351
|
Zhou Z, Liu X, Zhu D, Wang Y, Zhang Z, Zhou X, Qiu N, Chen X, Shen Y. Nonviral cancer gene therapy: Delivery cascade and vector nanoproperty integration. Adv Drug Deliv Rev 2017; 115:115-154. [PMID: 28778715 DOI: 10.1016/j.addr.2017.07.021] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
Gene therapy represents a promising cancer treatment featuring high efficacy and limited side effects, but it is stymied by a lack of safe and efficient gene-delivery vectors. Cationic polymers and lipid-based nonviral gene vectors have many advantages and have been extensively explored for cancer gene delivery, but their low gene-expression efficiencies relative to viral vectors limit their clinical translations. Great efforts have thus been devoted to developing new carrier materials and fabricating functional vectors aimed at improving gene expression, but the overall efficiencies are still more or less at the same level. This review analyzes the cancer gene-delivery cascade and the barriers, the needed nanoproperties and the current strategies for overcoming these barriers, and outlines PEGylation, surface-charge, size, and stability dilemmas in vector nanoproperties to efficiently accomplish the cancer gene-delivery cascade. Stability, surface, and size transitions (3S Transitions) are proposed to resolve those dilemmas and strategies to realize these transitions are comprehensively summarized. The review concludes with a discussion of the future research directions to design high-performance nonviral gene vectors.
Collapse
Affiliation(s)
- Zhuxian Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xiangrui Liu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Dingcheng Zhu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Yue Wang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Zhen Zhang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xuefei Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Nasha Qiu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xuesi Chen
- Changchun Institute of Applied Chemistry, Key Lab of Polymer Ecomaterials, Changchun, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China.
| |
Collapse
|
352
|
Schmøkel J, Voldum A, Tsakiridou G, Kuhlmann M, Cameron J, Sørensen ES, Wengel J, Howard KA. Site-selective conjugation of an anticoagulant aptamer to recombinant albumins and maintenance of neonatal Fc receptor binding. NANOTECHNOLOGY 2017; 28:204004. [PMID: 28362634 DOI: 10.1088/1361-6528/aa6a9b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Aptamers are an attractive molecular medicine that offers high target specificity. Nucleic acid-based aptamers, however, are prone to nuclease degradation and rapid renal excretion that require blood circulatory half-life extension enabling technologies. The long circulatory half-life, predominately facilitated by engagement with the cellular recycling neonatal Fc receptor (FcRn), and ligand transport properties of albumin promote it as an attractive candidate to improve the pharmacokinetic profile of aptamers. This study investigates the effect of Cys34 site-selective covalent attachment of a factor IXa anticoagulant aptamer on aptamer functionality and human FcRn (hFcRn) engagement using recombinant human albumin (rHA) of either a wild type (WT) or an engineered human FcRn high binding variant (HB). Albumin-aptamer conjugates, connected covalently through a heterobifunctional succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate linker, were successfully prepared and purified by high performance liquid chromatography as confirmed by gel electrophoresis band-shift analysis and matrix-assisted laser desorption/ionization time of flight. Minimal reduction (∼25%) in activity of WT-linked aptamer to that of aptamer alone was found using an anticoagulant activity assay measuring temporal levels of activated partial thrombin. Covalent albumin-aptamer conjugation, however, substantially compromized binding to hFcRn, to 10% affinity of that of non-conjugated WT, determined by biolayer interferometry. Binding could be rescued by aptamer conjugation to recombinant albumin engineered for higher FcRn affinity (HB) that exhibited an 8-fold affinity compared to WT alone. This work describes a novel albumin-based aptamer delivery system whose hFcRn binding can be increased using a HB engineered albumin.
