1
|
Liu H, Ji M, Xiao P, Gou J, Yin T, He H, Tang X, Zhang Y. Glucocorticoids-based prodrug design: Current strategies and research progress. Asian J Pharm Sci 2024; 19:100922. [PMID: 38966286 PMCID: PMC11222810 DOI: 10.1016/j.ajps.2024.100922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/04/2024] [Accepted: 03/06/2024] [Indexed: 07/06/2024] Open
Abstract
Attributing to their broad pharmacological effects encompassing anti-inflammation, antitoxin, and immunosuppression, glucocorticoids (GCs) are extensively utilized in the clinic for the treatment of diverse diseases such as lupus erythematosus, nephritis, arthritis, ulcerative colitis, asthma, keratitis, macular edema, and leukemia. However, long-term use often causes undesirable side effects, including metabolic disorders-induced Cushing's syndrome (buffalo back, full moon face, hyperglycemia, etc.), osteoporosis, aggravated infection, psychosis, glaucoma, and cataract. These notorious side effects seriously compromise patients' quality of life, especially in patients with chronic diseases. Therefore, glucocorticoid-based advanced drug delivery systems for reducing adverse effects have received extensive attention. Among them, prodrugs have the advantages of low investment, low risk, and high success rate, making them a promising strategy. In this review, we propose the strategies for the design and summarize current research progress of glucocorticoid-based prodrugs in recent decades, including polymer-based prodrugs, dendrimer-based prodrugs, antibody-drug conjugates, peptide-drug conjugates, carbohydrate-based prodrugs, aliphatic acid-based prodrugs and so on. Besides, we also raise issues that need to be focused on during the development of glucocorticoid-based prodrugs. This review is expected to be helpful for the research and development of novel GCs and prodrugs.
Collapse
Affiliation(s)
- Hongbing Liu
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Muse Ji
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Peifu Xiao
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingxin Gou
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tian Yin
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haibing He
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xing Tang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Zhang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
2
|
Bapat P, Paul S, Tseng YC, Taylor LS. Interplay of Drug-Polymer Interactions and Release Performance for HPMCAS-Based Amorphous Solid Dispersions. Mol Pharm 2024; 21:1466-1478. [PMID: 38346390 DOI: 10.1021/acs.molpharmaceut.3c01106] [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] [Indexed: 03/05/2024]
Abstract
The interplay between drug and polymer chemistry and its impact on drug release from an amorphous solid dispersion (ASD) is a relatively underexplored area. Herein, the release rates of several drugs of diverse chemistry from hydroxypropyl methylcellulose acetate succinate (HPMCAS)-based ASDs were explored using surface area normalized dissolution. The tendency of the drug to form an insoluble complex with HPMCAS was determined through coprecipitation experiments. The role of pH and the extent of drug ionization were probed to evaluate the role of electrostatic interactions in complex formation. Relationships between the extent of complexation and the drug release rate from an ASD were observed, whereby the drugs could be divided into two groups. Drugs with a low extent of insoluble complex formation with HPMCAS tended to be neutral or anionic and showed reasonable release at pH 6.8 even at higher drug loadings. Cationic drugs formed insoluble complexes with HPMCAS and showed poor release when formulated as an ASD. Thus, and somewhat counterintuitively, a weakly basic drug showed a reduced release rate from an ASD at a bulk solution pH where it was ionized, relative to when unionized. The opposite trend was observed in the absence of polymer for the neat amorphous drug. In conclusion, electrostatic interactions between HPMCAS and lipophilic cationic drugs led to insoluble complex formation, which in turn resulted in ASDs with poor release performance.
Collapse
Affiliation(s)
- Pradnya Bapat
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Shubhajit Paul
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Yin-Chao Tseng
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
3
|
Dimiou S, McCabe J, Booth R, Booth J, Nidadavole K, Svensson O, Sparén A, Lindfors L, Paraskevopoulou V, Mead H, Coates L, Workman D, Martin D, Treacher K, Puri S, Taylor LS, Yang B. Selecting Counterions to Improve Ionized Hydrophilic Drug Encapsulation in Polymeric Nanoparticles. Mol Pharm 2023; 20:1138-1155. [PMID: 36653946 DOI: 10.1021/acs.molpharmaceut.2c00855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hydrophobic ion pairing (HIP) can successfully increase the drug loading and control the release kinetics of ionizable hydrophilic drugs, addressing challenges that prevent these molecules from reaching the clinic. Nevertheless, polymeric nanoparticle (PNP) formulation development requires trial-and-error experimentation to meet the target product profile, which is laborious and costly. Herein, we design a preformulation framework (solid-state screening, computational approach, and solubility in PNP-forming emulsion) to understand counterion-drug-polymer interactions and accelerate the PNP formulation development for HIP systems. The HIP interactions between a small hydrophilic molecule, AZD2811, and counterions with different molecular structures were investigated. Cyclic counterions formed amorphous ion pairs with AZD2811; the 0.7 pamoic acid/1.0 AZD2811 complex had the highest glass transition temperature (Tg; 162 °C) and the greatest drug loading (22%) and remained as phase-separated amorphous nanosized domains inside the polymer matrix. Palmitic acid (linear counterion) showed negligible interactions with AZD2811 (crystalline-free drug/counterion forms), leading to a significantly lower drug loading despite having similar log P and pKa with pamoic acid. Computational calculations illustrated that cyclic counterions interact more strongly with AZD2811 than linear counterions through dispersive interactions (offset π-π interactions). Solubility data indicated that the pamoic acid/AZD2811 complex has a lower organic phase solubility than AZD2811-free base; hence, it may be expected to precipitate more rapidly in the nanodroplets, thus increasing drug loading. Our work provides a generalizable preformulation framework, complementing traditional performance-indicating parameters, to identify optimal counterions rapidly and accelerate the development of hydrophilic drug PNP formulations while achieving high drug loading without laborious trial-and-error experimentation.
