51
|
Nawaz HA, Schröck K, Schmid M, Krieghoff J, Maqsood I, Kascholke C, Kohn-Polster C, Schulz-Siegmund M, Hacker MC. Injectable oligomer-cross-linked gelatine hydrogels via anhydride-amine-conjugation. J Mater Chem B 2021; 9:2295-2307. [PMID: 33616150 DOI: 10.1039/d0tb02861d] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Injectable gelatine-based hydrogels are valuable tools for drug and cell delivery due to their extracellular matrix-like properties that can be adjusted by the degree of cross-linking. We have established anhydride-containing oligomers for the cross-linking of gelatine via anhydride-amine-conjugation. So far, this conversion required conditions not compatible with cell encapsulation or in vivo injection. In order to overcome this limitation, we developed an array of quarter-oligomers varying in comonomer composition and contents of reactive anhydride units reactive towards amine groups under physiological conditions. The oligomers were of low molecular weight (Mn < 5 kDa) with a high degree of chemically intact anhydrides. Chemical comonomer composition was determined by 1H-NMR. Dissolutions experiments confirmed improved hydrophilicity of the synthesized oligomers over our established compositions. Injectable formulations are described utilizing cytocompatible concentrations of constituent materials and proton-scavenging base. Degree of cross-linking and stiffness of injectable hydrogels were controlled by composition. The gels hold promise as injectable drug or cell carrier and as bioink.
Collapse
Affiliation(s)
- Hafiz Awais Nawaz
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany and Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Abdul Qadir Jillani road, Lahore, Pakistan
| | - Kathleen Schröck
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Maximilian Schmid
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Jan Krieghoff
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Iram Maqsood
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Christian Kascholke
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Caroline Kohn-Polster
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Michaela Schulz-Siegmund
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Michael C Hacker
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany and Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-Universität, Universitätsstraße 1, Düsseldorf, 40225 Düsseldorf, Germany.
| |
Collapse
|
52
|
F Nahhas A, F Nahhas A, J Webster T. Nanoscale pathogens treated with nanomaterial-like peptides: a platform technology appropriate for future pandemics. Nanomedicine (Lond) 2021; 16:1237-1254. [PMID: 33988037 PMCID: PMC8120868 DOI: 10.2217/nnm-2020-0447] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/29/2021] [Indexed: 01/13/2023] Open
Abstract
Viral infections are historically very difficult to treat. Although imperfect and time-consuming to develop, we do have some conventional vaccine and therapeutic approaches to stop viral spreading. Most importantly, all of this takes significant time while viruses continue to wreak havoc on our healthcare system. Furthermore, viral infections are accompanied by a weakened immune system which is often overlooked in antiviral drug strategies and requires additional drug development. In this review, for the first time, we touch on some promising alternative approaches to treat viral infections, specifically those focused on the use of platform nanomaterials with antiviral peptides. In doing so, this review presents a timely discussion of how we need to change our old way of treating viruses into one that can quickly meet the demands of COVID-19, as well as future pandemic-causing viruses, which will come.
Collapse
Affiliation(s)
- Alaa F Nahhas
- Biochemistry Department, College of Science, King Abdulaziz University, Jeddah 21589, KSA
| | - Alrayan F Nahhas
- Biochemistry Department, College of Science, King Abdulaziz University, Jeddah 21589, KSA
| | - Thomas J Webster
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
53
|
Pitorre M, Gazaille C, Pham LTT, Frankova K, Béjaud J, Lautram N, Riou J, Perrot R, Geneviève F, Moal V, Benoit JP, Bastiat G. Polymer-free hydrogel made of lipid nanocapsules, as a local drug delivery platform. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112188. [PMID: 34082987 DOI: 10.1016/j.msec.2021.112188] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 11/15/2022]
Abstract
Nanoparticle-loaded hydrogels are attractive pharmaceutical drug delivery systems that combine the advantages of both hydrogel (local administration and/or sustained drug release) and nanoparticle (stealthiness, targeting and decreased toxicity). The design of nanoparticle-loaded hydrogels is largely conventional, consisting of the dispersion of nanoparticles in a natural or synthetic polymer matrix to form a gel network. Novel nanoparticle-loaded hydrogels architecture could provide advantages in terms of innovation and application. We focused on the development of lipid nanocapsule (LNC)-based hydrogels without the use of a polymer matrix as a platform for drug delivery. Cytidine was modified by grafting palmitoyl chains (CytC16) and the new entity was added during the LNC phase-inversion formulation process allowing spontaneous gelation. Positioned at the oil/water interface, CytC16 acts as a crosslinking agent between LNCs. Association of the LNCs in a three-dimensional network led to the formation of polymer-free hydrogels. The viscoelastic properties of the LNC-based hydrogels depended on the LNC concentration and CytC16 loading but were not affected by the LNC size distribution. The LNC and drug-release profiles were controlled by the mechanical properties of the LNC-based hydrogels (slower release profiles correlated with higher viscoelasticity). Finally, the subcutaneous administration of LNC-based hydrogels led to classic inflammatory reactions of the foreign body-reaction type due to the endogenous character of CytC16, shown by cellular viability assays. New-generation nanoparticle-loaded hydrogels (LNC-based polymer-free hydrogels) show promise as implants for pharmaceutical applications. Once LNC release is completed, no gel matrix remains at the injection site, minimizing the additional toxicity due to the persistence of polymeric implants. Sustained drug-release profiles can be controlled by the mechanical properties of the hydrogels and could be tailor-made, depending on the therapeutic strategy chosen.
Collapse
Affiliation(s)
- Marion Pitorre
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Claire Gazaille
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | | | | | - Jérôme Béjaud
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Nolwenn Lautram
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Jérémie Riou
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Rodolphe Perrot
- Univ Angers, Service Commun d'Imageries et d'Analyses Microscopiques (SCIAM), SFR ICAT, F-49000 Angers, France
| | | | - Valérie Moal
- Biochemistry and Molecular Biology Department, University Hospital, Angers, France
| | | | | |
Collapse
|
54
|
Chen KJ, Plaunt AJ, Leifer FG, Kang JY, Cipolla D. Recent advances in prodrug-based nanoparticle therapeutics. Eur J Pharm Biopharm 2021; 165:219-243. [PMID: 33979661 DOI: 10.1016/j.ejpb.2021.04.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 03/10/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022]
Abstract
Extensive research into prodrug modification of active pharmaceutical ingredients and nanoparticle drug delivery systems has led to unprecedented levels of control over the pharmacological properties of drugs and resulted in the approval of many prodrug or nanoparticle-based therapies. In recent years, the combination of these two strategies into prodrug-based nanoparticle drug delivery systems (PNDDS) has been explored as a way to further advance nanomedicine and identify novel therapies for difficult-to-treat indications. Many of the PNDDS currently in the clinical development pipeline are expected to enter the market in the coming years, making the rapidly evolving field of PNDDS highly relevant to pharmaceutical scientists. This review paper is intended to introduce PNDDS to the novice reader while also updating those working in the field with a comprehensive summary of recent efforts. To that end, first, an overview of FDA-approved prodrugs is provided to familiarize the reader with their advantages over traditional small molecule drugs and to describe the chemistries that can be used to create them. Because this article is part of a themed issue on nanoparticles, only a brief introduction to nanoparticle-based drug delivery systems is provided summarizing their successful application and unfulfilled opportunities. Finally, the review's centerpiece is a detailed discussion of rationally designed PNDDS formulations in development that successfully leverage the strengths of prodrug and nanoparticle approaches to yield highly effective therapeutic options for the treatment of many diseases.
Collapse
|
55
|
Sustained Release Systems for Delivery of Therapeutic Peptide/Protein. Biomacromolecules 2021; 22:2299-2324. [PMID: 33957752 DOI: 10.1021/acs.biomac.1c00160] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Peptide/protein therapeutics have been significantly applied in the clinical treatment of various diseases such as cancer, diabetes, etc. owing to their high biocompatibility, specificity, and therapeutic efficacy. However, due to their immunogenicity, instability stemming from its complex tertiary and quaternary structure, vulnerability to enzyme degradation, and rapid renal clearance, the clinical application of protein/peptide therapeutics is significantly confined. Though nanotechnology has been demonstrated to prevent enzyme degradation of the protein therapeutics and thus enhance the half-life, issues such as initial burst release and uncontrollable release kinetics are still unsolved. Moreover, the traditional administration method results in poor patient compliance, limiting the clinical application of protein/peptide therapeutics. Exploiting the sustained-release formulations for more controllable delivery of protein/peptide therapeutics to decrease the frequency of injection and enhance patient compliance is thus greatly meaningful. In this review, we comprehensively summarize the substantial advancements of protein/peptide sustained-release systems in the past decades. In addition, the advantages and disadvantages of all these sustained-release systems in clinical application together with their future challenges are also discussed in this review.