Collapse
Affiliation(s)
- Julie Schmøkel
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
353
|
Influence of Defined Hydrophilic Blocks within Oligoaminoamide Copolymers: Compaction versus Shielding of pDNA Nanoparticles. Polymers (Basel) 2017; 9:polym9040142. [PMID: 30970822 PMCID: PMC6432433 DOI: 10.3390/polym9040142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/07/2017] [Accepted: 04/11/2017] [Indexed: 01/07/2023] Open
Abstract
Cationic polymers are promising components of the versatile platform of non-viral nucleic acid (NA) delivery agents. For a successful gene delivery system, these NA vehicles need to comprise several functionalities. This work focuses on the modification of oligoaminoamide carriers with hydrophilic oligomer blocks mediating nanoparticle shielding potential, which is necessary to prevent aggregation or dissociation of NA polyplexes in vitro, and hinder opsonization with blood components in vivo. Herein, the shielding agent polyethylene glycol (PEG) in three defined lengths (12, 24, or 48 oxyethylene repeats) is compared with two peptidic shielding blocks composed of four or eight repeats of sequential proline-alanine-serine (PAS). With both types of shielding agents, we found opposing effects of the length of hydrophilic segments on shielding and compaction of formed plasmid DNA (pDNA) nanoparticles. Two-arm oligoaminoamides with 37 cationizable nitrogens linked to 12 oxyethylene units or four PAS repeats resulted in very compact 40⁻50 nm pDNA nanoparticles, whereas longer shielding molecules destabilize the investigated polyplexes. Thus, the balance between sufficiently shielded but still compact and stable particles can be considered a critical optimization parameter for non-viral nucleic acid vehicles based on hydrophilic-cationic block oligomers.
Collapse
|
354
|
Tsuruta JK, Schaub NP, Rojas JD, Streeter J, Klauber-DeMore N, Dayton P. Optimizing ultrasound molecular imaging of secreted frizzled related protein 2 expression in angiosarcoma. PLoS One 2017; 12:e0174281. [PMID: 28333964 PMCID: PMC5363853 DOI: 10.1371/journal.pone.0174281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/05/2017] [Indexed: 11/19/2022] Open
Abstract
Secreted frizzled related protein 2 (SFRP2) is a tumor endothelial marker expressed in angiosarcoma. Previously, we showed ultrasound molecular imaging with SFRP2-targeted contrast increased average video pixel intensity (VI) of angiosarcoma vessels by 2.2 ± 0.6 VI versus streptavidin contrast. We hypothesized that redesigning our contrast agents would increase imaging performance. Improved molecular imaging reagents were created by combining NeutrAvidin™-functionalized microbubbles with biotinylated SFRP2 or IgY control antibodies. When angiosarcoma tumors in nude mice reached 8 mm, time-intensity, antibody loading, and microbubble dose experiments optimized molecular imaging. 10 minutes after injection, the control-subtracted time-intensity curve (TIC) for SFRP2-targeted contrast reached a maximum, after subtracting the contribution of free-flowing contrast. SFRP2 antibody-targeted VI was greater when contrast was formulated with 10-fold molar excess of maleimide-activated NeutrAvidin™ versus 3-fold (4.5 ± 0.18 vs. 0.32 ± 0.15, VI ± SEM, 5 x 106 dose, p < 0.001). Tumor vasculature returned greater average video pixel intensity using 5 x 107 versus 5 x 106 microbubbles (21.2 ± 2.5 vs. 4.5 ± 0.18, p = 0.0011). Specificity for tumor vasculature was confirmed by low VI for SFRP2-targeted, and control contrast in peri-tumoral vasculature (3.2 ± 0.52 vs. 1.6 ± 0.71, p = 0.92). After optimization, average video pixel intensity of tumor vasculature was 14.2 ± 3.0 VI units higher with SFRP2-targeted contrast versus IgY-targeted control (22.1 ± 2.5 vs. 7.9 ± 1.6, p < 0.001). After log decompression, 14.2 ΔVI was equal to ~70% higher signal, in arbitray acoustic units (AU), for SFRP2 versus IgY. This provided ~18- fold higher acoustic signal enhancement than provided previously by 2.2 ΔVI. Basing our targeted contrast on NeutrAvidin™-functionalized microbubbles, using IgY antibodies for our control contrast, and optimizing our imaging protocol significantly increased the SFRP2-specific signal returned from angiosarcoma vasculature, and may provide new opportunities for targeted molecular imaging.