Collapse
Affiliation(s)
- Savvas Dimiou
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D AstraZeneca, Granta Park, CambridgeCB21 6GH, U.K.,UCL School of Pharmacy, 29-39 Brunswick Square, LondonWC1N 1AX, U.K
| | - James McCabe
- Early Product Development, Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Rebecca Booth
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Jonathan Booth
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Kalyan Nidadavole
- Early Product Development, Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Olof Svensson
- Pharmaceutical Technology & Development, Operations, AstraZeneca, GothenburgSE-43183, Sweden
| | - Anders Sparén
- Pharmaceutical Technology & Development, Operations, AstraZeneca, GothenburgSE-43183, Sweden
| | - Lennart Lindfors
- Advanced Drug Delivery, Pharmaceutical Science, R&D AstraZeneca, GothenburgSE-43183, Sweden
| | - Vasiliki Paraskevopoulou
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Heather Mead
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Lydia Coates
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - David Workman
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D AstraZeneca, Granta Park, CambridgeCB21 6GH, U.K
| | - Dave Martin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Kevin Treacher
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Sanyogitta Puri
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D AstraZeneca, Granta Park, CambridgeCB21 6GH, U.K
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana47907, United States
| | - Bin Yang
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D AstraZeneca, Granta Park, CambridgeCB21 6GH, U.K
| |
Collapse
|
4
|
Bhilare NV, Marulkar VS, Shirote PJ, Dombe SA, Pise VJ, Salve PL, Biradar SM, Yadav VD, Jadhav PD, Bodhe AA, Borkar SP, Ghadge PM, Shelar PA, Jadhav AV, Godse KC. Mannich Bases: Centrality in Cytotoxic Drug Design. Med Chem 2021; 18:735-756. [PMID: 34931967 DOI: 10.2174/1573406418666211220124119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/04/2021] [Accepted: 10/18/2021] [Indexed: 11/22/2022]
Abstract
Mannich bases identified by Professor Carl Mannich have been the most extensively explored scaffolds for more than 100 years now. The versatile biological roles that they play have promoted their applications in many clinical conditions. The present review highlights the application of Mannich bases as cytotoxic agents, categorizing them into synthetic, semisynthetic and prodrugs classes and gives an exhaustive account of the work reported in the last two decades. The methods of synthesis of these cytotoxic agents, their anti-cancer potential in various cell lines and promising leads for future drug development have also been discussed. Structure-activity relationships along with the targets on which these cytotoxic Mannich bases act have been included as well.
Collapse
Affiliation(s)
- Neha V Bhilare
- Department of Pharmaceutical Chemistry, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Vinayak S Marulkar
- Department of Pharmaceutical Chemistry, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Pramodkumar J Shirote
- Department of Pharmaceutical Chemistry, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Shailaja A Dombe
- Department of Pharmaceutics, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Vilas J Pise
- Department of Pharmaceutical Chemistry, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Pallavi L Salve
- Department of Pharmaceutical Chemistry, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Shantakumar M Biradar
- Department of Pharmaceutical Chemistry, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Vishal D Yadav
- Department of Pharmaceutics, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Prakash D Jadhav
- Department of Pharmaceutics, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Anjali A Bodhe
- Department of Pharmaceutics, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Smita P Borkar
- Department of Pharmaceutics, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Prachi M Ghadge
- Department of Pharmacology, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Pournima A Shelar
- Department of Pharmaceutics, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Apurva V Jadhav
- Department of Pharmaceutics, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| | - Kirti C Godse
- Department of Pharmacology, Arvind Gavali College of Pharmacy, Satara-415004, Maharashtra, India
| |
Collapse
|
5
|
Molinier C, Picot-Groz M, Malval O, Le Lamer-Déchamps S, Richard J, Lopez-Noriega A, Grizot S. Impact of octreotide counterion nature on the long-term stability and release kinetics from an in situ forming depot technology. J Control Release 2021; 336:457-468. [PMID: 34214596 DOI: 10.1016/j.jconrel.2021.06.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/17/2021] [Accepted: 06/27/2021] [Indexed: 01/03/2023]
Abstract
The generation of acylated impurities has represented an important hurdle in the development of long acting injectables for therapeutic peptides using biocompatible polymers with a polyester moiety. We investigated here an in situ forming depot (ISFD) technology that uses polyethylene glycol - polyester copolymers and a solvent exchange mechanism to promote depot formation. This technology has shown promise in formulating small molecules as well as therapeutic proteins. In the present work, using the well-known somatostatin analog octreotide acetate (OctAc) as a model molecule, we evaluated this delivery platform to release therapeutic peptides. Peptide acylation was found to be pronounced in the formulation, while it was very limited once the depot was formed and during the release process. The octreotide acylation pattern was fully characterized by LC-MS/MS. Moreover, it was demonstrated that exchanging the acetate anion with more hydrophobic counterions like pamoate or lauryl sulfate allowed to greatly improve the peptide stability profile, as well as the formulation release performance. Finally, the in vivo evaluation through pharmacokinetics studies in rat of these new octreotide salts in ISFD formulations showed that octreotide was quantifiable up to four weeks post-administration with a high bioavailability and an acceptable initial burst.
