Department of Veterinary Physiology and Biochemistry, Sardar Vallabhbhai Patel University of Agriculture and Technology, Meerut, Uttar Pradesh, 250110, India
| | - Rekha Rao
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India
| | - Sheefali Mahant
- Department of Pharmaceutical Sciences, Maharishi Dayanand University, Rohtak, Haryana, 124001, India
| | - Sandip Kumar Khurana
- Central Institute for Research on Buffaloes, Sirsa Road, Hisar, Haryana, 125001, India
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, N. L., CP 64849, Mexico
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, India
| | - Jyoti Misri
- Division of Animal Health, Indian Council of Agriculture Research, New Delhi, India
| | - Gaya Prasad
- Sardar Vallabhbhai Patel University of Agriculture and Technology, Meerut, Uttar Pradesh, 250110, India
| |
Collapse
|
31
|
Alaarg A, Pérez-Medina C, Metselaar JM, Nahrendorf M, Fayad ZA, Storm G, Mulder WJM. Applying nanomedicine in maladaptive inflammation and angiogenesis. Adv Drug Deliv Rev 2017; 119:143-158. [PMID: 28506745 PMCID: PMC5682240 DOI: 10.1016/j.addr.2017.05.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 04/12/2017] [Accepted: 05/09/2017] [Indexed: 12/11/2022]
Abstract
Inflammation and angiogenesis drive the development and progression of multiple devastating diseases such as atherosclerosis, cancer, rheumatoid arthritis, and inflammatory bowel disease. Though these diseases have very different phenotypic consequences, they possess several common pathophysiological features in which monocyte recruitment, macrophage polarization, and enhanced vascular permeability play critical roles. Thus, developing rational targeting strategies tailored to the different stages of the journey of monocytes, from bone marrow to local lesions, and their extravasation from the vasculature in diseased tissues will advance nanomedicine. The integration of in vivo imaging uniquely allows studying nanoparticle kinetics, accumulation, clearance, and biological activity, at levels ranging from subcellular to an entire organism, and will shed light on the fate of intravenously administered nanomedicines. We anticipate that convergence of nanomedicines, biomedical engineering, and life sciences will help to advance clinically relevant therapeutics and diagnostic agents for patients with chronic inflammatory diseases.
Collapse
Affiliation(s)
- Amr Alaarg
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Josbert M Metselaar
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Institute for Experimental Molecular Imaging, University Clinic, Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gert Storm
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
32
|
|
33
|
Alaarg A, Senders ML, Varela-Moreira A, Pérez-Medina C, Zhao Y, Tang J, Fay F, Reiner T, Fayad ZA, Hennink WE, Metselaar JM, Mulder WJM, Storm G. A systematic comparison of clinically viable nanomedicines targeting HMG-CoA reductase in inflammatory atherosclerosis. J Control Release 2017; 262:47-57. [PMID: 28700897 DOI: 10.1016/j.jconrel.2017.07.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/27/2017] [Accepted: 07/07/2017] [Indexed: 12/21/2022]
Abstract
Atherosclerosis is a leading cause of worldwide morbidity and mortality whose management could benefit from novel targeted therapeutics. Nanoparticles are emerging as targeted drug delivery systems in chronic inflammatory disorders. To optimally exploit nanomedicines, understanding their biological behavior is crucial for further development of clinically relevant and efficacious nanotherapeutics intended to reduce plaque inflammation. Here, three clinically relevant nanomedicines, i.e., high-density lipoprotein ([S]-HDL), polymeric micelles ([S]-PM), and liposomes ([S]-LIP), that are loaded with the HMG-CoA reductase inhibitor simvastatin [S], were evaluated in the apolipoprotein E-deficient (Apoe-/-) mouse model of atherosclerosis. We systematically employed quantitative techniques, including in vivo positron emission tomography imaging, gamma counting, and flow cytometry to evaluate the biodistribution, nanomedicines' uptake by plaque-associated macrophages/monocytes, and their efficacy to reduce macrophage burden in atherosclerotic plaques. The three formulations demonstrated distinct biological behavior in Apoe-/- mice. While [S]-PM and [S]-LIP possessed longer circulation half-lives, the three platforms accumulated to similar levels in atherosclerotic plaques. Moreover, [S]-HDL and [S]-PM showed higher uptake by plaque macrophages in comparison to [S]-LIP, while [S]-PM demonstrated the highest uptake by Ly6Chigh monocytes. Among the three formulations, [S]-PM displayed the highest efficacy in reducing macrophage burden in advanced atherosclerotic plaques. In conclusion, our data demonstrate that [S]-PM is a promising targeted drug delivery system, which can be advanced for the treatment of atherosclerosis and other inflammatory disorders in the clinical settings. Our results also emphasize the importance of a thorough understanding of nanomedicines' biological performance, ranging from the whole body to the target cells, as well drug retention in the nanoparticles. Such systematic investigations would allow rational applications of nanomaterials', beyond cancer, facilitating the expansion of the nanomedicine horizon.