Collapse
Affiliation(s)
- James K. Tsuruta
- Joint Department of Biomedical Engineering, North Carolina State University, and The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Nicholas P. Schaub
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Juan D. Rojas
- Joint Department of Biomedical Engineering, North Carolina State University, and The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jason Streeter
- Joint Department of Biomedical Engineering, North Carolina State University, and The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Nancy Klauber-DeMore
- Department of Surgery, Medical College of South Carolina, Charleston, South Carolina, United States of America
| | - Paul Dayton
- Joint Department of Biomedical Engineering, North Carolina State University, and The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
355
|
Bell SM, Wendt DJ, Zhang Y, Taylor TW, Long S, Tsuruda L, Zhao B, Laipis P, Fitzpatrick PA. Formulation and PEGylation optimization of the therapeutic PEGylated phenylalanine ammonia lyase for the treatment of phenylketonuria. PLoS One 2017; 12:e0173269. [PMID: 28282402 PMCID: PMC5345807 DOI: 10.1371/journal.pone.0173269] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 02/11/2017] [Indexed: 01/03/2023] Open
Abstract
Phenylketonuria (PKU) is a genetic metabolic disease in which the decrease or loss of phenylalanine hydroxylase (PAH) activity results in elevated, neurotoxic levels of phenylalanine (Phe). Due to many obstacles, PAH enzyme replacement therapy is not currently an option. Treatment of PKU with an alternative enzyme, phenylalanine ammonia lyase (PAL), was first proposed in the 1970s. However, issues regarding immunogenicity, enzyme production and mode of delivery needed to be overcome. Through the evaluation of PAL enzymes from multiple species, three potential PAL enzymes from yeast and cyanobacteria were chosen for evaluation of their therapeutic potential. The addition of polyethylene glycol (PEG, MW = 20,000), at a particular ratio to modify the protein surface, attenuated immunogenicity in an animal model of PKU. All three PEGylated PAL candidates showed efficacy in a mouse model of PKU (BTBR Pahenu2) upon subcutaneous injection. However, only PEGylated Anabaena variabilis (Av) PAL-treated mice demonstrated sustained low Phe levels with weekly injection and was the only PAL evaluated that maintained full enzymatic activity upon PEGylation. A PEGylated recombinant double mutant version of AvPAL (Cys503Ser/Cys565Ser), rAvPAL-PEG, was selected for drug development based on its positive pharmacodynamic profile and favorable expression titers. PEGylation was shown to be critical for rAvPAL-PEG efficacy as under PEGylated rAvPAL had a lower pharmacodynamic effect. rAvPAL and rAvPAL-PEG had poor stability at 4°C. L-Phe and trans-cinnamate were identified as activity stabilizing excipients. rAvPAL-PEG is currently in Phase 3 clinical trials to assess efficacy in PKU patients.