Collapse
Affiliation(s)
| | | | - Océane Malval
- MedinCell, 3 Rue des Frères Lumière, 34830 Jacou, France
| | | | - Joël Richard
- MedinCell, 3 Rue des Frères Lumière, 34830 Jacou, France
| | | | | |
Collapse
|
6
|
Ribeiro SB, de Araújo AA, Oliveira MMB, dos Santos Silva AM, da Silva-Júnior AA, Guerra GCB, Brito GADC, Leitão RFDC, de Araújo Júnior RF, Garcia VB, Vasconcelos RC, de Medeiros CACX. Effect of Dexamethasone-Loaded PLGA Nanoparticles on Oral Mucositis Induced by 5-Fluorouracil. Pharmaceutics 2021; 13:53. [PMID: 33406583 PMCID: PMC7823510 DOI: 10.3390/pharmaceutics13010053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/23/2020] [Accepted: 12/01/2020] [Indexed: 01/06/2023] Open
Abstract
Oral mucositis (OM) is characterized by the presence of severe ulcers in the oral region that affects patients treated with chemotherapy. It occurs in almost all patients who receive radiotherapy of the head and neck, as well as patients who undergo hematopoietic cell transplantation. The pathophysiology of OM is complex, and there is no effective therapy. The aim of this study was to evaluate the effect of dexamethasone-loaded poly(d,l-Lactic-co-glycolic) nanoparticles (PLGA-DEX NPs) on an OM model induced in hamsters. The NPs were synthesized using the emulsification-solvent evaporation method and were characterized by the size, zeta potential, encapsulation efficiency, atomic force microscopy, physicochemical stability, and the in vitro release. The OM was induced by the administration of 5-FU on the first and second days and mechanical trauma on the 4th day of the experiment. PLGA-DEX NPs were administered to treat OM. The animals were euthanized on the 10th day. Macroscopic and histopathological analyses were performed, measurement of malonaldehyde (MDA) and ELISA was used to determine the levels of IL-1β and TNF-α. Immunoexpressions of NF-κB, COX-2, and TGF-β were determined by immunohistochemistry, and qRT-PCR was used to quantify the gene expression of the GILZ, MKP1, and NF-κB p65. The PLGA-DEX NPs (0.1 mg/kg) significantly reduced macroscopic and histopathological scores, decreased MDA, TNF-α and IL-1β levels, immunostaining for NF-κB, COX-2, TGF-β, and suppressed NF-κB p65 mRNA expression, but increased GILZ and MKP1 expression.
Collapse
Affiliation(s)
- Susana Barbosa Ribeiro
- Post Graduate Program Biotechnology-RENORBIO, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil; (S.B.R.); (M.M.B.O.)
| | - Aurigena Antunes de Araújo
- Post Graduate Program Dental Sciences, Post Graduate Program Pharmaceutical Science, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil;
| | - Maisie Mitchele Barbosa Oliveira
- Post Graduate Program Biotechnology-RENORBIO, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil; (S.B.R.); (M.M.B.O.)
| | - Alaine Maria dos Santos Silva
- Laboratory of Pharmaceutical Technology & Biotechnology (TecBioFar), Post Graduate Program Pharmaceutical Sciences, Pharmacy Department, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Faria St, Petrópolis, Natal RN 59012-570, Brazil; (A.M.d.S.S.); (A.A.d.S.-J.)
| | - Arnóbio Antônio da Silva-Júnior
- Laboratory of Pharmaceutical Technology & Biotechnology (TecBioFar), Post Graduate Program Pharmaceutical Sciences, Pharmacy Department, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Faria St, Petrópolis, Natal RN 59012-570, Brazil; (A.M.d.S.S.); (A.A.d.S.-J.)
| | - Gerlane Coelho Bernardo Guerra
- Post Graduate Program Biochemistry and Molecular Biology, Post Graduate Program Pharmaceutical Science, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil;
| | - Gerly Anne de Castro Brito
- Post Graduate Program Morphofunctional Sciences, Department of Morphology, Faculty of Medicine, Federal University of Ceará, Delmiro de Farias St, Rodolfo Teófilo, Fortaleza CE 60416-030, Brazil; (G.A.d.C.B.); (R.F.d.C.L.)
| | - Renata Ferreira de Carvalho Leitão
- Post Graduate Program Morphofunctional Sciences, Department of Morphology, Faculty of Medicine, Federal University of Ceará, Delmiro de Farias St, Rodolfo Teófilo, Fortaleza CE 60416-030, Brazil; (G.A.d.C.B.); (R.F.d.C.L.)
| | - Raimundo Fernandes de Araújo Júnior
- Post Graduate Program Functional and Structural Biology, Post Graduate Program Health Science, Department of Morphology, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil;
| | - Vinícius Barreto Garcia
- Post Graduate Program Health Science, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Faria St, Petrópolis, Natal RN 59012-570, Brazil;
| | | | - Caroline Addison Carvalho Xavier de Medeiros
- Post Graduate Program Biotechnology-RENORBIO, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil; (S.B.R.); (M.M.B.O.)