Collapse
Affiliation(s)
- Amr Alaarg
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands; Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Max L Senders
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medical Biochemistry, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Aida Varela-Moreira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands; Department of Clinical Chemistry and Haematology, University Medical Centre Utrecht, Utrecht 3584 CX, The Netherlands
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yiming Zhao
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jun Tang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Francois Fay
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Chemistry, York College of The City University of New York, New York, NY 11451, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Josbert M Metselaar
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands; Department of Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen 52074, Germany
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medical Biochemistry, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands.
| | - Gert Storm
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands; Imaging Division, University Medical Centre Utrecht, Utrecht 3584 CX, The Netherlands.
| |
Collapse
|
34
|
Palange AL, Palomba R, Rizzuti IF, Ferreira M, Decuzzi P. Deformable Discoidal Polymeric Nanoconstructs for the Precise Delivery of Therapeutic and Imaging Agents. Mol Ther 2017; 25:1514-1521. [PMID: 28341562 DOI: 10.1016/j.ymthe.2017.02.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/10/2017] [Accepted: 02/10/2017] [Indexed: 02/05/2023] Open
Abstract
Over the last 15 years, a plethora of materials and different formulations have been proposed for the realization of nanomedicines. Yet drug-loading efficiency, sequestration by phagocytic cells, and tumor accumulation are sub-optimal. This would imply that radically new design approaches are needed to propel the clinical integration of nanomedicines, overcoming well-accepted clichés. This work briefly reviews the use of deformable discoidal nanoconstructs as a novel delivery strategy for therapeutic and imaging agents. Inspired by blood cell behavior, these nanoconstructs are designed to efficiently navigate the circulatory system, minimize sequestration by phagocytic cells, and recognize the tortuous angiogenic microvasculature of neoplastic masses. This article discusses the notion of nanoparticle margination and vascular adhesion, as well as advantages associated with deformable particles. Finally, details on the synthesis, physico-chemical properties, and in vivo characterization of discoidal polymeric nanoconstructs are provided, with particular emphasis on their ability to independently control size, shape, surface properties, and mechanical stiffness. These nanoconstructs could help in gaining a deeper understanding of the mechanisms regulating the behavior of nanomedicines and identifying optimal delivery strategies for patient-specific therapeutic interventions.