Collapse
|
56
|
Castro LS, Lobo GS, Pereira P, Freire MG, Neves MC, Pedro AQ. Interferon-Based Biopharmaceuticals: Overview on the Production, Purification, and Formulation. Vaccines (Basel) 2021; 9:328. [PMID: 33915863 PMCID: PMC8065594 DOI: 10.3390/vaccines9040328] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
The advent of biopharmaceuticals in modern medicine brought enormous benefits to the treatment of numerous human diseases and improved the well-being of many people worldwide. First introduced in the market in the early 1980s, the number of approved biopharmaceutical products has been steadily increasing, with therapeutic proteins, antibodies, and their derivatives accounting for most of the generated revenues. The success of pharmaceutical biotechnology is closely linked with remarkable developments in DNA recombinant technology, which has enabled the production of proteins with high specificity. Among promising biopharmaceuticals are interferons, first described by Isaacs and Lindenmann in 1957 and approved for clinical use in humans nearly thirty years later. Interferons are secreted autocrine and paracrine proteins, which by regulating several biochemical pathways have a spectrum of clinical effectiveness against viral infections, malignant diseases, and multiple sclerosis. Given their relevance and sustained market share, this review provides an overview on the evolution of interferon manufacture, comprising their production, purification, and formulation stages. Remarkable developments achieved in the last decades are herein discussed in three main sections: (i) an upstream stage, including genetically engineered genes, vectors, and hosts, and optimization of culture conditions (culture media, induction temperature, type and concentration of inducer, induction regimens, and scale); (ii) a downstream stage, focusing on single- and multiple-step chromatography, and emerging alternatives (e.g., aqueous two-phase systems); and (iii) formulation and delivery, providing an overview of improved bioactivities and extended half-lives and targeted delivery to the site of action. This review ends with an outlook and foreseeable prospects for underdeveloped aspects of biopharma research involving human interferons.
Collapse
Affiliation(s)
- Leonor S. Castro
- CICECO–Aveiro Institute of Materials, Chemistry Department, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (L.S.C.); (G.S.L.); (M.G.F.)
| | - Guilherme S. Lobo
- CICECO–Aveiro Institute of Materials, Chemistry Department, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (L.S.C.); (G.S.L.); (M.G.F.)
| | - Patrícia Pereira
- Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal;
| | - Mara G. Freire
- CICECO–Aveiro Institute of Materials, Chemistry Department, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (L.S.C.); (G.S.L.); (M.G.F.)
| | - Márcia C. Neves
- CICECO–Aveiro Institute of Materials, Chemistry Department, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (L.S.C.); (G.S.L.); (M.G.F.)
| | - Augusto Q. Pedro
- CICECO–Aveiro Institute of Materials, Chemistry Department, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (L.S.C.); (G.S.L.); (M.G.F.)
| |
Collapse
|
57
|
Ni Z, Yu H, Wang L, Shen D, Elshaarani T, Fahad S, Khan A, Haq F, Teng L. Recent research progress on polyphosphazene-based drug delivery systems. J Mater Chem B 2021; 8:1555-1575. [PMID: 32025683 DOI: 10.1039/c9tb02517k] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In recent years, synthetic polymer materials have become a research hotspot in the field of drug delivery. Compared with natural polymer materials, synthetic polymer materials have more flexible structural adjustability, and can be designed to obtain clinically required delivery vehicles. Polyphosphazenes are one of the most promising biomedical materials in the future due to their controllable degradation properties and structural flexibility. These materials can be designed by controlling the hydrophilic and hydrophobic balance, introducing functional groups or drugs to form different forms of administration, such as nanoparticles, polyphosphazene-drug conjugates, injectable hydrogels, coatings, etc. In addition, the flexible backbone of polyphosphazenes and the flexibility of substitution enable them to meet researchers' design requirements in terms of stereochemistry, nanostructures, and topologies. At present, researchers have achieved a lot of successful practices in the field of targeted delivery of anticancer drugs/proteins/genes, bone tissue engineering repair, cell imaging tracking, photothermal therapy, and immunologic preparations. This review provides a summary of the progress of the recent 10 years of polyphosphazene-based drug delivery systems in terms of of chemical structure and functions.
Collapse
Affiliation(s)
- Zhipeng Ni
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Haojie Yu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Li Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Di Shen
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Tarig Elshaarani
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Shah Fahad
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Amin Khan
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Fazal Haq
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Lison Teng
- Biological Surgery and Cancer Center, The First Affiliated Hospital, Zhejiang University, 310003, P. R. China
| |
Collapse
|
58
|
Sharma DS, Wadhwa S, Gulati M, Kadukkattil Ramanunny A, Awasthi A, Singh SK, Khursheed R, Corrie L, Chitranshi N, Gupta VK, Vishwas S. Recent advances in intraocular and novel drug delivery systems for the treatment of diabetic retinopathy. Expert Opin Drug Deliv 2020; 18:553-576. [PMID: 33143473 DOI: 10.1080/17425247.2021.1846518] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction: Diabetic retinopathy (DR) is associated with damage to the retinal blood vessels that lead eventually to vision loss. The existing treatments of DR are invasive, expensive, and cumbersome. To overcome challenges associated with existing therapies, various intraocular sustained release and novel drug delivery systems (NDDS) have been explored.Areas covered: The review discusses recently developed intraocular devices for sustained release of drugs as well as novel noninvasive drug delivery systems that have met a varying degree of success in local delivery of drugs to retinal circulation.Expert opinion: The intraocular devices have got very good success in providing sustained release of drugs in patients. The development of NDDS and their application through the ocular route has certainly provided an edge to treat DR over existing therapies such as anti-VEGF administration but their success rate is quite low. Moreover, most of them have proved to be effective only in animal models. In addition, the extent of targeting the drug to the retina still remains variable and unpredictable. The toxicity aspect of the NDDS has generally been neglected. In order to have successful commercialization of nanotechnology-based innovations well-designed clinical research studies need to be conducted to evaluate their clinical superiority over that of the existing formulations.
Collapse
Affiliation(s)
- Deep Shikha Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Sheetu Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | | | - Ankit Awasthi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Rubiya Khursheed
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Leander Corrie
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Nitin Chitranshi
- Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Australia
| | - Vivek Kumar Gupta
- Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Australia
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| |
Collapse
|
59
|
Cottura N, Howarth A, Rajoli RKR, Siccardi M. The Current Landscape of Novel Formulations and the Role of Mathematical Modeling in Their Development. J Clin Pharmacol 2020; 60 Suppl 1:S77-S97. [PMID: 33205431 DOI: 10.1002/jcph.1715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/25/2020] [Indexed: 12/15/2022]
Abstract
Drug delivery is an integral part of the drug development process, influencing safety and efficacy of active pharmaceutical ingredients. The application of nanotechnology has enabled the discovery of novel formulations for numerous therapeutic purposes across multiple disease areas. However, evaluation of novel formulations in clinical scenarios is slow and hampered due to various ethical and logistical barriers. Computational models have the ability to integrate existing domain knowledge and mathematical correlations, to rationalize the feasibility of using novel formulations for safely enhancing drug delivery, identifying suitable candidates, and reducing the burden on preclinical and clinical studies. In this review, types of novel formulations and their application through several routes of administration and the use of modeling approaches that can find application in different stages of the novel formulation development process are discussed.
Collapse
Affiliation(s)
- Nicolas Cottura
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Alice Howarth
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Rajith K R Rajoli
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Marco Siccardi
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
60
|
Zaghmi A, Drouin-Ouellet J, Brambilla D, Gauthier MA. Treating brain diseases using systemic parenterally-administered protein therapeutics: Dysfunction of the brain barriers and potential strategies. Biomaterials 2020; 269:120461. [PMID: 33218788 DOI: 10.1016/j.biomaterials.2020.120461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022]
Abstract
The parenteral administration of protein therapeutics is increasingly gaining importance for the treatment of human diseases. However, the presence of practically impermeable blood-brain barriers greatly restricts access of such pharmaceutics to the brain. Treating brain disorders with proteins thus remains a great challenge, and the slow clinical translation of these therapeutics may be largely ascribed to the lack of appropriate brain delivery system. Exploring new approaches to deliver proteins to the brain by circumventing physiological barriers is thus of great interest. Moreover, parallel advances in the molecular neurosciences are important for better characterizing blood-brain interfaces, particularly under different pathological conditions (e.g., stroke, multiple sclerosis, Parkinson's disease, and Alzheimer's disease). This review presents the current state of knowledge of the structure and the function of the main physiological barriers of the brain, the mechanisms of transport across these interfaces, as well as alterations to these concomitant with brain disorders. Further, the different strategies to promote protein delivery into the brain are presented, including the use of molecular Trojan horses, the formulation of nanosystems conjugated/loaded with proteins, protein-engineering technologies, the conjugation of proteins to polymers, and the modulation of intercellular junctions. Additionally, therapeutic approaches for brain diseases that do not involve targeting to the brain are presented (i.e., sink and scavenging mechanisms).
Collapse
Affiliation(s)
- A Zaghmi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada
| | - J Drouin-Ouellet
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - D Brambilla
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - M A Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada.
| |
Collapse
|
61
|
Khodaverdi E, Delroba K, Mohammadpour F, Khameneh B, Sajadi Tabassi SA, Tafaghodi M, Kamali H, Hadizadeh F. In-vitro Release Evaluation of Growth Hormone from an Injectable In-Situ Forming Gel Using PCL-PEG-PCL Thermosensitive Triblock. Curr Drug Deliv 2020; 17:174-183. [PMID: 31987020 DOI: 10.2174/1567201817666200120120105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 10/02/2019] [Accepted: 12/31/2019] [Indexed: 12/16/2022]
Abstract
OBJECTIVE An injectable long acting In-Situ Forming Gel (ISFG) of human Growth Hormone (hGH) was prepared by using triblock PCL-PEG-PCL (Mw 1500-1500-1500). Ring-Opening Polymerization (ROP) of triblock using microwave was applied. METHODS The BCA protein assay Kit was used to determine the concentration of hGH in the in-vitro release medium. Finally, Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE) tests and Circular Dichroism (CD) spectrum were done to approve the stability of released hGH. The result of ROP demonstrated that the proportion of PCL to PEG accorded with the initial molar ratio of the monomers. The cross-section of the Surface Electron Microscopy (SEM) indicated the porous framework of the hydrogel could load the drug into its tridimensional matrixes structure. There is the low initial burst release of hGH from the supramolecular hydrogel. RESULTS The maximum in-vitro release of hGH was 71.2 % ± 1.5 that were due to hGH degrading after this time (21 days). The CD spectrum and SDS-PAGE results confirmed the stability of hGH during invitro release evaluation. CONCLUSION The results suggest that the sustained-release formulation using PCL-PEG-PCL can be applied to control the release of hGH.