- Post Graduate Program Biochemistry and Molecular Biology, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil
| |
Collapse
|
7
|
Ristroph K, Salim M, Wilson BK, Clulow AJ, Boyd BJ, Prud'homme RK. Internal liquid crystal structures in nanocarriers containing drug hydrophobic ion pairs dictate drug release. J Colloid Interface Sci 2021; 582:815-824. [DOI: 10.1016/j.jcis.2020.08.045] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/28/2022]
|
8
|
Ristroph KD, Prud'homme RK. Hydrophobic ion pairing: encapsulating small molecules, peptides, and proteins into nanocarriers. NANOSCALE ADVANCES 2019; 1:4207-4237. [PMID: 33442667 PMCID: PMC7771517 DOI: 10.1039/c9na00308h] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/18/2019] [Indexed: 05/26/2023]
Abstract
Hydrophobic ion pairing has emerged as a method to modulate the solubility of charged hydrophilic molecules ranging in class from small molecules to large enzymes. Charged hydrophilic molecules are ionically paired with oppositely-charged molecules that include hydrophobic moieties; the resulting uncharged complex is water-insoluble and will precipitate in aqueous media. Here we review one of the most prominent applications of hydrophobic ion pairing: efficient encapsulation of charged hydrophilic molecules into nano-scale delivery vehicles - nanoparticles or nanocarriers. Hydrophobic complexes are formed and then encapsulated using techniques developed for poorly-water-soluble therapeutics. With this approach, researchers have reported encapsulation efficiencies up to 100% and drug loadings up to 30%. This review covers the fundamentals of hydrophobic ion pairing, including nomenclature, drug eligibility for the technique, commonly-used counterions, and drug release of encapsulated ion paired complexes. We then focus on nanoformulation techniques used in concert with hydrophobic ion pairing and note strengths and weaknesses specific to each. The penultimate section bridges hydrophobic ion pairing with the related fields of polyelectrolyte coacervation and polyelectrolyte-surfactant complexation. We then discuss the state of the art and anticipated future challenges. The review ends with comprehensive tables of reported hydrophobic ion pairing and encapsulation from the literature.
Collapse
Affiliation(s)
- Kurt D. Ristroph
- Department of Chemical and Biological Engineering, Princeton UniversityPrincetonNew Jersey 08544USA
| | - Robert K. Prud'homme
- Department of Chemical and Biological Engineering, Princeton UniversityPrincetonNew Jersey 08544USA
| |
Collapse
|
9
|
Formulation of Antimicrobial Tobramycin Loaded PLGA Nanoparticles via Complexation with AOT. J Funct Biomater 2019; 10:jfb10020026. [PMID: 31200522 PMCID: PMC6617385 DOI: 10.3390/jfb10020026] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 05/22/2019] [Accepted: 06/10/2019] [Indexed: 01/01/2023] Open
Abstract
Tobramycin is a potent antimicrobial aminoglycoside and its effective delivery by encapsulation within nanoparticle carriers could increase its activity against infections through a combination of sustained release and enhanced uptake. Effective antimicrobial therapy against a clinically relevant model bacteria (Pseudomonas aeruginosa) requires sufficient levels of therapeutic drug to maintain a drug concentration above the microbial inhibitory concentration (MIC) of the bacteria. Previous studies have shown that loading of aminoglycoside drugs in poly(lactic-co-glycolic) acid (PLGA)-based delivery systems is generally poor due to weak interactions between the drug and the polymer. The formation of complexes of tobramycin with dioctylsulfosuccinate (AOT) allows the effective loading of the drug in PLGA-nanoparticles and such nanoparticles can effectively deliver the antimicrobial aminoglycoside with retention of tobramycin antibacterial function.
Collapse
|
10
|
Zhou Z, Kennell C, Jafari M, Lee JY, Ruiz-Torres SJ, Waltz SE, Lee JH. Sequential delivery of erlotinib and doxorubicin for enhanced triple negative Breast cancer treatment using polymeric nanoparticle. Int J Pharm 2017; 530:300-307. [PMID: 28778627 DOI: 10.1016/j.ijpharm.2017.07.085] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/13/2017] [Accepted: 07/31/2017] [Indexed: 01/25/2023]
Abstract
Recent studies of signaling networks point out that an order of drugs to be administrated to the cancerous cells can be critical for optimal therapeutic outcomes of recalcitrant metastatic and drug-resistant cell types. In this study, a development of a polymeric nanoparticle system for sequential delivery is reported. The nanoparticle system can co-encapsulate and co-deliver a combination of therapeutic agents with different physicochemical properties [i.e. epidermal growth factor receptor (EGFR) inhibitor, erlotinib (Ei), and doxorubicin (Dox)]. Dox is hydrophilic and was complexed with anionic lipid, 1,2-dioleoyl-sn-glycero-3-phosphate (DOPA), via ion pairing to form a hydrophobic entity. Then it was co-encapsulated with hydrophobic Ei in a poly(L-lactide)-b-polyethylene glycol (PLA-b-PEG) nanoparticle by nanoprecipitation. The complexation of Dox with DOPA greatly helps the encapsulation of Dox, and substantially reduces the release rate of Dox. This nanoparticle system was found to burst the release of Ei with a slow and sustained profile of Dox, which is an optimal course of administration for these two drugs as previously reported. The efficacy of this sequential delivery nanoparticle system was validated in vitro and its in vivo potential applicability was substantiated by fluorescent imaging of high tumor accumulation.