Collapse
Affiliation(s)
- Sean M. Bell
- BioMarin Pharmaceutical, Novato, California, United States of America
- * E-mail:
| | - Dan J. Wendt
- BioMarin Pharmaceutical, Novato, California, United States of America
| | - Yanhong Zhang
- BioMarin Pharmaceutical, Novato, California, United States of America
| | - Timothy W. Taylor
- BioMarin Pharmaceutical, Novato, California, United States of America
| | - Shinong Long
- BioMarin Pharmaceutical, Novato, California, United States of America
| | - Laurie Tsuruda
- BioMarin Pharmaceutical, Novato, California, United States of America
| | - Bin Zhao
- BioMarin Pharmaceutical, Novato, California, United States of America
| | - Phillip Laipis
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, United States of America
| | | |
Collapse
|
356
|
Magarkar A, Róg T, Bunker A. A computational study suggests that replacing PEG with PMOZ may increase exposure of hydrophobic targeting moiety. Eur J Pharm Sci 2017; 103:128-135. [PMID: 28285174 DOI: 10.1016/j.ejps.2017.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/28/2017] [Accepted: 03/07/2017] [Indexed: 12/18/2022]
Abstract
In a previous study we showed that the cause of failure of a new, proposed, targeting ligand, the AETP moiety, when attached to a PEGylated liposome, was occlusion by the poly(ethylene glycol) (PEG) layer due to its hydrophobic nature, given that PEG is not entirely hydrophilic. At the time we proposed that possible replacement with a more hydrophilic protective polymer could alleviate this problem. In this study we have used computational molecular dynamics modelling, using a model with all atom resolution, to suggest that a specific alternative protective polymer, poly(2-methyloxazoline) (PMOZ), would perform exactly this function. Our results show that when PEG is replaced by PMOZ the relative exposure to the solvent of AETP is increased to a level even greater than that we found in previous simulations for the RGD peptide, a targeting moiety that has previously been used successfully in PEGylated liposome based therapies. While the AETP moiety itself is no longer under consideration, the results of this computational study have broader significance: the use of PMOZ as an alternative polymer coating to PEG could be efficacious in the context of more hydrophobic targeting ligands. In addition to PMOZ we studied another polyoxazoline, poly(2-ethyloxazoline) (PEOZ), that has also been mooted as a possible alternate protective polymer. It was also found that the RDG peptide occlusion was significantly greater for the case of both oxazolines as opposed to PEG and that, unlike PEG, neither oxazoline entered the membrane. As far as we are aware this is the first time that polyoxazolines have been studied using molecular dynamics simulation with all atom resolution.
Collapse
Affiliation(s)
- Aniket Magarkar
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland; Institute of Organic Chemistry and Biochemistry, Academy of the Sciences of the Czech Republic Prague, Czech Republic
| | - Tomasz Róg
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Alex Bunker
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
357
|
|
358
|
Carmali S, Murata H, Cummings C, Matyjaszewski K, Russell AJ. Polymer-Based Protein Engineering: Synthesis and Characterization of Armored, High Graft Density Polymer-Protein Conjugates. Methods Enzymol 2017; 590:347-380. [PMID: 28411645 DOI: 10.1016/bs.mie.2016.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Atom transfer radical polymerization (ATRP) from the surface of a protein can generate remarkably dense polymer shells that serve as armor and rationally tune protein function. Using straightforward chemistry, it is possible to covalently couple or display multiple small molecule initiators onto a protein surface. The chemistry is fine-tuned to be sequence specific (if one desires a single targeted site) at controlled density. Once the initiator is anchored on the protein surface, ATRP is used to grow polymers on protein surface, in situ. The technique is so powerful that a single-protein polymer conjugate molecule can contain more than 90% polymer coating by weight. If desired, stimuli-responsive polymers can be "grown" from the initiated sites to prepare enzyme conjugates that respond to external triggers such as temperature or pH, while still maintaining enzyme activity and stability. Herein, we focus mainly on the synthesis of chymotrypsin-polymer conjugates. Control of the number of covalently coupled initiator sites by changing the stoichiometric ratio between enzyme and the initiator during the synthesis of protein-initiator complexes allowed fine-tuning of the grafting density. For example, very high grafting density chymotrypsin conjugates were prepared from protein-initiator complexes to grow the temperature-responsive polymers, poly(N-isopropylacrylamide), and poly[N,N'-dimethyl(methacryloyloxyethyl) ammonium propane sulfonate]. Controlled growth of polymers from protein surfaces enables one to predictably manipulate enzyme kinetics and stability without the need for molecular biology-dependent mutagenesis.