Collapse
Affiliation(s)
- Elham Khodaverdi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Delroba
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mohammadpour
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahman Khameneh
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sayyed A Sajadi Tabassi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Tafaghodi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Kamali
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
62
|
Tryfonidou MA, de Vries G, Hennink WE, Creemers LB. "Old Drugs, New Tricks" - Local controlled drug release systems for treatment of degenerative joint disease. Adv Drug Deliv Rev 2020; 160:170-185. [PMID: 33122086 DOI: 10.1016/j.addr.2020.10.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022]
Abstract
Osteoarthritis (OA) and chronic low back pain (CLBP) caused by intervertebral disc (IVD) degeneration are joint diseases that have become major causes for loss of quality of life worldwide. Despite the unmet need, effective treatments other than invasive, and often ineffective, surgery are lacking. Systemic administration of drugs entails suboptimal local drug exposure in the articular joint and IVD. This review provides an overview of the potency of biomaterial-based drug delivery systems as novel treatment modality, with a focus on the biological effects of drug release systems that have reached translation at the level of in vivo models and relevant ex vivo models. These studies have shown encouraging results of biomaterial-based local delivery of several types of drugs, mostly inhibitors of inflammatory cytokines or other degenerative factors. Prevention of inflammation and degeneration and pain relief was achieved, although mainly in small animal models, with interventions applied at an early disease stage. Less convincing data were obtained with the delivery of regenerative factors. Multidisciplinary efforts towards tackling the discord between in vitro and in vivo release, combined with adaptations in the regulatory landscape may be needed to enhance safe and expeditious introduction of more and more effective controlled release-based treatments with the OA and CLBP patients.
Collapse
|
63
|
Lanier OL, Ficarrotta JM, Adjei I, Wable D, Lewis C, Nacea C, Sharma B, Dobson J, McFetridge P. Magnetically Responsive Polymeric Microparticles for the Triggered Delivery of a Complex Mixture of Human Placental Proteins. Macromol Biosci 2020; 21:e2000249. [PMID: 33015960 DOI: 10.1002/mabi.202000249] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/09/2020] [Indexed: 12/21/2022]
Abstract
Bone loss through traumatic injury is a significant clinical issue. Researchers have created many scaffold types to mimic an extracellular matrix to provide structural support for the formation of new bone, however functional regeneration of larger scaffolds has not been fully achieved. Newer scaffolds aim to deliver bioactive molecules to improve tissue regeneration. To achieve a more comprehensive regenerative response, a magnetically triggerable polymeric microparticle platform is developed for the on-demand release of a complex mixture of isolated human placental proteins. This system is composed of polycaprolactone (PCL) microparticles, encapsulating magnetic nanoparticles (MNPs), and placental proteins. When subjected to an alternating magnetic field (AMF), the MNPs heat and melt the PCL, enhancing the diffusion of proteins from microparticles. When the field is off, the PCL re-solidifies. This potentially allows for cyclic drug delivery. Here the design, synthesis, and proof-of-concept experiments for this system are reported. In addition, it is shown that the proteins retain function after being magnetically released. The ability to trigger the release of complex protein mixtures on-demand may provide a significant advantage with wounds where stagnation of healing processes can occur (e.g., large segmented bone defects).
Collapse
Affiliation(s)
- Olivia L Lanier
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Joseph M Ficarrotta
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Isaac Adjei
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Dayita Wable
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Camryn Lewis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Christopher Nacea
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Jon Dobson
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Peter McFetridge
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
64
|
|
65
|
Wang S, Liu R, Fu Y, Kao WJ. Release mechanisms and applications of drug delivery systems for extended-release. Expert Opin Drug Deliv 2020; 17:1289-1304. [PMID: 32619149 DOI: 10.1080/17425247.2020.1788541] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Drug delivery systems with extended-release profiles are ideal in improving patient compliance with enhanced efficacy. To develop devices capable of a prolonged delivery kinetics, it is crucial to understand the various underlying mechanisms contributing to extended drug release and the impact thereof on modulating the long-term performance of such systems in a practical application environment. AREAS COVERED This review article intends to provide a comprehensive summary of release mechanisms in extended-release drug delivery systems, particularly polymer-based systems; however, other material types will also be mentioned. Selected current research in the delivery of small molecule drugs and macromolecules is highlighted. Emphasis is placed on the combined impact of different release mechanisms and drug properties on the long-term release kinetics in vitro and in vivo. EXPERT OPINION The development of drug delivery systems over an extended duration is promising but also challenging when considering the numerous interrelated delivery-related parameters. Achieving a well-controlled extended drug release requires advanced techniques to minimize burst release and lag phase, a better understanding of the dynamic interrelationship between drug properties and release profiles over time, and a thorough elucidation of the impact of multiple in vivo conditions to methodically evaluate the eventual clinical efficacy.
Collapse
Affiliation(s)
- Shuying Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University , Chengdu, China
| | - Renhe Liu
- Global Health Drug Discovery Institute , Beijing, China
| | - Yao Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University , Chengdu, China
| | - W John Kao
- Department of Industrial and Manufacturing Systems Engineering, Biomedical Engineering Programme, Chemical Biology Centre, and Li Ka Shing Faculty of Medicine, The University of Hong Kong , Pokfulam, China
| |
Collapse
|
66
|
Xu C, Hu W, Zhang N, Qi Y, Nie JJ, Zhao N, Yu B, Xu FJ. Genetically multimodal therapy mediated by one polysaccharides-based supramolecular nanosystem. Biomaterials 2020; 248:120031. [DOI: 10.1016/j.biomaterials.2020.120031] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/20/2022]
|
67
|
Soltani R, Guo S, Bianco A, Ménard‐Moyon C. Carbon Nanomaterials Applied for the Treatment of Inflammatory Diseases: Preclinical Evidence. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Rym Soltani
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572 University of Strasbourg, ISIS Strasbourg 67000 France
| | - Shi Guo
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572 University of Strasbourg, ISIS Strasbourg 67000 France
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572 University of Strasbourg, ISIS Strasbourg 67000 France
| | - Cécilia Ménard‐Moyon
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572 University of Strasbourg, ISIS Strasbourg 67000 France
| |
Collapse
|
68
|
Bayer IS. Hyaluronic Acid and Controlled Release: A Review. Molecules 2020; 25:molecules25112649. [PMID: 32517278 PMCID: PMC7321085 DOI: 10.3390/molecules25112649] [Citation(s) in RCA: 198] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
Hyaluronic acid (HA) also known as hyaluronan, is a natural polysaccharide—an anionic, non-sulfated glycosaminoglycan—commonly found in our bodies. It occurs in the highest concentrations in the eyes and joints. Today HA is used during certain eye surgeries and in the treatment of dry eye disease. It is a remarkable natural lubricant that can be injected into the knee for patients with knee osteoarthritis. HA has also excellent gelling properties due to its capability to bind water very quickly. As such, it is one the most attractive controlled drug release matrices and as such, it is frequently used in various biomedical applications. Due to its reactivity, HA can be cross-linked or conjugated with assorted bio-macromolecules and it can effectively encapsulate several different types of drugs, even at nanoscale. Moreover, the physiological significance of the interactions between HA and its main membrane receptor, CD44 (a cell-surface glycoprotein that modulates cell–cell interactions, cell adhesion and migration), in pathological processes, e.g., cancer, is well recognized and this has resulted in an extensive amount of studies on cancer drug delivery and tumor targeting. HA acts as a therapeutic but also as a tunable matrix for drug release. Thus, this review focuses on controlled or sustained drug release systems assembled from HA and its derivatives. More specifically, recent advances in controlled release of proteins, antiseptics, antibiotics and cancer targeting drugs from HA and its derivatives were reviewed. It was shown that controlled release from HA has many benefits such as optimum drug concentration maintenance, enhanced therapeutic effects, improved efficiency of treatment with less drug, very low or insignificant toxicity and prolonged in vivo release rates.