Collapse
Affiliation(s)
- Zilan Zhou
- Chemical Engineering Program, Department of Biomedical, Environmental, and Chemical Engineering, University of Cincinnati, Cincinnati, OH 45221-0012, USA
| | - Carly Kennell
- Chemical Engineering Program, Department of Biomedical, Environmental, and Chemical Engineering, University of Cincinnati, Cincinnati, OH 45221-0012, USA
| | - Mina Jafari
- Chemical Engineering Program, Department of Biomedical, Environmental, and Chemical Engineering, University of Cincinnati, Cincinnati, OH 45221-0012, USA
| | - Joo-Youp Lee
- Chemical Engineering Program, Department of Biomedical, Environmental, and Chemical Engineering, University of Cincinnati, Cincinnati, OH 45221-0012, USA.
| | - Sasha J Ruiz-Torres
- Department of Cancer Biology, College of Medicine University of Cincinnati, Cincinnati, OH 45267-0521, USA
| | - Susan E Waltz
- Department of Cancer Biology, College of Medicine University of Cincinnati, Cincinnati, OH 45267-0521, USA; Research Service, Cincinnati Veteran Hospital Medical Center, Cincinnati, OH 45267-0521, USA
| | - Jing-Huei Lee
- Biomedical Engineering Program, Department of Biomedical, Environmental, and Chemical Engineering, University of Cincinnati, Cincinnati, OH 45221-0012, USA
| |
Collapse
|
11
|
Ashton S, Song YH, Nolan J, Cadogan E, Murray J, Odedra R, Foster J, Hall PA, Low S, Taylor P, Ellston R, Polanska UM, Wilson J, Howes C, Smith A, Goodwin RJA, Swales JG, Strittmatter N, Takáts Z, Nilsson A, Andren P, Trueman D, Walker M, Reimer CL, Troiano G, Parsons D, De Witt D, Ashford M, Hrkach J, Zale S, Jewsbury PJ, Barry ST. Aurora kinase inhibitor nanoparticles target tumors with favorable therapeutic index in vivo. Sci Transl Med 2016; 8:325ra17. [PMID: 26865565 DOI: 10.1126/scitranslmed.aad2355] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Efforts to apply nanotechnology in cancer have focused almost exclusively on the delivery of cytotoxic drugs to improve therapeutic index. There has been little consideration of molecularly targeted agents, in particular kinase inhibitors, which can also present considerable therapeutic index limitations. We describe the development of Accurin polymeric nanoparticles that encapsulate the clinical candidate AZD2811, an Aurora B kinase inhibitor, using an ion pairing approach. Accurins increase biodistribution to tumor sites and provide extended release of encapsulated drug payloads. AZD2811 nanoparticles containing pharmaceutically acceptable organic acids as ion pairing agents displayed continuous drug release for more than 1 week in vitro and a corresponding extended pharmacodynamic reduction of tumor phosphorylated histone H3 levels in vivo for up to 96 hours after a single administration. A specific AZD2811 nanoparticle formulation profile showed accumulation and retention in tumors with minimal impact on bone marrow pathology, and resulted in lower toxicity and increased efficacy in multiple tumor models at half the dose intensity of AZD1152, a water-soluble prodrug of AZD2811. These studies demonstrate that AZD2811 can be formulated in nanoparticles using ion pairing agents to give improved efficacy and tolerability in preclinical models with less frequent dosing. Accurins specifically, and nanotechnology in general, can increase the therapeutic index of molecularly targeted agents, including kinase inhibitors targeting cell cycle and oncogenic signal transduction pathways, which have to date proved toxic in humans.