Collapse
Affiliation(s)
- Anna Lisa Palange
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Roberto Palomba
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Ilaria F Rizzuti
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Miguel Ferreira
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa 16163, Italy.
| |
Collapse
|
35
|
Gandomi N, Varshochian R, Atyabi F, Ghahremani MH, Sharifzadeh M, Amini M, Dinarvand R. Solid lipid nanoparticles surface modified with anti-Contactin-2 or anti-Neurofascin for brain-targeted delivery of medicines. Pharm Dev Technol 2017; 22:426-435. [DOI: 10.1080/10837450.2016.1226901] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Nargess Gandomi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Varshochian
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Sakurai Y, Kajimoto K, Harashima H. Anti-angiogenic nanotherapy via active targeting systems to tumors and adipose tissue vasculature. Biomater Sci 2017; 3:1253-65. [PMID: 26261854 DOI: 10.1039/c5bm00113g] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Sophisticated drug delivery systems (DDS) are required for delivering drugs, especially macromolecules such as nucleic acids or proteins, to their sites of action. Therefore it is a prerequisite that future DDS are designed to selectively target a tissue. In this review, we focus on systems that actively target the vasculature in tumors or adipose tissues. For targeting tumor vasculatur, a new strategy referred to as dual-targeting is proposed that uses a combination of a receptor specific ligand and a cell penetrating peptide, which can induce the synergistic enhancement of tissue selectivity under in vivo conditions. A novel pH-sensitive cationic lipid was designed to enhance the endosomal release of encapsulated compounds such as siRNA as well as to improve the stability in blood circulation after intravenous administration. A cyclic RGD peptide is used as an active targeting ligand. For targeting adipose vasculature, prohibitin, which is expressed on the surface of adipose endothelial cells, was targeted with KGGRAKD peptides on the surface of PEGylated nanoparticles. Prohibitin targeted nanoparticles (PTNP) encapsulating Cytochrome c (CytC) can selectively target adipose vasculature by optimizing the lengths of the PEG linkers and can deliver CytC to adipose endothelial cells. PTNP can successfully induce anti-obese effects as well as apoptosis by delivering CytC to the cytosol in endothelial cells. Unexpectedly, the EPR (enhanced permeability and retention) effect, which is usually observed in tumor tissue, was also observed in the adipose vasculature, especially in obese mice, where PEGylated nanoparticles can pass through the endothelial barriers in adipose tissue. We believe that these achievements in active targeting will allow a greatly expanded use of DDS for nanomedicines.
Collapse
Affiliation(s)
- Yu Sakurai
- Faculty of Pharmaceutical Sciences, Hokkaido University, Japan.
| | | | | |
Collapse
|
37
|
Smith AAA, Zuwala K, Pilgram O, Johansen KS, Tolstrup M, Dagnæs-Hansen F, Zelikin AN. Albumin-Polymer-Drug Conjugates: Long Circulating, High Payload Drug Delivery Vehicles. ACS Macro Lett 2016; 5:1089-1094. [PMID: 35658186 DOI: 10.1021/acsmacrolett.6b00544] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Albumin is an exquisite tool of nature used in biomedicine to achieve long blood residence time for drugs, but the payload it can carry is typically limited to one molecule per protein. In contrast, synthetic macromolecular prodrugs contain multiple copies of drugs per polymer chain but offer only a marginal increase in the circulation lifetime of the drugs. We combine the benefits of the two platforms and at the same time overcome their respective limitations. Specifically, we develop the synthesis of albumin-polymer-drug conjugates to obtain long circulating, high payload drug delivery vehicles. In vivo data validate that albumin endows the conjugate with a blood residence time similar to that of the protein and well exceeding that of the polymer. Therapeutic activity of the conjugates is validated using prodrugs of panobinostat, an HIV latency reversal agent, in which case the conjugates matched the drug in terms of efficacy of treatment.