Collapse
Affiliation(s)
- Sheiliza Carmali
- Center for Polymer-Based Protein Engineering, ICES, Carnegie Mellon University, Pittsburgh, PA, United States; Carnegie Mellon University, Pittsburgh, PA, United States
| | - Hironobu Murata
- Center for Polymer-Based Protein Engineering, ICES, Carnegie Mellon University, Pittsburgh, PA, United States; Carnegie Mellon University, Pittsburgh, PA, United States
| | - Chad Cummings
- Center for Polymer-Based Protein Engineering, ICES, Carnegie Mellon University, Pittsburgh, PA, United States; Carnegie Mellon University, Pittsburgh, PA, United States
| | - Krzysztof Matyjaszewski
- Center for Polymer-Based Protein Engineering, ICES, Carnegie Mellon University, Pittsburgh, PA, United States; Carnegie Mellon University, Pittsburgh, PA, United States
| | - Alan J Russell
- Center for Polymer-Based Protein Engineering, ICES, Carnegie Mellon University, Pittsburgh, PA, United States; Carnegie Mellon University, Pittsburgh, PA, United States.
| |
Collapse
|
359
|
Schwartz S. Unmet needs in developing nanoparticles for precision medicine. Nanomedicine (Lond) 2017; 12:271-274. [PMID: 28093937 DOI: 10.2217/nnm-2016-0390] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Simo Schwartz
- Molecular Biology & Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Barcelona, Spain.,Networking Research Centre for Bioengineering, Biomaterials, & Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza, Spain
| |
Collapse
|
360
|
Nie Q, Jia D, Yang H, Feng Y, Fan Q, Shi Q, Wan L, Lu X. Conjugation to 10 kDa Linear PEG Extends Serum Half-Life and Preserves the Receptor-Binding Ability of mmTRAIL with Minimal Stimulation of PEG-Specific Antibodies. Mol Pharm 2017; 14:502-512. [PMID: 28029256 DOI: 10.1021/acs.molpharmaceut.6b00964] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The poor in vivo potencies of most therapeutic proteins might be attributed to their short serum half-lives. PEGylation is a well-established method and has been clinically proven to improve pharmacokinetics. mmTRAIL exhibited supercytotoxicity in a variety of tumor cells, but its serum half-life was less than 10 min in mice. Here, mmTRAIL-5K, mmTRAIL-10K, and mmTRAIL-20K were produced by N-terminus-specific PEGylation of mmTRAIL with 5, 10, or 20 kDa mPEG, respectively. The particle sizes of mmTRAIL-5K, mmTRAIL-10K, and mmTRAIL-20K were 9.09 ± 2.76, 12.62 ± 4.05, and 15.68 ± 4.95 nm, which were higher than the threshold (∼7 nm) of renal clearance. Accordingly, mmTRAIL-5K exhibited a serum half-life of 30 min only 3 times longer than that of mmTRAIL. However, both mmTRAIL-10K and mmTRAIL-20K exhibited similar serum half-lives ranging from 350 to 400 min, indicating that PEGylation with 10 or 20 kDa mPEG significantly improved the pharmacokinetics of mmTRAIL. However, death receptor binding of mmTRAIL-20K was reduced 5- to 8-fold, resulting in a 3-fold reduction of cytotoxicity. Additionally, repeated administration of mmTRAIL-20K elicited both mPEG-specific IgG and IgM antibody responses in rats. In contrast, the receptor binding and cytotoxicity of mmTRAIL-10K were similar to those of mmTRAIL. Repeated administration of mmTRAIL-10K did not obviously stimulate mPEG-specific antibody responses in rats and rhesus monkeys. Of the three PEGylated mmTRAIL analogues, mmTRAIL-10K exerted the greatest tumor suppression in mice bearing human tumor xenografts. These results demonstrated that conjugation of mmTRAIL to 10 kDa mPEG was better than that to 5 or 20 kDa mPEG for enhancing antitumor effects.