Collapse
Affiliation(s)
- Susan Ashton
- Oncology iMED, AstraZeneca, Macclesfield, Cheshire SK10 4TG, UK
| | - Young Ho Song
- BIND Therapeutics, 325 Vassar Street, Cambridge, MA 02139, USA
| | - Jim Nolan
- BIND Therapeutics, 325 Vassar Street, Cambridge, MA 02139, USA
| | - Elaine Cadogan
- Oncology iMED, AstraZeneca, Macclesfield, Cheshire SK10 4TG, UK
| | - Jim Murray
- Pharmaceutical Development, AstraZeneca, Macclesfield, Cheshire SK10 2NX, UK
| | - Rajesh Odedra
- Oncology iMED, AstraZeneca, Macclesfield, Cheshire SK10 4TG, UK
| | - John Foster
- Drug Safety and Metabolism, Innovative Medicines, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - Peter A Hall
- Drug Safety and Metabolism, Innovative Medicines, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - Susan Low
- BIND Therapeutics, 325 Vassar Street, Cambridge, MA 02139, USA
| | - Paula Taylor
- Oncology iMED, AstraZeneca, Macclesfield, Cheshire SK10 4TG, UK
| | - Rebecca Ellston
- Oncology iMED, AstraZeneca, Macclesfield, Cheshire SK10 4TG, UK
| | | | - Joanne Wilson
- Oncology iMED, AstraZeneca, Macclesfield, Cheshire SK10 4TG, UK
| | - Colin Howes
- Oncology iMED, AstraZeneca, Macclesfield, Cheshire SK10 4TG, UK
| | - Aaron Smith
- Oncology iMED, AstraZeneca, Macclesfield, Cheshire SK10 4TG, UK
| | - Richard J A Goodwin
- Drug Safety and Metabolism, Innovative Medicines, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - John G Swales
- Drug Safety and Metabolism, Innovative Medicines, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | | | - Zoltán Takáts
- Department of Surgery and Cancer, Imperial College, London SW7 2AZ, UK
| | - Anna Nilsson
- Biomolecular Imaging and Proteomics, National Center for Mass Spectrometry Imaging, Uppsala University, Uppsala 751 05, Sweden
| | - Per Andren
- Biomolecular Imaging and Proteomics, National Center for Mass Spectrometry Imaging, Uppsala University, Uppsala 751 05, Sweden
| | - Dawn Trueman
- Oncology iMED, AstraZeneca, Macclesfield, Cheshire SK10 4TG, UK
| | - Mike Walker
- Oncology iMED, AstraZeneca, Macclesfield, Cheshire SK10 4TG, UK
| | - Corinne L Reimer
- Oncology iMED, AstraZeneca, Gatehouse Park, Waltham, Boston 02451, USA
| | - Greg Troiano
- BIND Therapeutics, 325 Vassar Street, Cambridge, MA 02139, USA
| | - Donald Parsons
- BIND Therapeutics, 325 Vassar Street, Cambridge, MA 02139, USA
| | - David De Witt
- BIND Therapeutics, 325 Vassar Street, Cambridge, MA 02139, USA
| | - Marianne Ashford
- Pharmaceutical Development, AstraZeneca, Macclesfield, Cheshire SK10 2NX, UK
| | - Jeff Hrkach
- BIND Therapeutics, 325 Vassar Street, Cambridge, MA 02139, USA
| | - Stephen Zale
- BIND Therapeutics, 325 Vassar Street, Cambridge, MA 02139, USA.
| | | | - Simon T Barry
- Oncology iMED, AstraZeneca, Macclesfield, Cheshire SK10 4TG, UK.
| |
Collapse
|
12
|
A novel in situ hydrophobic ion pairing (HIP) formulation strategy for clinical product selection of a nanoparticle drug delivery system. J Control Release 2016; 229:106-119. [DOI: 10.1016/j.jconrel.2016.03.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/23/2016] [Accepted: 03/16/2016] [Indexed: 11/18/2022]
|
13
|
Vaishya RD, Gokulgandhi M, Patel S, Minocha M, Mitra AK. Novel dexamethasone-loaded nanomicelles for the intermediate and posterior segment uveitis. AAPS PharmSciTech 2014; 15:1238-51. [PMID: 24895075 DOI: 10.1208/s12249-014-0100-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 02/23/2014] [Indexed: 11/30/2022] Open
Abstract
Development and characterization of dexamethasone (DEX)-encapsulated polymeric nanomicelles have been reported. A low molecular weight di-block copolymer was synthesized and characterized for its structure, molecular weights, critical micelle concentration (CMC), and cytotoxicity in ocular cells. In order to delineate the effects of drug-polymer interactions on drug solubilization in micelle core, a response surface methodology was generated with the help of SAS 9.02 (exploratory model). The method for preparing micelle was modified based on the results obtained from exploratory model. The formulation was optimized by response surface methodology (optimization model) to achieve DEX solubility of above 1 mg/mL. The optimized formulation was characterized for DEX solubility, nanomicelle size, polydispersity index, surface morphology, in vitro transport across conjunctival cell line, and ex vivo transport across excised rabbit sclera. Nanomicelles exhibited average sizes in range of 25-30 nm with unimodel size distribution and low polydispersity of 0.125. Nanomicelles increased DEX permeability by 2 times across conjunctival cell line and by 2.5 times across the excised rabbit sclera as compared to DEX suspension. A design of experiment (DOE) strategy was successfully applied to understand the effects of drug-polymer interaction on drug solubility. DOE was also employed to achieve optimal formulation with high DEX solubility. Nanomicellar formulation significantly enhanced DEX permeability across the excised rabbit sclera. Therefore, nanomicellar formulation may provide therapeutic levels in the back of the eye following topical administration.