Collapse
Affiliation(s)
- Ilker S Bayer
- Smart Materials, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| |
Collapse
|
69
|
Zhang N, Mei K, Guan P, Hu X, Zhao Y. Protein-Based Artificial Nanosystems in Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907256. [PMID: 32378796 DOI: 10.1002/smll.201907256] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 05/21/2023]
Abstract
Proteins, like actors, play different roles in specific applications. In the past decade, significant achievements have been made in protein-engineered biomedicine for cancer therapy. Certain proteins such as human serum albumin, working as carriers for drug/photosensitizer delivery, have entered clinical use due to their long half-life, biocompatibility, biodegradability, and inherent nonimmunogenicity. Proteins with catalytic abilities are promising as adjuvant agents for other therapeutic modalities or as anticancer drugs themselves. These catalytic proteins are usually defined as enzymes with high biological activity and substrate specificity. However, clinical applications of these kinds of proteins remain rare due to protease-induced denaturation and weak cellular permeability. Based on the characteristics of different proteins, tailor-made protein-based nanosystems could make up for their individual deficiencies. Therefore, elaborately designed protein-based nanosystems, where proteins serve as drug carriers, adjuvant agents, or therapeutic drugs to make full use of their intrinsic advantages in cancer therapy, are reviewed. Up-to-date progress on research in the field of protein-based nanomedicine is provided.
Collapse
Affiliation(s)
- Nan Zhang
- School of Natural and Applied Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Kun Mei
- School of Natural and Applied Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Ping Guan
- School of Natural and Applied Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Xiaoling Hu
- School of Natural and Applied Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| |
Collapse
|
70
|
Chaharband F, Daftarian N, Kanavi MR, Varshochian R, Hajiramezanali M, Norouzi P, Arefian E, Atyabi F, Dinarvand R. Trimethyl chitosan-hyaluronic acid nano-polyplexes for intravitreal VEGFR-2 siRNA delivery: Formulation and in vivo efficacy evaluation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 26:102181. [DOI: 10.1016/j.nano.2020.102181] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/06/2020] [Accepted: 02/26/2020] [Indexed: 12/29/2022]
|
71
|
Lee S, Park S, Nguyen MT, Lee E, Kim J, Baek S, Kim CJ, Jang YJ, Choe H. A chemical conjugate between HER2-targeting antibody fragment and Pseudomonas exotoxin A fragment demonstrates cytotoxic effects on HER2-expressing breast cancer cells. BMB Rep 2020. [PMID: 30670149 PMCID: PMC6726212 DOI: 10.5483/bmbrep.2019.52.8.250] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Conventionally, immunotoxins have been produced as a single polypeptide from fused genes of an antibody fragment and a toxin. In this study, we adopted a unique approach of chemical conjugation of a toxin protein and an antibody fragment. The two genes were separately expressed in Escherichia coli and purified to high levels of purity. The two purified proteins were conjugated using a chemical linker. The advantage of this approach is its ability to overcome the problem of low recombinant immunotoxin production observed in some immunotoxins. Another advantage is that various combinations of immunotoxins can be prepared with fewer efforts, because the chemical conjugation of components is relatively simpler than the processes involved in cloning, expression, and purification of multiple immunotoxins. As a proof of concept, the scFv of trastuzumab and the PE24 fragment of Pseudomonas exotoxin A were separately produced using E. coli and then chemically crosslinked. The new immunotoxin was tested on four breast cancer cell lines variably expressing HER2. The chemically crosslinked immunotoxin exhibited cytotoxicity in proportion to the expression level of HER2. In conclusion, the present study revealed an alternative method of generating an immunotoxin that could effectively reduce the viability of HER2-expressing breast cancer cells. These results suggest the effectiveness of this method of immunotoxin crosslinking as a suitable alternative for producing immunotoxins. [BMB
Collapse
Affiliation(s)
- Sunju Lee
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Sangsu Park
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Minh Tan Nguyen
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea; Department of Biotechnology, NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
| | - Eunyoung Lee
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Julee Kim
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Sangki Baek
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Chong Jai Kim
- Department of Pathology, Asan-Minnesota Institute for Innovating Transplantation, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Yeon Jin Jang
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Han Choe
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
72
|
Andrianov AK, Marin A, Deng J, Fuerst TR. Protein-loaded soluble and nanoparticulate formulations of ionic polyphosphazenes and their interactions on molecular and cellular levels. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 106:110179. [PMID: 31753403 PMCID: PMC6903416 DOI: 10.1016/j.msec.2019.110179] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/09/2019] [Accepted: 09/08/2019] [Indexed: 11/21/2022]
Abstract
Nanoparticulate and water-soluble formulations of ionic polyphosphazenes and protein cargo - lysozyme (LYZ) were prepared by their self-assembly in aqueous solutions at near physiological pH (pH 7.4) in the presence and absence of an ionic cross-linker - spermine tetrahydrochloride. Efficiency of LYZ encapsulation, physico-chemical characteristics of formulations, and the effect of reaction parameters were investigated using asymmetric flow field flow fractionation (AF4) and dynamic light scattering (DLS) methods. The effect of both polymer formulations on encapsulated LYZ was evaluated using soluble oligosaccharide substrate, whereas their ability to present the protein to cellular surfaces was assessed by measuring enzymatic activity of encapsulated LYZ against Micrococcus lysodeikticus cells. It was found that both soluble and cross-linked polymer matrices reduce lysis of bacterial cells by LYZ, whereas activity of encapsulated protein against oligosaccharide substrate remained practically unchanged indicating no adverse effect of polyphosphazene on protein integrity. Moreover, nanoparticulate formulations display distinctly different behavior in cellular assays when compared to their soluble counterparts. LYZ encapsulated in polyphosphazene nanoparticles shows approximately 2.5-fold higher activity in its ability to lyse cells as compared with water-soluble LYZ-PCPP formulations. A new approach to PEGylation of polyphosphazene nanoparticles was also developed. The method utilizes a new ionic polyphosphazene derivative, which contains graft (polyethylene glycol) chains. PEGylation allows for an improved control over the size of nanoparticles and broader modulation of their cross-linking density, while still permitting for protein presentation to cellular substrates.
Collapse
Affiliation(s)
- Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Dr., Rockville, MD 20850, United States.
| | - Alexander Marin
- Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Dr., Rockville, MD 20850, United States
| | - Joseph Deng
- Department of Biology, College of Computer, Mathematical, and Natural Sciences, 1210 Biology - Psychology Building, 4094 Campus Drive, College Park, MD 20742, United States
| | - Thomas R Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Dr., Rockville, MD 20850, United States; Department of Cell Biology and Molecular Genetics, 1109 Microbiology Building, University of Maryland, College Park, MD 20742, United States
| |
Collapse
|
73
|
Polley P, Gupta S, Singh R, Pradhan A, Basu SM, V. R, Yadava SK, Giri J. Protein–Sugar-Glass Nanoparticle Platform for the Development of Sustained-Release Protein Depots by Overcoming Protein Delivery Challenges. Mol Pharm 2019; 17:284-300. [DOI: 10.1021/acs.molpharmaceut.9b01022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Poulomi Polley
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Shivam Gupta
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
| | - Ruby Singh
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Arpan Pradhan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, Maharashtra, India
| | - Suparna Mercy Basu
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Remya V.
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Sunil Kumar Yadava
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| |
Collapse
|
74
|
Smith J, McMullen P, Yuan Z, Pfaendtner J, Jiang S. Elucidating Molecular Design Principles for Charge-Alternating Peptides. Biomacromolecules 2019; 21:435-443. [DOI: 10.1021/acs.biomac.9b01191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Josh Smith
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195-1750, United States
| | - Patrick McMullen
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195-1750, United States
| | - Zhefan Yuan
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195-1750, United States
| | - Jim Pfaendtner
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195-1750, United States
| | - Shaoyi Jiang
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195-1750, United States
| |
Collapse
|
75
|
Wu C, Mu H. Lipid and PLGA Microparticles for Sustained Delivery of Protein and Peptide Drugs. Pharm Nanotechnol 2019; 8:22-32. [PMID: 31663483 DOI: 10.2174/2211738507666191029160944] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/03/2019] [Accepted: 10/16/2019] [Indexed: 01/01/2023]
Abstract
Solid lipid particles have a great potential in sustained drug delivery, the lipid excipients are solid at room temperature with a slow degradation rate. Poly (D, L-lactic-coglycolic acid) (PLGA) has been successfully clinically applied for the sustained delivery of peptide drugs. A recent study showed the advantage of hybrid PLGA-lipid microparticles (MPs) over PLGA MPs for the sustained delivery of peptide drug in vivo. In this paper, we briefly present PLGA MPs, solid lipid MPs and PLGA lipid hybrid MP prepared by the double emulsion method and the spray drying method and discuss the effects of excipients on encapsulation efficiency of protein and peptide drugs in the MPs. The pros and cons of PLGA MPs, solid lipid MPs and PLGA lipid hybrid MP as carriers for sustained delivery of protein and peptide drugs are also discussed.