Collapse
Affiliation(s)
- Qianxue Nie
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Dianlong Jia
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Hao Yang
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Yanru Feng
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Qing Fan
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Qiuxiao Shi
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Lin Wan
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Xiaofeng Lu
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| |
Collapse
|
361
|
Abstract
The past decade has witnessed explosive growth in the development of nanoparticle-based therapies for the treatment of neurological disorders and diseases. The systemic delivery of therapeutic carriers to the central nervous system (CNS) is hindered by both the blood-brain barrier (BBB) and the porous and electrostatically charged brain extracellular matrix (ECM), which acts as a steric and adhesive barrier. Therapeutic delivery to the brain is influenced by changes in the brain microenvironment, which can occur as a function of physiology, biology, pathology, and developmental age. Brain-penetrating nanoparticles (BPNs) are an optimal platform not only for therapeutic delivery to the brain, but also for evaluating changes in the brain microenvironment. BPNs possess both the capability to readily move within their local environment to survey their surroundings and the ability to reach the diffuse disease cells often associated with CNS disorders. To achieve effective delivery of BPNs to specific locations within the brain requires careful control over the nanoparticle's transport properties. Here, we describe the process of conjugating a dense layer of poly(ethylene glycol) (PEG) to the surface of nonbiodegradable nanoparticles to achieve brain-penetrating capabilities.
Collapse
Affiliation(s)
- Elizabeth Nance
- Department of Chemical Engineering, University of Washington, 105 Benson Hall, Box 351750, Seattle, WA, 98195, USA.
| |
Collapse
|
362
|
Abstract
mRNA vaccines elicit a potent immune response including antibodies and cytotoxic T cells. mRNA vaccines are currently evaluated in clinical trials for cancer immunotherapy applications, but also have great potential as prophylactic vaccines. Efficient delivery of mRNA vaccines will be key for their success and translation to the clinic. Among potential nonviral vectors, lipid nanoparticles are particularly promising. Indeed, lipid nanoparticles can be synthesized with relative ease in a scalable manner, protect the mRNA against degradation, facilitate endosomal escape, can be targeted to the desired cell type by surface decoration with ligands, and as needed, can be codelivered with adjuvants.
Collapse
|
363
|
Yang Q, Jacobs TM, McCallen JD, Moore DT, Huckaby JT, Edelstein JN, Lai SK. Analysis of Pre-existing IgG and IgM Antibodies against Polyethylene Glycol (PEG) in the General Population. Anal Chem 2016; 88:11804-11812. [PMID: 27804292 DOI: 10.1021/acs.analchem.6b03437] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Circulating antibodies (Ab) that specifically bind polyethylene glycol (PEG), a biocompatible polymer routinely used in protein and nanoparticle therapeutics, have been associated with reduced efficacy of and/or adverse reactions to therapeutics modified with or containing PEG. Unlike most antidrug antibodies that are induced following initial drug dosing, anti-PEG Ab can be found in treatment-naïve individuals (i.e., individuals who have never undergone treatment with PEGylated drugs but most likely have been exposed to PEG through other means). Unfortunately, the true prevalence, quantitative levels, and Ab isotype of pre-existing anti-PEG Ab remain poorly understood. Here, using rigorously validated competitive ELISAs with engineered chimeric anti-PEG monoclonal Ab standards, we quantified the levels of anti-PEG IgM and different subclasses of anti-PEG IgG (IgG1-4) in both contemporary and historical human samples. We unexpectedly found, with 90% confidence, detectable levels of anti-PEG Ab in ∼72% of the contemporary specimens (18% IgG, 25% IgM, 30% both IgG and IgM). The vast majority of these samples contained low levels of anti-PEG Ab, with only ∼7% and ∼1% of all specimens possessing anti-PEG IgG and IgM in excess of 500 ng/mL, respectively. IgG2 was the predominant anti-PEG IgG subclass. Anti-PEG Ab's were also observed in ∼56% of serum samples collected during 1970-1999 (20% IgG, 19% IgM, and 16% both IgG and IgM), suggesting that the presence of PEG-specific antibodies may be a longstanding phenomenon. Anti-PEG IgG levels demonstrated correlation with patient age, but not with gender or race. The widespread prevalence of pre-existing anti-PEG Ab, coupled with high Ab levels in a subset of the population, underscores the potential importance of screening patients for anti-PEG Ab levels prior to administration of therapeutics containing PEG.