Collapse
|
14
|
Abstract
INTRODUCTION Proteins are effective biotherapeutics with applications in diverse ailments. Despite being specific and potent, their full clinical potential has not yet been realized. This can be attributed to short half-lives, complex structures, poor in vivo stability, low permeability, frequent parenteral administrations and poor adherence to treatment in chronic diseases. A sustained release system, providing controlled release of proteins, may overcome many of these limitations. AREAS COVERED This review focuses on recent development in approaches, especially polymer-based formulations, which can provide therapeutic levels of proteins over extended periods. Advances in particulate, gel-based formulations and novel approaches for extended protein delivery are discussed. Emphasis is placed on dosage form, method of preparation, mechanism of release and stability of biotherapeutics. EXPERT OPINION Substantial advancements have been made in the field of extended protein delivery via various polymer-based formulations over last decade despite the unique delivery-related challenges posed by protein biologics. A number of injectable sustained-release formulations have reached market. However, therapeutic application of proteins is still hampered by delivery-related issues. A large number of protein molecules are under clinical trials, and hence, there is an urgent need to develop new methods to deliver these highly potent biologics.
Collapse
Affiliation(s)
- Ravi Vaishya
- University of Missouri-Kansas City, Pharmaceutical Sciences , Kansas City, MO , USA
| | | | | | | |
Collapse
|
15
|
Pignatello R, Leonardi A, Petronio GP, Ruozi B, Puglisi G, Furneri PM. Preparation and Microbiological Evaluation of Amphiphilic Kanamycin-Lipoamino Acid Ion-Pairs. Antibiotics (Basel) 2014; 3:216-32. [PMID: 27025745 PMCID: PMC4790386 DOI: 10.3390/antibiotics3020216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/29/2014] [Accepted: 05/14/2014] [Indexed: 11/19/2022] Open
Abstract
Amphiphilic ion-pairs of kanamycin (KAN) were prepared by evaporation of a water-ethanol co-solution of KAN base and a lipoamino acid bearing a 12-carbon atoms alkyl side chain (LAA12), at different molar ratios. Infrared spectroscopy confirmed the structure of ion-pairs, while differential scanning calorimetry (DSC) and powder X-ray diffractometry (PXRD) studies supported the formation of new saline species with a different crystalline structure than the starting components. The solubility pattern shown in a range of both aqueous and organic solvents confirmed that the ion-pairs possess an amphiphilic character. The LAA12 counter-ion showed not to improve the antibacterial activity of KAN, suggesting that such chemical strategy is not able to favor the penetration of this drug inside the bacteria cells. Nevertheless, a slight improving, i.e., a one-fold dilution, was observed in E. coli. The present study can also serve as the basis for a further evaluation of LAA ion-pairing of antibiotics, as a means to improve the loading of hydrophilic drugs into lipid-based nanocarriers.
Collapse
Affiliation(s)
- Rosario Pignatello
- Department of Drug Sciences, University of Catania, Città Universitaria, viale A. Doria 6, I-95125 Catania, Italy.
- NANO-i, Research Centre for Ocular Nanotechnology, Department of Drug Sciences, University of Catania, viale A. Doria 6, I-95125 Catania, Italy.
| | - Antonio Leonardi
- Department of Drug Sciences, University of Catania, Città Universitaria, viale A. Doria 6, I-95125 Catania, Italy.
| | - Giulio Petronio Petronio
- Department of Biomedical Sciences, University of Catania, via Androne 83, I-95124 Catania, Italy.
- IRCCS San Raffaele Pisana, Via della Pisana 235, I-00163 Roma, Italy.
| | - Barbara Ruozi
- Pharmaceutical Technology, Te.Far.T.I. group, Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 183, I-41100 Modena, Italy.
| | - Giovanni Puglisi
- Department of Drug Sciences, University of Catania, Città Universitaria, viale A. Doria 6, I-95125 Catania, Italy.
- NANO-i, Research Centre for Ocular Nanotechnology, Department of Drug Sciences, University of Catania, viale A. Doria 6, I-95125 Catania, Italy.
| | - Pio Maria Furneri
- Department of Biomedical Sciences, University of Catania, via Androne 83, I-95124 Catania, Italy.
| |
Collapse
|
16
|
Patel A, Gaudana R, Mitra AK. A novel approach for antibody nanocarriers development through hydrophobic ion-pairing complexation. J Microencapsul 2014; 31:542-50. [PMID: 24697179 DOI: 10.3109/02652048.2014.885606] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
IgG-Fab fragment, a model antibody protein was hydrophobically modified by a novel approach of ion-pairing complexation. Three different sulphated ion-pairing agents were utilised including sodium dodecyl sulphate, taurocholic acid and dextran sulphate (DS). The formations of hydrophobic ion-pairing (HIP) complexes were dependant on pH and molar ratio of ion-pairing agent to Fab. Aqueous solubilities of HIP complexes were very low compared to Fab alone. In particular, when dextran sulphate was added as ion-pairing agent, formed Fab:DS HIP complexes were least soluble in water. Further, nanoparticles (NPs) loaded with drug and Fab:DS HIP complex were prepared and characterised with respect to encapsulation efficiency and size. We observed significant improvement in encapsulation efficiency for Fab:DS HIP complex-loaded nanoparticles. This study demonstrates a novel approach of formulating antibody-loaded nanoparticles which can also be employed for delivery of large antibodies.