Collapse
Affiliation(s)
- Chengyu Wu
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK 2100, Copenhagen, Denmark
| | - Huiling Mu
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK 2100, Copenhagen, Denmark
| |
Collapse
|
76
|
Sikder S, Gote V, Alshamrani M, Sicotte J, Pal D. Long-term delivery of protein and peptide therapeutics for cancer therapies. Expert Opin Drug Deliv 2019; 16:1113-1131. [DOI: 10.1080/17425247.2019.1662785] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Sadia Sikder
- Division of Pharmacological & Pharmaceutical Sciences, University of Missouri Kansas City, Kansas, MO, USA
| | - Vrinda Gote
- Division of Pharmacological & Pharmaceutical Sciences, University of Missouri Kansas City, Kansas, MO, USA
| | - Meshal Alshamrani
- Division of Pharmacological & Pharmaceutical Sciences, University of Missouri Kansas City, Kansas, MO, USA
| | - Jeff Sicotte
- Division of Pharmacological & Pharmaceutical Sciences, University of Missouri Kansas City, Kansas, MO, USA
| | - Dhananjay Pal
- Division of Pharmacological & Pharmaceutical Sciences, University of Missouri Kansas City, Kansas, MO, USA
| |
Collapse
|
77
|
Lim WQ, Phua SZF, Zhao Y. Redox-Responsive Polymeric Nanocomplex for Delivery of Cytotoxic Protein and Chemotherapeutics. ACS APPLIED MATERIALS & INTERFACES 2019; 11:31638-31648. [PMID: 31389684 DOI: 10.1021/acsami.9b09605] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Responsive delivery of anticancer proteins into cells is an emerging field in biological therapeutics. Currently, the delivery of proteins is highly compromised by multiple successive physiological barriers that reduce the therapeutic efficacy. Hence, there is a need to design a robust and sustainable nanocarrier to provide suitable protection of proteins and overcome the physiological barriers for better cellular accumulation. In this work, polyethylenimine (PEI) cross-linked by oxaliplatin(IV) prodrug (oxliPt(IV)) was used to fabricate a redox-responsive nanocomplex (PEI-oxliPt(IV)@RNBC/GOD) for the delivery of a reactive oxygen species-cleavable, reversibly caged RNase A protein (i.e., RNase A nitrophenylboronic conjugate, RNBC) and glucose oxidase (GOD) in order to realize efficient cancer treatment. The generation of hydrogen peroxide by GOD can uncage and restore the enzymatic activity of RNBC. On account of the responsiveness of the nanocomplex to highly reducing cellular environment, it would dissociate and release the protein and active oxaliplatin drug, causing cell death by both catalyzing RNA degradation and inhibiting DNA synthesis. As assessed by the RNA degradation assay, the activity of the encapsulated RNBC was recovered by the catalytic production of hydrogen peroxide from GOD and glucose substrate overexpressed in cancer cells. Monitoring of the changes in nanoparticle size confirmed that the nanocomplex could dissociate in the reducing environment, with the release of active oxaliplatin drug and protein. Confocal laser scanning microscopy (CLSM) and flow cytometry analysis revealed highly efficient accumulation of the nanocomplex as compared to free native proteins. In vitro cytotoxicity experiments using 4T1 cancer cells showed ∼80% cell killing efficacy, with highly efficient apoptosis induction. Assisted by the cationic polymeric carrier, it was evident from CLSM images that intracellular delivery of the therapeutic protein significantly depleted the RNA level. Thus, this work provides a promising platform for the delivery of therapeutic proteins and chemotherapeutic drugs for efficient cancer treatment.
Collapse
Affiliation(s)
- Wei Qi Lim
- NTU-Northwestern Institute for Nanomedicine, Interdisciplinary Graduate School , Nanyang Technological University , 50 Nanyang Drive , 637553 , Singapore
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences , Nanyang Technological University , 21 Nanyang Link , 637371 , Singapore
| | - Soo Zeng Fiona Phua
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences , Nanyang Technological University , 21 Nanyang Link , 637371 , Singapore
| | - Yanli Zhao
- NTU-Northwestern Institute for Nanomedicine, Interdisciplinary Graduate School , Nanyang Technological University , 50 Nanyang Drive , 637553 , Singapore
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences , Nanyang Technological University , 21 Nanyang Link , 637371 , Singapore
| |
Collapse
|
78
|
Cao Y, Samy KE, Bernards DA, Desai TA. Recent advances in intraocular sustained-release drug delivery devices. Drug Discov Today 2019; 24:1694-1700. [PMID: 31173915 PMCID: PMC6708500 DOI: 10.1016/j.drudis.2019.05.031] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/16/2019] [Accepted: 05/31/2019] [Indexed: 12/22/2022]
Abstract
Topical eye-drop administration and intravitreal injections are the current standard for ocular drug delivery. However, patient adherence to the drug regimen and insufficient administration frequency are well-documented challenges to this field. In this review, we describe recent advances in intraocular implants designed to deliver therapeutics for months to years, to obviate the issues of patient adherence. We highlight recent advances in monolithic ocular implants in the literature, the commercialization pipeline, and approved for the market. We also describe design considerations based on material selection, active pharmaceutical ingredient, and implantation site.
Collapse
Affiliation(s)
- Yiqi Cao
- UC Berkeley-UCSF Graduate Program in Bioengineering, 1700 4th Street, San Francisco, CA 94158, United States
| | - Karen E Samy
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1700 4th Street, San Francisco, CA 94158, USA
| | - Daniel A Bernards
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1700 4th Street, San Francisco, CA 94158, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1700 4th Street, San Francisco, CA 94158, USA.
| |
Collapse
|
79
|
Lee S, Park S, Nguyen MT, Lee E, Kim J, Baek S, Kim CJ, Jang YJ, Choe H. A chemical conjugate between HER2-targeting antibody fragment and Pseudomonas exotoxin A fragment demonstrates cytotoxic effects on HER2-expressing breast cancer cells. BMB Rep 2019; 52:496-501. [PMID: 30670149 PMCID: PMC6726212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/11/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2023] Open
Abstract
Conventionally, immunotoxins have been produced as a single polypeptide from fused genes of an antibody fragment and a toxin. In this study, we adopted a unique approach of chemical conjugation of a toxin protein and an antibody fragment. The two genes were separately expressed in Escherichia coli and purified to high levels of purity. The two purified proteins were conjugated using a chemical linker. The advantage of this approach is its ability to overcome the problem of low recombinant immunotoxin production observed in some immunotoxins. Another advantage is that various combinations of immunotoxins can be prepared with fewer efforts, because the chemical conjugation of components is relatively simpler than the processes involved in cloning, expression, and purification of multiple immunotoxins. As a proof of concept, the scFv of trastuzumab and the PE24 fragment of Pseudomonas exotoxin A were separately produced using E. coli and then chemically crosslinked. The new immunotoxin was tested on four breast cancer cell lines variably expressing HER2. The chemically crosslinked immunotoxin exhibited cytotoxicity in proportion to the expression level of HER2. In conclusion, the present study revealed an alternative method of generating an immunotoxin that could effectively reduce the viability of HER2-expressing breast cancer cells. These results suggest the effectiveness of this method of immunotoxin crosslinking as a suitable alternative for producing immunotoxins. [BMB Reports 2019; 52(8): 496-501].
Collapse
Affiliation(s)
- Sunju Lee
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Sangsu Park
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Minh Tan Nguyen
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
- Department of Biotechnology, NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City 70000,
Vietnam
| | - Eunyoung Lee
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Julee Kim
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Sangki Baek
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Chong Jai Kim
- Department of Pathology, Asan-Minnesota Institute for Innovating Transplantation, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Yeon Jin Jang
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Han Choe
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505,
Korea
| |
Collapse
|
80
|
Arrighi A, Marquette S, Peerboom C, Denis L, Goole J, Amighi K. Development of PLGA microparticles with high immunoglobulin G-loaded levels and sustained-release properties obtained by spray-drying a water-in-oil emulsion. Int J Pharm 2019; 566:291-298. [DOI: 10.1016/j.ijpharm.2019.05.070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 11/24/2022]
|
81
|
Shen X, Wang L, Xu C, Yang J, Peng R, Hu X, Wang F, Zheng H, Lao X. Fusion of thymosin alpha 1 with mutant IgG1 CH3 prolongs half-life and enhances antitumor effects in vivo. Int Immunopharmacol 2019; 74:105662. [PMID: 31220695 DOI: 10.1016/j.intimp.2019.05.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/13/2019] [Accepted: 05/23/2019] [Indexed: 10/26/2022]
Abstract
Thymosin alpha 1 (Tα1) is an immunomodulatory polypeptide secreted from the thymus. Tα1 has a wide range of biological functions, such as immunomodulation and endocrine regulation. Tα1 also displays antiviral and antitumor activities. Tα1 has been successfully used in clinical adjuvant therapy for solid tumors to improve the immune response of patients undergoing chemotherapy and radiotherapy. However, the half-life of Tα1 in the body is short, so frequent administration is required to maintain efficacy. In order to improve the pharmacokinetic profile of Tα1, we linked the mutated CH3 (mCH3) fragment of IgG1 (human) to the C-terminus of Tα1 to produce a long-acting fusion protein, Tα1-mCH3. The half-life of Tα1-mCH3 (47 h) was substantially increased compared with that of the parent molecule Tα1 (3 h). In vivo studies indicated that mCH3 fusion retained the original biological activity of Tα1, and Tα1-mCH3 showed slightly better immunomodulatory effect than Ta1. In the 4 T1 and B16F10 tumor xenograft models, Tα1-mCH3 induced a greater abundance of CD4+ and CD8+ T-cells in tumor tissues compared with Ta1. Tα1-mCH3 exhibited better effect in promoting the production of IL-2 and IFN-γ compared with Tα1. Therefore, Tα1-mCH3 more efficiently inhibited the growth of 4 T1 and B16F10 tumors than Tα1. In conclusion, fusion with mCH3 is an attractive strategy to lengthen the half-life and increase the activity of Tα1.