Collapse
Affiliation(s)
- Qi Yang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Timothy M Jacobs
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Justin D McCallen
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Dominic T Moore
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Justin T Huckaby
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Jasmine N Edelstein
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Samuel K Lai
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
364
|
Engineered Nanomedicine with Alendronic Acid Corona Improves Targeting to Osteosarcoma. Sci Rep 2016; 6:36707. [PMID: 27824143 PMCID: PMC5099855 DOI: 10.1038/srep36707] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/19/2016] [Indexed: 12/29/2022] Open
Abstract
We engineered nanomedicine with the stealth corona made up of densely packed bone seeking ligand, alendronic acid. In a typical nanoconstruct, alendronic acid is conjugated with hydrophilic head moiety of phospholipid that has an ability to self-assemble with hydrophobic polymeric core through its hydrophobic long carbon-chain. Proposed nanomedicine has three distinct compartments namely; poly(l-lactic-co-glycolic acid) polymeric core acting as a drug reservoir and skeleton of the nanoconstruct, phospholipid monolayer covers the core acting as a diffusion barrier, and a densely packed alendronic acid corona acting as a stabilizer and targeting moiety. Thus engineered nanomedicine attain spherical entity with ~90 ± 6 nm having negative zeta potential, −37.7 ± 2 mV, and has an ability to load 7 ± 0.3 wt% of doxorubicin. In-vitro bone targeting efficiency of nanomedicine was studied using hydroxyapatite crystals as a bone model, and found significant accumulation of nanoparticle in the crystals. Moreover, cellular internalization studies with mouse osteosarcoma confirm the selectivity of nanomedicine when compared to its internalization in non-targeted mouse melanoma. This nanomedicine shows prolong stability in serum and deliver the drug into the cell exhibiting an IC50 of 3.7 μM. Given the strong interacting property of alendronic acid with bone, the proposed nanomedicine hold promises in delivering drug to bone microenvironment.
Collapse
|
365
|
Gabizon AA, Patil Y, La-Beck NM. New insights and evolving role of pegylated liposomal doxorubicin in cancer therapy. Drug Resist Updat 2016; 29:90-106. [DOI: 10.1016/j.drup.2016.10.003] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/16/2016] [Accepted: 10/24/2016] [Indexed: 12/16/2022]
|
366
|
Abstract
Treatment and management of kidney disease currently presents an enormous global burden, and the application of nanotechnology principles to renal disease therapy, although still at an early stage, has profound transformative potential. The increasing translation of nanomedicines to the clinic, alongside research efforts in tissue regeneration and organ-on-a-chip investigations, are likely to provide novel solutions to treat kidney diseases. Our understanding of renal anatomy and of how the biological and physico-chemical properties of nanomedicines (the combination of a nanocarrier and a drug) influence their interactions with renal tissues has improved dramatically. Tailoring of nanomedicines in terms of kidney retention and binding to key membranes and cell populations associated with renal diseases is now possible and greatly enhances their localization, tolerability, and efficacy. This Review outlines nanomedicine characteristics central to improved targeting of renal cells and highlights the prospects, challenges, and opportunities of nanotechnology-mediated therapies for renal diseases.