Collapse
Affiliation(s)
| | - Kaja Zuwala
- Department of Infectious Diseases, Aarhus University Hospital, DK-8000 Aarhus C, Denmark
| | | | | | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, DK-8000 Aarhus C, Denmark
| | | | | |
Collapse
|
38
|
Nam EJ, Kang JH, Sa KH, Sung S, Park JY, Jo DG, Park JH, Kim IS, Kang YM. Robust Therapeutic Efficacy of Matrix Metalloproteinase-2-Cleavable Fas-1-RGD Peptide Complex in Chronic Inflammatory Arthritis. PLoS One 2016; 11:e0164102. [PMID: 27741237 PMCID: PMC5065154 DOI: 10.1371/journal.pone.0164102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 09/20/2016] [Indexed: 12/29/2022] Open
Abstract
Objective Therapeutic agents that are transformable via introducing cleavable linkage by locally enriched MMP-2 within inflamed synovium would enhance therapeutic efficacy on chronic inflammatory arthritis. Transforming growth factor-β-inducible gene-h3 (βig-h3), which consists of four fas-1 domains and an Arg-Gly-Asp (RGD) motif, intensifies inflammatory processes by facilitating adhesion and migration of fibroblast-like synoviocyte in the pathogenesis of rheumatoid arthritis (RA). The aim of this study was to investigate whether a MMP-2-cleavable peptide complex consisting of a fas-1 domain and an RGD peptide blocks the interaction between βig-h3 and resident cells and leads to the amelioration of inflammatory arthritis. Methods We designed βig-h3-derivatives, including the fourth fas-1 domain truncated for H1 and H2 sequences of mouse (MFK00) and MMP-2-cleavable peptide complex (MFK902). MMP-2 selectivity was examined by treatment with a series of proteases. MFK902 efficacy was determined by the adhesion and migration assay with NIH3T3 cells in vitro and collagen-induced arthritis (CIA) model using male DBA/1J mice in vivo. The mice were treated intraperitoneally with MFK902 at different dosages. Results MFK902 was specifically cleaved by active MMP-2 in a concentration-dependent manner, and βig-h3-mediated adhesion and migration were more effectively inhibited by MFK902, compared with RGD or MFK00 peptides. The arthritis activity of murine CIA, measured by clinical arthritis index and incidence of arthritic paws, was significantly ameliorated after treatment with all dosages of MFK902 (1, 10, and 30 mg/kg). MFK902 ameliorated histopathologic deterioration and reduced the expression of inflammatory mediators simultaneously with improvement of clinical features. In addition, a favorable safety profile of MFK902 was demonstrated in vivo. Conclusion The present study revealed that MMP-2-cleavable peptide complex based on βig-h3 structure is a potent and safe therapeutic agent for chronic inflammatory arthritis, thus providing reliable evidence for a MMP-2-cleavable mechanism as a tissue-targeted strategy for treatment of RA.
Collapse
Affiliation(s)
- Eon Jeong Nam
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University, School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu 41944, South Korea
- Cell and Matrix Research Institute, Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu 41944, South Korea
| | - Jin Hee Kang
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University, School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu 41944, South Korea
| | - Keum Hee Sa
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University, School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu 41944, South Korea
| | - Shijin Sung
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University, School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu 41944, South Korea
| | - Jae Yong Park
- Cell and Matrix Research Institute, Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu 41944, South Korea
- Division of Pulmonology, Department of Internal Medicine, Kyungpook National University, School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu 41944, South Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangangu, Suwon 16419, Republic of Korea
| | - Jae Hyung Park
- Department of Polymer Science and Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangangu, Suwon 16419, Republic of Korea
| | - In San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongukgu, Seoul 02792, Republic of Korea
| | - Young Mo Kang
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University, School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu 41944, South Korea
- Cell and Matrix Research Institute, Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu 41944, South Korea
- Department of Biochemistry and Cellular Biology, Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea
- * E-mail:
| |
Collapse
|
39
|
van der Geest T, Laverman P, Gerrits D, Walgreen B, Helsen MM, Klein C, Nayak TK, Storm G, Metselaar JM, Koenders MI, Boerman OC. Liposomal Treatment of Experimental Arthritis Can Be Monitored Noninvasively with a Radiolabeled Anti-Fibroblast Activation Protein Antibody. J Nucl Med 2016; 58:151-155. [PMID: 27493266 DOI: 10.2967/jnumed.116.177931] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 07/13/2016] [Indexed: 11/16/2022] Open
Abstract
Rheumatoid arthritis is a chronic autoimmune disorder resulting in synovial inflammation. Fibroblast activation protein (FAP) is overexpressed by fibroblastlike synoviocytes in arthritic joints. Radioimmunoimaging with an anti-FAP antibody might be used to monitor the response to therapy, thus enabling tailored therapy strategies and therapeutic outcomes. The aim of this study was to assess whether a radiolabeled anti-FAP antibody could be used to monitor the efficacy of treatment with long-circulating liposomes (LCL) containing prednisolone phosphate (PLP-LCL) in a mouse model of arthritis. METHODS Collagen-induced arthritis (CIA) was induced in male DBA/1J mice. Mice were treated with a single injection (10 mg/kg) of PLP-LCL or empty LCL as a control. SPECT and CT images were acquired 24 h after injection of 99mTc-labeled succinimidyl-hydrazinonicotinamide (99mTc-S-HYNIC)-conjugated anti-FAP antibody 28H1 at 2, 5, and 9 d after treatment. The uptake of 99mTc-S-HYNIC-28H1 in all joints was quantified and correlated with macroscopic arthritis scores. RESULTS Treatment of CIA with PLP-LCL significantly suppressed joint swelling. At just 1 d after treatment, the macroscopic arthritis scores had decreased by 50%. Scores decreased further, to only 10% of the initial scores, at 5 and 9 d after treatment. In contrast, macroscopic arthritis scores had increased up to 600% in untreated mice at 9 d after the injection of empty LCL. 99mTc-S-HYNIC-28H1 uptake ranged from 1.5 percentage injected dose per gram in noninflamed joints to 22.6 percentage injected dose per gram in severely inflamed joints. The uptake of radiolabeled 28H1 in inflamed joints (percentage injected dose) correlated with the arthritis score (Spearman ρ, 0.77; P < 0.0001). Moreover, the uptake of 99mTc-S-HYNIC-28H1 was slightly increased at 9 d after therapy but was not seen macroscopically, indicating that SPECT/CT imaging might be more sensitive than the macroscopic arthritis scoring method. CONCLUSION SPECT/CT imaging with 99mTc-S-HYNIC-28H1 specifically monitored the response to therapy, and tracer accumulation correlated with the severity of inflammation. In addition, SPECT/CT imaging was potentially more sensitive than the macroscopic arthritis scoring method. This study showed that SPECT/CT with 99mTc-S-HYNIC-28H1 could be used to noninvasively monitor the course of CIA in mice.
Collapse
Affiliation(s)
- Tessa van der Geest
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Laverman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Danny Gerrits
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Birgitte Walgreen
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Monique M Helsen
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christian Klein
- Roche Pharmaceutical Research and Early Development, Innovation Center Zurich, Schlieren, Switzerland
| | - Tapan K Nayak
- Roche Pharmaceutical Research and Early Development, Innovation Center Basel, Basel, Switzerland
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Department of Targeted Therapeutics, MIRA Institute, University of Twente, Zuidhorst, The Netherlands; and
| | - Josbert M Metselaar
- Department of Targeted Therapeutics, MIRA Institute, University of Twente, Zuidhorst, The Netherlands; and.,Department of Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Marije I Koenders
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
40
|
Kelley WJ, Safari H, Lopez-Cazares G, Eniola-Adefeso O. Vascular-targeted nanocarriers: design considerations and strategies for successful treatment of atherosclerosis and other vascular diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 8:909-926. [PMID: 27194461 DOI: 10.1002/wnan.1414] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 04/07/2016] [Accepted: 04/21/2016] [Indexed: 02/02/2023]
Abstract