Collapse
Affiliation(s)
- Xutong Shen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Liping Wang
- Department of Clinical Oncology, the First City Hospital of Chenzhou, Hunan 423000, PR China
| | - Caoying Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jiahui Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Renhao Peng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xinyi Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Fanwen Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Heng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xingzhen Lao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
82
|
Condict L, Paramita VD, Kasapis S. Dairy protein–ligand interactions upon thermal processing and targeted delivery for the design of functional foods. Curr Opin Food Sci 2019. [DOI: 10.1016/j.cofs.2019.03.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
83
|
The long-term delivery of proteins and peptides using micro/nanoparticles: overview and perspectives. Ther Deliv 2019; 10:269-272. [PMID: 31094301 DOI: 10.4155/tde-2019-0016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
84
|
Souery WN, Arun Kumar S, Prasca-Chamorro D, Moore DM, Good J, Bishop CJ. Controlling and quantifying the stability of amino acid-based cargo within polymeric delivery systems. J Control Release 2019; 300:102-113. [PMID: 30826372 DOI: 10.1016/j.jconrel.2019.02.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 12/23/2022]
Abstract
In recent years, the rapid growth and availability of protein and peptide therapeutics has not only expanded the boundaries of modern science but has also revolutionized the practice of medicine today. The potential of such therapies, however, is greatly limited by the innate instabilities of proteins and peptides, which is further magnified during therapeutic formulation processing, transport, storage, and administration. In this paper, we will consider the unique stability challenges associated with protein/peptide polymeric delivery systems from an engineering approach oriented towards the quantification and modification of amino acid-based cargo stability. While a number of methods have been developed for the purposes of quantifying factors affecting protein and peptide stability, current measurement techniques remain largely limited in scope in regard to polymeric drug delivery systems. This paper will primarily describe the influence of water content, pH, and temperature on protein and peptide stability within polymer-based delivery systems. Moreover, we will review current instrumentation used to quantify factors affecting protein/peptide stability with respect to water content, pH, and temperature. Lastly, we will outline several recommendations to help guide future research efforts to develop methods more specific to quantifying protein/peptide stability within polymer-based delivery systems.
Collapse
Affiliation(s)
- Whitney Nicole Souery
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 101 Bizzell St., College Station, TX 77843, USA.
| | - Shreedevi Arun Kumar
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 101 Bizzell St., College Station, TX 77843, USA.
| | - Daniel Prasca-Chamorro
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 101 Bizzell St., College Station, TX 77843, USA.
| | - David Mitchell Moore
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 101 Bizzell St., College Station, TX 77843, USA.
| | - Jacob Good
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 101 Bizzell St., College Station, TX 77843, USA.
| | - Corey J Bishop
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 101 Bizzell St., College Station, TX 77843, USA.
| |
Collapse
|
85
|
Chaparro FJ, Presley KF, Coutinho da Silva MA, Mandan N, Colachis ML, Posner M, Arnold RM, Fan F, Moraes CR, Lannutti JJ. Sintered electrospun poly(ɛ‐caprolactone)–poly(ethylene terephthalate) for drug delivery. J Appl Polym Sci 2019. [DOI: 10.1002/app.47731] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Francisco J. Chaparro
- Department of Materials Science and EngineeringThe Ohio State University 2041 College Road, Columbus Ohio 43210
| | - Kayla F. Presley
- Department of Materials Science and EngineeringThe Ohio State University 2041 College Road, Columbus Ohio 43210
| | - Marco A. Coutinho da Silva
- Department of Veterinary Clinical SciencesThe Ohio State University 601 Vernon Tharp Street, Columbus Ohio 43210
| | - Nayan Mandan
- Department of Materials Science and EngineeringThe Ohio State University 2041 College Road, Columbus Ohio 43210
| | - Matthew L. Colachis
- Department of Materials Science and EngineeringThe Ohio State University 2041 College Road, Columbus Ohio 43210
| | - Michael Posner
- Department of Materials Science and EngineeringThe Ohio State University 2041 College Road, Columbus Ohio 43210
| | - Ryan M. Arnold
- Department of Materials Science and EngineeringThe Ohio State University 2041 College Road, Columbus Ohio 43210
| | - Fan Fan
- Department of Materials Science and EngineeringThe Ohio State University 2041 College Road, Columbus Ohio 43210
| | - Christa R. Moraes
- Department of Veterinary Clinical SciencesThe Ohio State University 601 Vernon Tharp Street, Columbus Ohio 43210
| | - John J. Lannutti
- Department of Materials Science and EngineeringThe Ohio State University 2041 College Road, Columbus Ohio 43210
| |
Collapse
|
86
|
O'Neill E, Rajpura K, Carbone EJ, Awale G, Kan HM, Lo KWH. Repositioning Tacrolimus: Evaluation of the Effect of Short-Term Tacrolimus Treatment on Osteoprogenitor Cells and Primary Cells for Bone Regenerative Engineering. Assay Drug Dev Technol 2019; 17:77-88. [DOI: 10.1089/adt.2018.876] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Edward O'Neill
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
| | - Komal Rajpura
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Connecticut Institute for Clinical and Translational Science, University of Connecticut Health Center, Farmington, Connecticut
| | - Erica J. Carbone
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- UConn Stem Cell Institute, University of Connecticut Health Center, Farmington, Connecticut
| | - Guleid Awale
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
| | - Ho-Man Kan
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Department of Orthopaedic Surgery, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
| | - Kevin W.-H. Lo
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Connecticut Institute for Clinical and Translational Science, University of Connecticut Health Center, Farmington, Connecticut
- UConn Stem Cell Institute, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
87
|
van Dijk F, Teekamp N, Post E, Schuppan D, Kim YO, Zuidema J, Steendam R, Klose MHM, Meier-Menches SM, Casini A, Horvatovich PL, Sijbrandi NJ, Frijlink HW, Hinrichs WLJ, Poelstra K, Beljaars L, Olinga P. The antifibrotic potential of a sustained release formulation of a PDGFβ-receptor targeted rho kinase inhibitor. J Control Release 2019; 296:250-257. [PMID: 30682444 DOI: 10.1016/j.jconrel.2018.12.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 11/29/2018] [Accepted: 12/21/2018] [Indexed: 12/22/2022]
Abstract
Rho kinase activity in hepatic stellate cells (HSCs) is associated with activation, transformation and contraction of these cells, leading to extracellular matrix production and portal hypertension in liver cirrhosis. Inhibition of rho kinase activity can reduce these activities, but may also lead to side effects, for instance systemic hypotension. This can be circumvented by liver-specific delivery of a rho kinase inhibitor to effector cells. Therefore, we targeted the rho kinase inhibitor Y27632 to the key pathogenic cells in liver fibrosis, i.e. myofibroblasts including activated HSCs that highly express the PDGFβ-receptor, using the drug carrier pPB-MSA. This carrier consists of mouse serum albumin (MSA) covalently coupled to several PDGFβR-recognizing moieties (pPB). We aimed to create a prolonged release system of such a targeted construct, by encapsulating pPB-MSA-Y27632 in biodegradable polymeric microspheres, thereby reducing short-lasting peak concentrations and the need for frequent administrations. Firstly, we confirmed the vasodilating potency of PDGFβ-receptor targeted Y27632 in vitro in a contraction assay using HSCs seeded on a collagen gel. We subsequently demonstrated the in vivo antifibrotic efficacy of pPB-MSA-Y27632-loaded microspheres in the Mdr2-/- mouse model of progressive biliary liver fibrosis. A single subcutaneous microsphere administration followed by organ harvest one week later clearly attenuated liver fibrosis progression and significantly suppressed the expression of fibrosis related genes, such as several collagens, profibrotic cytokines and matrix metalloproteinases. In conclusion, we demonstrate that polymeric microspheres are suitable as drug delivery system for the sustained systemic delivery of targeted protein constructs with antifibrotic potential, such as pPB-MSA-Y27632. This formulation appears suitable for the sustained treatment of liver fibrosis and possibly other chronic diseases.
Collapse
Affiliation(s)
- F van Dijk
- Groningen Research Institute of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands; Groningen Research Institute of Pharmacy, Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands
| | - N Teekamp
- Groningen Research Institute of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - E Post
- Groningen Research Institute of Pharmacy, Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands
| | - D Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Y O Kim
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - J Zuidema
- InnoCore Pharmaceuticals, Groningen, The Netherlands
| | - R Steendam
- InnoCore Pharmaceuticals, Groningen, The Netherlands
| | - Matthias H M Klose
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria
| | - Samuel M Meier-Menches
- School of Chemistry, Cardiff University, Park Place, CF10 3AT, Cardiff, UK; Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria
| | - A Casini
- School of Chemistry, Cardiff University, Park Place, CF10 3AT, Cardiff, UK
| | - P L Horvatovich
- Groningen Research Institute of Pharmacy, Department of Analytical Biochemistry, University of Groningen, Groningen, the Netherlands
| | | | - H W Frijlink
- Groningen Research Institute of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - W L J Hinrichs
- Groningen Research Institute of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - K Poelstra
- Groningen Research Institute of Pharmacy, Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands
| | - L Beljaars
- Groningen Research Institute of Pharmacy, Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands
| | - P Olinga
- Groningen Research Institute of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
88
|
Sequeira JAD, Santos AC, Serra J, Estevens C, Seiça R, Veiga F, Ribeiro AJ. Subcutaneous delivery of biotherapeutics: challenges at the injection site. Expert Opin Drug Deliv 2019; 16:143-151. [PMID: 30632401 DOI: 10.1080/17425247.2019.1568408] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Biotherapeutics are primarily delivered subcutaneously due to better compliance and prolonged rate of absorption compared to other parenteral administration routes. Recent research has allowed for the development of biotherapeutic formulations for subcutaneous delivery that require a lower frequency of administration by increasing drug half-life. Formulations determine shelf-life stability as well as features and transient behaviors that influence stability once implanted in the subcutaneous space. AREAS COVERED This review provides an overview of the factors affecting subcutaneous absorption with a focus on transient effects at the injection site following administration of biotherapeutics and the subsequent impact on absorption and stability. EXPERT OPINION Advances have been made in understanding subcutaneous tissue and the complex interplay of factors that regulate its homeostasis. The issue of poor stability after injection has been neglected, and many biotherapeutics are hampered by low bioavailability. With the advent of new in vitro techniques that account for properties of the injection site, stability studies evaluating subcutaneous tissues and impacts on pharmacokinetics of biotherapeutics may be useful in the development of new formulations.