Collapse
Affiliation(s)
- Ashaben Patel
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City , Kansas City, MO , USA
| | | | | |
Collapse
|
17
|
He Y, Liu H, Xie Z, Liao Q, Lai X, Du Z. PVP and surfactant combined carrier as an effective absorption enhancer of poorly soluble astilbin in vitro and in vivo. Drug Dev Ind Pharm 2013; 40:237-43. [PMID: 23350723 DOI: 10.3109/03639045.2012.756008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Astilbin is considered to be a new and promising immunosuppressant for immune related diseases, but limited in clinical application due to its poor water solubility, difficult oral absorption and low bioavailability. OBJECTIVE The present work studied the effect of PVP and surfactant combined carrier on its capability to improve drug absorption. MATERIALS AND METHODS PVP K30-Tween 80 combined carries was applied into the astilbin solid dispersions, tested both in vivo in beagle dogs and in vitro in transport experiments across Caco-2 cell monolayers. RESULTS AND DISCUSSION In the animal studies a many fold increase in plasma AUC was observed for the solid dispersions of drug in PVP K30-Tween 80 combined carries compared to active pharmaceutical ingredient (API). The applicability of Caco-2 monolayers as a tool for predicting the in vivo transport behavior of Astilbin in combination with a solubility enhancing carries was shown. In vitro transport studies confirmed the effect of combined carries on the absorption behavior of the astilbin. MTT studies showed the cell viability gradually decreased with the increase of the drug concentration in a dose dependent manner for astilbin and that in solid dispersions. The permeability and apparent permeability coefficients (Papp) increased with drug in the Caco-2 cell. CONCLUSION In this study, it was found that PVP K30 and Tween 80 promoted the permeability of drugs best within a certain amount. For astilbin PVP K30 and surfactant combined carrier had a strong potential to improve oral bioavailability.
Collapse
Affiliation(s)
- Yan He
- School of Chemical Engineering and Light Industry , Guangdong University of Technology, Guangzhou, Guangdong, PR China
| | | | | | | | | | | |
Collapse
|
18
|
Barot M, Bagui M, Gokulgandhi MR, Mitra AK. Prodrug strategies in ocular drug delivery. Med Chem 2012; 8:753-68. [PMID: 22530907 DOI: 10.2174/157340612801216283] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 03/07/2012] [Accepted: 03/22/2012] [Indexed: 11/22/2022]
Abstract
Poor bioavailability of topically instilled drug is the major concern in the field of ocular drug delivery. Efflux transporters, static and dynamic ocular barriers often possess rate limiting factors for ocular drug therapy. Different formulation strategies like suspension, ointment, gels, nanoparticles, implants, dendrimers and liposomes have been employed in order to improve drug permeation and retention by evading rate limiting factors at the site of absorption. Chemical modification such as prodrug targeting various nutrient transporters (amino acids, peptide and vitamin) has evolved a great deal of interest to improve ocular drug delivery. In this review, we have discussed various prodrug strategies which have been widely applied for enhancing therapeutic efficacy of ophthalmic drugs. The purpose of this review is to provide an update on the utilization of prodrug concept in ocular drug delivery. In addition, this review will highlight ongoing academic and industrial research and development in terms of ocular prodrug design and delivery.
Collapse
Affiliation(s)
- Megha Barot
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | | | | | | |
Collapse
|
19
|
Functional polymeric nanoparticles for dexamethasone loading and release. Colloids Surf B Biointerfaces 2012; 93:59-66. [DOI: 10.1016/j.colsurfb.2011.12.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 12/06/2011] [Accepted: 12/08/2011] [Indexed: 11/20/2022]
|
20
|
Pignatello R, Mangiafico A, Basile L, Ruozi B, Furneri PM. Amphiphilic ion pairs of tobramycin with lipoamino acids. Eur J Med Chem 2011; 46:1665-71. [PMID: 21396748 DOI: 10.1016/j.ejmech.2011.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Revised: 01/11/2011] [Accepted: 02/11/2011] [Indexed: 10/18/2022]
|
21
|
Encapsulation of Protein-Polysaccharide HIP Complex in Polymeric Nanoparticles. JOURNAL OF DRUG DELIVERY 2011; 2011:458128. [PMID: 21603214 PMCID: PMC3095424 DOI: 10.1155/2011/458128] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 02/18/2011] [Indexed: 12/04/2022]
Abstract
The objective of the present study is to formulate and characterize a nanoparticulate-based formulation of a macromolecule in a hydrophobic ion pairing (HIP) complex form. So far, HIP complexation approach has been studied only for proteins with molecular weight of 10–20 kDa. Hence, we have selected bovine serum albumin (BSA) having higher molecular weight (66.3 kDa) as a model protein and dextran sulphate (DS) as a complexing polymer to generate HIP complex. We have prepared and optimized the HIP complex formation process of BSA with DS. Ionic interactions between basic amino acids of BSA with sulphate groups of DS were confirmed by FTIR analysis. Further, nanoparticles were prepared and characterized with respect to size and surface morphology. We observed significant entrapment of BSA in nanoparticles prepared with minimal amounts of PLGA polymer. Finally, results of circular dichroism and intrinsic fluorescence assay have clearly indicated that HIP complexation and method of nanoparticle preparation did not alter the secondary and tertiary structures of BSA.
Collapse
|