Vascular-targeted nanocarriers are an attractive option for the treatment of a number of cardiovascular diseases, as they allow for more specific delivery and increased efficacy of many small molecule drugs. However, immune clearance, limited cellular uptake, and particle-cell dynamics in blood flow can hinder nanocarrier efficacy in many applications. This review aims to investigate successful strategies for the use of vascular-targeted nanocarriers in the treatment of cardiovascular diseases such as atherosclerosis. In particular, the review will highlight strategies employed for actively targeting the components of the atherosclerotic plaque, including endothelial cells, macrophages, and platelets and passive targeting via endothelial permeability, as well as design specifications (such as size, shape, and density) aimed at enhancing the ability of nanocarriers to reach the vascular wall. WIREs Nanomed Nanobiotechnol 2016, 8:909-926. doi: 10.1002/wnan.1414 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- William J Kelley
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Hanieh Safari
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | | |
Collapse
|
41
|
Surprising lack of liposome-induced complement activation by artificial 1,3-diamidophospholipids in vitro. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:845-849. [DOI: 10.1016/j.nano.2015.12.364] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 11/25/2015] [Accepted: 12/05/2015] [Indexed: 11/22/2022]
|
42
|
Yang M, Ding J, Zhang Y, Chang F, Wang J, Gao Z, Zhuang X, Chen X. Activated macrophage-targeted dextran-methotrexate/folate conjugate prevents deterioration of collagen-induced arthritis in mice. J Mater Chem B 2016; 4:2102-2113. [PMID: 32263177 DOI: 10.1039/c5tb02479j] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease, leading to articular synovial hyperplasia, cartilage destruction, and bone erosion. In RA pathophysiology, the activated macrophages contribute to the initiation and maintenance of the disease. Folate receptor, an overexpressed receptor on the activated macrophages, becomes a promising target site for RA treatment. In this work, the folate-modified dextran-methotrexate conjugate (noted as Dex-g-MTX/FA) was synthesized with an untargeted dextran-methotrexate prodrug (referred as Dex-g-MTX) as the control. The two prodrugs self-assembled into spherical micelles with both scales of about 90 nm and exhibited sustained MTX release. Dex-g-MTX/FA exhibited more superior cellular uptake mediated by the folate receptor and higher cytotoxicity toward macrophages activated by lipopolysaccharide (LPS) compared with Dex-g-MTX. Moreover, Dex-g-MTX/FA possessed improved biodistribution at the lesion site and stronger remission of RA through the inhibition of proinflammatory cytokines in comparison with both Dex-g-MTX and free MTX. These results demonstrated that the folate-targeted prodrug, i.e., Dex-g-MTX/FA, is a potential strategy for activated macrophage-targeted therapy of RA.
Collapse
Affiliation(s)
- Modi Yang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Karabey-Akyurek Y, Gurcay AG, Gurcan O, Turkoglu OF, Yabanoglu-Ciftci S, Eroglu H, Sargon MF, Bilensoy E, Oner L. Localized delivery of methylprednisolone sodium succinate with polymeric nanoparticles in experimental injured spinal cord model. Pharm Dev Technol 2016; 22:972-981. [PMID: 26895158 DOI: 10.3109/10837450.2016.1143002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
With important social and economic consequences, spinal cord injuries (SCIs) still exist among major health problems. Although many therapeutic agents and methods investigated for the treatment of acute SCI, only high dose methylprednisolone (MP) is being used currently in practice. Due to the serious side effects, high dose systemic MP administration after SCI is a critical issue that is mostly considered controversial. In our study, it is aimed to develop a nanoparticle-gel combined drug delivery system for localization of MP on trauma site and eliminating dose-dependent side effects by lowering the administered dose. For this purpose, methyl prednisolone sodium succinate (MPSS) loaded polycaprolactone based nanoparticles were developed and embedded in an implantable fibrin gel. The effects of MPSS delivery system are evaluated on an acute SCI rat model, by quantification the levels of three inflammatory cytokines (interleukin-1β, interleukin-6 and caspase-3) and assessment of the damage on ultrastructural level by transmission electron microscopy. Developed NP-gel system showed very similar results with systemic high dose of MPSS. It is believed that developed system may be used as a tool for the safe and effective localized delivery of several other therapeutic molecules on injured spinal cord cases.