Collapse
Affiliation(s)
- Joana A D Sequeira
- a Department of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal
| | - Ana C Santos
- a Department of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,b REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal
| | - João Serra
- c Tecnimede , Sociedade Técnico-Medicinal , Sintra , Portugal
| | | | - Raquel Seiça
- d Institute of Physiology, Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Francisco Veiga
- a Department of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,b REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal
| | - António J Ribeiro
- a Department of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,e i3S, Group Genetics of Cognitive Dysfunction , Institute for Molecular and Cell Biology , Porto , Portugal
| |
Collapse
|
89
|
Schlesinger EB, Bernards DA, Chen HH, Feindt J, Cao J, Dix D, Romano C, Bhisitkul RB, Desai TA. Device design methodology and formulation of a protein therapeutic for sustained release intraocular delivery. Bioeng Transl Med 2019; 4:152-163. [PMID: 30680326 PMCID: PMC6336666 DOI: 10.1002/btm2.10121] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/28/2022] Open
Abstract
Despite years of effort, sustained delivery of protein therapeutics remains an unmet need due to three primary challenges - dose, duration, and stability. The work presented here provides a design methodology for polycaprolactone reservoir-based thin film devices suitable for long-acting protein delivery to the back of the eye. First, the challenge of formulating highly concentrated protein in a device reservoir was addressed by improving stability with solubility-reducing excipients. Next, predictive correlations between design parameters and device performance were developed to provide a methodology to achieve a target product profile. Prototype devices were designed using this methodology to achieve desired device size, release rate, therapeutic payload, and protein stability, assessed by in vitro studies. Finally, prototype tolerability was established in a non-human primate model. The design methodology presented here is widely applicable to reservoir-based sustained delivery devices for proteins and provides a general device design framework.
Collapse
Affiliation(s)
- Erica B. Schlesinger
- Graduate Program in BioengineeringUniversity of CaliforniaSan FranciscoCA 94158
- Formulation Development GroupRegeneron PharmaceuticalsTarrytownNY 10591
| | - Daniel A. Bernards
- Dept. of Bioengineering and Therapeutic SciencesUniversity of CaliforniaSan FranciscoCA 94158
| | - Hunter H. Chen
- Formulation Development GroupRegeneron PharmaceuticalsTarrytownNY 10591
| | - James Feindt
- Formulation Development GroupRegeneron PharmaceuticalsTarrytownNY 10591
| | - Jingtai Cao
- Ophthalmology ResearchRegeneron PharmaceuticalsTarrytownNY 10591
| | - Daniel Dix
- Formulation Development GroupRegeneron PharmaceuticalsTarrytownNY 10591
| | - Carmelo Romano
- Ophthalmology ResearchRegeneron PharmaceuticalsTarrytownNY 10591
| | | | - Tejal A. Desai
- Graduate Program in BioengineeringUniversity of CaliforniaSan FranciscoCA 94158
- Dept. of Bioengineering and Therapeutic SciencesUniversity of CaliforniaSan FranciscoCA 94158
| |
Collapse
|
90
|
Xue K, Zhao X, Zhang Z, Qiu B, Tan QSW, Ong KH, Liu Z, Parikh BH, Barathi VA, Yu W, Wang X, Lingam G, Hunziker W, Su X, Loh XJ. Sustained delivery of anti-VEGFs from thermogel depots inhibits angiogenesis without the need for multiple injections. Biomater Sci 2019; 7:4603-4614. [DOI: 10.1039/c9bm01049a] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Polyurethane thermogels show sustained delivery of bioactive anti-VEGFs therapeutics to the eye.
Collapse
|
91
|
A Novel Biodegradable and Thermosensitive Poly(Ester-Amide) Hydrogel for Cartilage Tissue Engineering. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2710892. [PMID: 30662902 PMCID: PMC6313982 DOI: 10.1155/2018/2710892] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/18/2018] [Indexed: 01/09/2023]
Abstract
Thermosensitive hydrogels are attractive alternative scaffolding materials for minimally invasive surgery through a simple injection and in situ gelling. In this study, a novel poly(ester-amide) polymer, methoxy poly(ethylene glycol)-poly(pyrrolidone-co-lactide) (mPDLA, P3L7) diblock copolymer, was synthesized and characterized for cartilage tissue engineering. A series of amphiphilic diblock copolymers was synthesized by ring-opening polymerization of mPEG 550, D,L-lactide, and 2-pyrrolidone. By dynamic light scattering analysis and tube-flipped-upside-down method, viscoelastic properties of the mPDLA diblock copolymer solution exhibited sol-gel transition behavior as a function of temperature. An in vitro degradation assay showed that degradation acidity was effectively reduced by introducing the 2-pyrrolidone monomer into the polyester hydrogel. Besides, mPDLA exhibited great biocompatibility in vitro for cell encapsulation due to a high swelling ratio. Moreover, cell viability and biochemical analysis proved that the mPDLA hydrogel presented a great chondrogenic response. Taken together, these results demonstrate that mPDLA hydrogels are promising injectable scaffolds potentially applicable to cartilage tissue engineering.
Collapse
|
92
|
Sankaran S, Del Campo A. Optoregulated Protein Release from an Engineered Living Material. ACTA ACUST UNITED AC 2018; 3:e1800312. [PMID: 32627372 DOI: 10.1002/adbi.201800312] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/04/2018] [Indexed: 12/20/2022]
Abstract
Developing materials to encapsulate and deliver functional proteins inside the body is a challenging yet rewarding task for therapeutic purposes. High production costs, mostly associated with the purification process, short-term stability in vivo, and controlled and prolonged release are major hurdles for the clinical application of protein-based biopharmaceuticals. In an attempt to overcome these hurdles, herein, the possibility of incorporating bacteria as protein factories into a material and externally controlling protein release using optogenetics is demonstrated. By engineering bacteria to express and secrete a red fluorescent protein in response to low doses of blue light irradiation and embedding them in agarose hydrogels, living materials are fabricated capable of releasing proteins into the surrounding medium when exposed to light. These bacterial hydrogels allow spatially confined protein expression and dosed protein release over several weeks, regulated by the area and extent of light exposure. The possibility of incorporating such complex functions in a material using relatively simple material and genetic engineering strategies highlights the immense potential and versatility offered by living materials for protein-based biopharmaceutical delivery.
Collapse
Affiliation(s)
| | - Aránzazu Del Campo
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany.,Chemistry Department, Saarland University, 66123, Saarbrücken, Germany
| |
Collapse
|
93
|
Shen X, Li Q, Wang F, Bao J, Dai M, Zheng H, Lao X. Generation of a novel long-acting thymosin alpha1-Fc fusion protein and its efficacy for the inhibition of breast cancer in vivo. Biomed Pharmacother 2018; 108:610-617. [DOI: 10.1016/j.biopha.2018.09.064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/09/2018] [Accepted: 09/11/2018] [Indexed: 12/21/2022] Open
|
94
|
Zhu S, Wang J, He Y, Meng N, Yan GR. Peptides/Proteins Encoded by Non-coding RNA: A Novel Resource Bank for Drug Targets and Biomarkers. Front Pharmacol 2018; 9:1295. [PMID: 30483132 PMCID: PMC6243196 DOI: 10.3389/fphar.2018.01295] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
Non-coding RNAs (ncRNAs) are defined as RNA molecules that do not encode proteins, but recent evidence has proven that peptides/proteins encoded by ncRNAs do indeed exist and usually contain less than 100 amino acids. These peptides/proteins play an important role in regulating tumor energy metabolism, epithelial to mesenchymal transition of cancer cells, the stability of the c-Myc oncoprotein, and the ubiquitination and degradation of proliferating cell nuclear antigen (PCNA). These peptides/proteins represent promising drug targets for fighting against tumor growth or biomarkers for predicting the prognosis of cancer patients. In this review, we summarize the characteristics of peptides/proteins that have recently been identified as putative ncRNA translation products and their outlook for small molecule peptide drugs, drug targets, and biomarkers.
Collapse
Affiliation(s)
- Song Zhu
- Biomedicine Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Jizhong Wang
- Biomedicine Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Yutian He
- Biomedicine Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Nan Meng
- Biomedicine Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Guang-Rong Yan
- Biomedicine Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
95
|
Pudlarz A, Szemraj J. Nanoparticles as Carriers of Proteins, Peptides and Other Therapeutic Molecules. Open Life Sci 2018; 13:285-298. [PMID: 33817095 PMCID: PMC7874720 DOI: 10.1515/biol-2018-0035] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/08/2018] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles have many applications both in industry and medicine. Depending upon their physical and chemical properties, they can be used as carriers of therapeutic molecules or as therapeutics. Nanoparticles are made of synthetic or natural polymers, lipids or metals. Their use allows for faster transport to the place of action, thus prolonging its presence in the body and limiting side effects. In addition, the use of such a drug delivery system protects the drug from rapid disintegration and elimination from the body. In recent years, the use of proteins and peptides as therapeutic molecules has grown significantly. Unfortunately, proteins are subject to enzymatic digestion and can cause unwanted immune response beyond therapeutic action. The use of drug carriers can minimize undesirable side effects and reduce the dose of medication needed to achieve the therapeutic effect. The current study presents the use of several selected drug delivery systems for the delivery of proteins, peptides and other therapeutic molecules.
Collapse
Affiliation(s)
- Agnieszka Pudlarz
- Department of Medical Biochemistry, Medical University of Lodz, Lodz, Poland
- E-mail:
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
96
|
Liu X, Wang C, Liu Z. Protein-Engineered Biomaterials for Cancer Theranostics. Adv Healthc Mater 2018; 7:e1800913. [PMID: 30260583 DOI: 10.1002/adhm.201800913] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/19/2018] [Indexed: 12/18/2022]
Abstract
Proteins are an important class of biomaterials promising a variety of applications such as drug delivery, and imaging or therapy, owing to their biodegradability, biocompatibility, as well as inherent biological activities acting as enzymes, recognizing molecules, or therapeutics by themselves. Over the few past decades, different types of proteins with desired properties have been widely explored for biomedical applications. Many therapeutic proteins have now entered clinical use. This review therefore summarizes various strategies in the engineering of biomaterials for delivery of therapeutic proteins, as well as the recent development of protein-based biomaterials for cancer theranostics.
Collapse
Affiliation(s)
- Xiaowen Liu
- Pharmacology; Department of Basic Medical Sciences; Faculty of Medical Science; Jinan University; Guangzhou Guangdong 510632 China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM); Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices; Soochow University; Suzhou Jiangsu 215123 China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM); Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices; Soochow University; Suzhou Jiangsu 215123 China
| |
Collapse
|
97
|
Clements AEB, Groves ER, Chamberlain CS, Vanderby R, Murphy WL. Microparticles Locally Deliver Active Interleukin-1 Receptor Antagonist In Vivo. Adv Healthc Mater 2018; 7:e1800263. [PMID: 29974661 DOI: 10.1002/adhm.201800263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/23/2018] [Indexed: 11/11/2022]
Abstract
Despite significant research in therapeutic protein delivery, localized and sustained delivery of active therapeutic proteins remains a challenge. Delivery is a particular challenge for therapeutic proteins with a short half-life. Herein, localized delivery of interleukin-1 receptor antagonist (IL-1Ra) by mineral coated microparticles (MPs) is assessed in a healing rat medial collateral ligament (MCL). The local tissue concentration and systemic serum concentration of IL-1Ra, the anti-inflammatory activity of IL-1Ra delivered with MPs, and whether IL-1Ra loaded MPs (IL-1Ra MPs) are immunogenic in a healing ligament are also examined. IL-1Ra MPs significantly increase the local concentration of IL-1Ra compared to soluble IL-1Ra at 7 and 14 days after treatment but do not elevate the systemic concentration of IL-1Ra at these time points, indicating localized delivery of IL-1Ra. IL-1Ra MPs significantly reduce inflammation caused by the MPs themselves, indicating the IL-1Ra is active. Finally, IL-1Ra MPs do not induce a foreign body response and decrease the immunogenicity of human IL-1Ra in a healing rat MCL. Overall, mineral coated microparticles have the ability to locally deliver active therapeutic proteins for an extended period of time.
Collapse
Affiliation(s)
- Anna E. B. Clements
- University of Wisconsin; Madison, 1111 Highland Ave., 5405 WIMR II Madison WI 53705 USA
| | - Emily R. Groves
- University of Wisconsin; Madison, 1111 Highland Ave., 5405 WIMR II Madison WI 53705 USA
| | - Connie S. Chamberlain
- University of Wisconsin; Madison, 1111 Highland Ave., 5405 WIMR II Madison WI 53705 USA
| | - Ray Vanderby
- University of Wisconsin; Madison, 1111 Highland Ave., 5405 WIMR II Madison WI 53705 USA
| | - William L. Murphy
- University of Wisconsin; Madison, 1111 Highland Ave., 5405 WIMR II Madison WI 53705 USA
| |
Collapse
|
98
|
Andrianov AK, Marin A, Martinez AP, Weidman JL, Fuerst TR. Hydrolytically Degradable PEGylated Polyelectrolyte Nanocomplexes for Protein Delivery. Biomacromolecules 2018; 19:3467-3478. [PMID: 29953203 DOI: 10.1021/acs.biomac.8b00785] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Novel oppositely charged polyphosphazene polyelectrolytes containing grafted poly(ethylene glycol) (PEG) chains were synthesized as modular components for the assembly of biodegradable PEGylated protein delivery vehicles. These macromolecular counterparts, which contained either carboxylic acid or tertiary amino groups, were then formulated at near physiological conditions into supramolecular assemblies of nanoscale level, below 100 nm. Nanocomplexes with electroneutral surface charge, as assessed by zeta potential measurements, were stable in aqueous solutions, which suggests their compact polyelectrolyte complex "core"-hydrophilic PEG "shell" structure. Investigation of PEGylated polyphosphazene nanocomplexes as agents for noncovalent PEGylation of the therapeutic protein l-asparaginase (L-ASP) in vitro demonstrated their ability to dramatically reduce protein antigenicity, as measured by antibody binding using enzyme linked immunosorbent assay (ELISA). Encapsulation in nanocomplexes did not affect enzymatic activity of L-ASP, but improved its thermal stability and proteolytic resistance. Gel permeation chromatography (GPC) experiments revealed that all synthesized polyphosphazenes exhibited composition controlled hydrolytic degradability in aqueous solutions at neutral pH and showed greater stability at lower temperatures. Overall, novel hydrolytically degradable polyphosphazene polyelectrolytes capable of spontaneous self-assembly into PEGylated nanoparticulates in aqueous solutions can potentially enable a simple and effective approach to modifying therapeutic proteins without the need for their covalent modification.
Collapse
Affiliation(s)
- Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research , University of Maryland , 9600 Gudelsky Drive , Rockville , Maryland 20850 , United States
| | - Alexander Marin
- Institute for Bioscience and Biotechnology Research , University of Maryland , 9600 Gudelsky Drive , Rockville , Maryland 20850 , United States
| | - Andre P Martinez
- Institute for Bioscience and Biotechnology Research , University of Maryland , 9600 Gudelsky Drive , Rockville , Maryland 20850 , United States
| | - Jacob L Weidman
- Institute for Bioscience and Biotechnology Research , University of Maryland , 9600 Gudelsky Drive , Rockville , Maryland 20850 , United States
| | - Thomas R Fuerst
- Institute for Bioscience and Biotechnology Research , University of Maryland , 9600 Gudelsky Drive , Rockville , Maryland 20850 , United States.,Department of Cell Biology and Molecular Genetics , 1109 Microbiology Building , University of Maryland , College Park , Maryland 20742 , United States
| |
Collapse
|
99
|
Awwad S, Angkawinitwong U. Overview of Antibody Drug Delivery. Pharmaceutics 2018; 10:E83. [PMID: 29973504 PMCID: PMC6161251 DOI: 10.3390/pharmaceutics10030083] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/29/2018] [Accepted: 06/29/2018] [Indexed: 12/11/2022] Open
Abstract
Monoclonal antibodies (mAbs) are one of the most important classes of therapeutic proteins, which are used to treat a wide number of diseases (e.g., oncology, inflammation and autoimmune diseases). Monoclonal antibody technologies are continuing to evolve to develop medicines with increasingly improved safety profiles, with the identification of new drug targets being one key barrier for new antibody development. There are many opportunities for developing antibody formulations for better patient compliance, cost savings and lifecycle management, e.g., subcutaneous formulations. However, mAb-based medicines also have limitations that impact their clinical use; the most prominent challenges are their short pharmacokinetic properties and stability issues during manufacturing, transport and storage that can lead to aggregation and protein denaturation. The development of long acting protein formulations must maintain protein stability and be able to deliver a large enough dose over a prolonged period. Many strategies are being pursued to improve the formulation and dosage forms of antibodies to improve efficacy and to increase the range of applications for the clinical use of mAbs.
Collapse
Affiliation(s)
- Sahar Awwad
- UCL School of Pharmacy, London WC1N 1AX, UK.
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1 V9EL, UK.
| | | |
Collapse
|
100
|
Leconet W, Liu H, Guo M, Le Lamer-Déchamps S, Molinier C, Kim S, Vrlinic T, Oster M, Liu F, Navarro V, Batra JS, Noriega AL, Grizot S, Bander NH. Anti-PSMA/CD3 Bispecific Antibody Delivery and Antitumor Activity Using a Polymeric Depot Formulation. Mol Cancer Ther 2018; 17:1927-1940. [DOI: 10.1158/1535-7163.mct-17-1138] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 04/05/2018] [Accepted: 06/04/2018] [Indexed: 11/16/2022]
|