Collapse
Affiliation(s)
- Yasemin Karabey-Akyurek
- a Department of Pharmaceutical Technology , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey
| | - Ahmet Gurhan Gurcay
- b Department of Neurosurgery , Ankara Ataturk Research & Education Hospital , Ankara , Turkey
| | - Oktay Gurcan
- b Department of Neurosurgery , Ankara Ataturk Research & Education Hospital , Ankara , Turkey
| | - Omer Faruk Turkoglu
- b Department of Neurosurgery , Ankara Ataturk Research & Education Hospital , Ankara , Turkey
| | - Samiye Yabanoglu-Ciftci
- c Department of Biochemistry , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey , and
| | - Hakan Eroglu
- a Department of Pharmaceutical Technology , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey
| | - Mustafa Fevzi Sargon
- d Department of Anatomy , Faculty of Medicine, Hacettepe University , Ankara , Turkey
| | - Erem Bilensoy
- a Department of Pharmaceutical Technology , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey
| | - Levent Oner
- a Department of Pharmaceutical Technology , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey
| |
Collapse
|
44
|
van der Meel R, Vehmeijer LJC, Kok RJ, Storm G, van Gaal EVB. Ligand-targeted Particulate Nanomedicines Undergoing Clinical Evaluation: Current Status. INTRACELLULAR DELIVERY III 2016. [DOI: 10.1007/978-3-319-43525-1_7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
45
|
Synthesis and Controlled Release Behavior of Biodegradable Polymers with Pendant Ibuprofen Group. INT J POLYM SCI 2016. [DOI: 10.1155/2016/5861419] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The continuous use of nonsteroidal anti-inflammatory drugs such as ibuprofen frequently leads to some serious side-effects including stomach ulcers and bleeding. In this paper, two kinds of new biocompatible polyesters (PIGB, PIGH) and polyester-amide (PIGA) comprising biodegradable components (L-glutamic acid,1,4-butanediol, and1,6-hexanediol and6-amino hexanol) and ibuprofen as pendant group have been prepared by the melting polycondensation. The chemical structures of the monomer and polymers are characterized by FTIR,1H NMR spectrum, GPC, and contact angle measurements. The drug loading of ibuprofen reaches very high level (35–37%) for PIGB, PIGH, and PIGA carriers. The free ibuprofen molecules are releasedin vitrofrom polymer carriers in a controlled manner without a burst release, different from the release pattern observed in the other drug-encapsulated systems. It is also found that the different hydrophilicity among PIGB, PIGH, and PIGA plays a key role in the time-controlled release of ibuprofen. In addition, the viability of HeLa cells after 48 h of incubation reaches more than 100%, indicating no cytotoxicity for PIGB, PIGH, and PIGA carriers.
Collapse
|
46
|
Lopes-de-Araújo J, Neves AR, Gouveia VM, Moura CC, Nunes C, Reis S. Oxaprozin-Loaded Lipid Nanoparticles towards Overcoming NSAIDs Side-Effects. Pharm Res 2015; 33:301-14. [PMID: 26350105 DOI: 10.1007/s11095-015-1788-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/31/2015] [Indexed: 01/15/2023]
Abstract
PURPOSE Nanostructured Lipid Carriers (NLCs) loading oxaprozin were developed to address an effective drug packaging and targeted delivery, improving the drug pharmacokinetics and pharmacodynamics properties and avoiding the local gastric side-effects. Macrophages actively phagocyte particles with sizes larger than 200 nm and, when activated, over-express folate beta receptors - features that in the case of this work constitute the basis for passive and active targeting strategies. METHODS Two formulations containing oxaprozin were developed: NLCs with and without folate functionalization. In order to target the macrophages folate receptors, a DSPE-PEG2000-FA conjugate was synthesized and added to the NLCs. RESULTS These formulations presented a relatively low polydispersity index (approximately 0.2) with mean diameters greater than 200 nm and zeta potential inferior to -40 mV. The encapsulation efficiency of the particles was superior to 95% and the loading capacity was of 9%, approximately. The formulations retained the oxaprozin release in simulated gastric fluid (only around 10%) promoting its release on simulated intestinal fluid. MTT and LDH assays revealed that the formulations only presented cytotoxicity in Caco-2 cells for oxaprozin concentrations superior to 100 μM. Permeability studies in Caco-2 cells shown that oxaprozin encapsulation did not interfered with oxaprozin permeability (around 0.8 × 10(-5) cm/s in simulated intestinal fluid and about 1.45 × 10(-5) cm/s in PBS). Moreover, in RAW 264.7 cells NLCs functionalization promoted an increased uptake over time mainly mediated by a caveolae uptake mechanism. CONCLUSIONS The developed nanoparticles enclose a great potential for oxaprozin oral administration with significant less gastric side-effects.
Collapse
Affiliation(s)
- José Lopes-de-Araújo
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Ana Rute Neves
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Virgínia M Gouveia
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Catarina C Moura
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Cláudia Nunes
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Salette Reis
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| |
Collapse
|
47
|
Medvedeva NV, Prozorovskiy VN, Ignatov DV, Druzilovskaya OS, Kudinov VA, Kasatkina EO, Tikhonova EG, Ipatova OM. Pharmacological agents and transport nanosystems based on plant phospholipids. BIOCHEMISTRY (MOSCOW) SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2015. [DOI: 10.1134/s199075081503004x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
48
|
Affiliation(s)
- Bhushan S Pattni
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States
| | - Vladimir V Chupin
- Laboratory for Advanced Studies of Membrane Proteins, Moscow Institute of Physics and Technology , Dolgoprudny 141700, Russia
| | - Vladimir P Torchilin
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States.,Department of Biochemistry, Faculty of Science, King Abdulaziz University , Jeddah 21589, Saudi Arabia
| |
Collapse
|
49
|
Gonçalves RM, Pereira ACL, Pereira IO, Oliveira MJ, Barbosa MA. Macrophage response to chitosan/poly-(γ-glutamic acid) nanoparticles carrying an anti-inflammatory drug. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:167. [PMID: 25791458 DOI: 10.1007/s10856-015-5496-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/14/2015] [Indexed: 06/04/2023]
Abstract
The inflammatory response to biomaterials, traditionally viewed as detrimental, is nowadays considered essential for tissue repair/regeneration, being macrophages recognized as the key players in resolving inflammation. Here, the preparation of chitosan (Ch)/poly-(γ-glutamic acid) (γ-PGA) nanoparticles (NPs) as vehicle for a non-steroid anti-inflammatory drug, diclofenac (Df), is described and the response of primary human macrophages to this system is evaluated. Df was incorporated in Ch/γ-PGA NPs at controlled pH (5.0) (maximum 0.05 mg/ml). The components molar ratio and order of addition revealed to be critical to obtain NPs (315 ± 50 nm with 0.36 ± 0.06 polydispersion index). Df was released at physiological pH and this drug-delivery system was proved to be non toxic to macrophages, being rapidly internalized (95 %). Importantly, efficacy of Df-NPs was confirmed by their ability of inhibit/revert PGE2 production of activated macrophages. Therefore, Df-NPs could contribute to stifle local inflammatory reactions, namely those associated with biomaterials.
Collapse
|
50
|
Rabenhold M, Steiniger F, Fahr A, Kontermann RE, Rüger R. Bispecific single-chain diabody-immunoliposomes targeting endoglin (CD105) and fibroblast activation protein (FAP) simultaneously. J Control Release 2015; 201:56-67. [DOI: 10.1016/j.jconrel.2015.01.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/16/2015] [Accepted: 01/20/2015] [Indexed: 01/09/2023]
|