Collapse
|
367
|
Chen BM, Su YC, Chang CJ, Burnouf PA, Chuang KH, Chen CH, Cheng TL, Chen YT, Wu JY, Roffler SR. Measurement of Pre-Existing IgG and IgM Antibodies against Polyethylene Glycol in Healthy Individuals. Anal Chem 2016; 88:10661-10666. [PMID: 27726379 DOI: 10.1021/acs.analchem.6b03109] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Polyethylene glycol (PEG) is a biocompatible polymer that is often attached to therapeutic molecules to improve bioavailability and therapeutic efficacy. Although antibodies with specificity for PEG may compromise the safety and effectiveness of PEGylated medicines, the prevalence of pre-existing anti-PEG antibodies in healthy individuals is unclear. Chimeric human anti-PEG antibody standards were created to accurately measure anti-PEG IgM and IgG antibodies by direct ELISA with confirmation by a competition assay in the plasma of 1504 healthy Han Chinese donors residing in Taiwan. Anti-PEG antibodies were detected in 44.3% of healthy donors with a high prevalence of both anti-PEG IgM (27.1%) and anti-PEG IgG (25.7%). Anti-PEG IgM and IgG antibodies were significantly more common in females as compared to males (32.0% vs 22.2% for IgM, p < 0.0001 and 28.3% vs 23.0% for IgG, p = 0.018). The prevalence of anti-PEG IgG antibodies was higher in younger (up to 60% for 20 year olds) as opposed to older (20% for >50 years) male and female donors. Anti-PEG IgG concentrations were negatively associated with donor age in both females (p = 0.0073) and males (p = 0.026). Both anti-PEG IgM and IgG strongly bound PEGylated medicines. The described assay can assist in the elucidation of the impact of anti-PEG antibodies on the safety and therapeutic efficacy of PEGylated medicines.
Collapse
Affiliation(s)
- Bing-Mae Chen
- Institute of Biomedical Sciences, Academia Sinica , Taipei 11529, Taiwan
| | - Yu-Cheng Su
- Institute of Biomedical Sciences, Academia Sinica , Taipei 11529, Taiwan
| | - Chia-Jung Chang
- Institute of Biomedical Sciences, Academia Sinica , Taipei 11529, Taiwan
| | | | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University , Taipei 11031, Taiwan
| | - Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica , Taipei 11529, Taiwan.,School of Chinese Medicine, China Medical University , Taichung 40447, Taiwan
| | - Tian-Lu Cheng
- Department of Biomedical Science and Environmental Biology, Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University , Kaohsiung 80708, Taiwan
| | - Yuan-Tsong Chen
- Institute of Biomedical Sciences, Academia Sinica , Taipei 11529, Taiwan.,Department of Pediatrics, Duke University Medical Center , Durham, North Carolina 27710, United States
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica , Taipei 11529, Taiwan.,School of Chinese Medicine, China Medical University , Taichung 40447, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica , Taipei 11529, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University , Kaohsiung 80708, Taiwan
| |
Collapse
|
368
|
Bunker A, Magarkar A, Viitala T. Rational design of liposomal drug delivery systems, a review: Combined experimental and computational studies of lipid membranes, liposomes and their PEGylation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2334-2352. [DOI: 10.1016/j.bbamem.2016.02.025] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/09/2016] [Accepted: 02/10/2016] [Indexed: 01/22/2023]
|
369
|
Xie L, Wang G, Zhou H, Zhang F, Guo Z, Liu C, Zhang X, Zhu L. Functional long circulating single walled carbon nanotubes for fluorescent/photoacoustic imaging-guided enhanced phototherapy. Biomaterials 2016; 103:219-228. [DOI: 10.1016/j.biomaterials.2016.06.058] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 06/10/2016] [Accepted: 06/23/2016] [Indexed: 12/29/2022]
|
370
|
Henry CE, Wang YY, Yang Q, Hoang T, Chattopadhyay S, Hoen T, Ensign LM, Nunn KL, Schroeder H, McCallen J, Moench T, Cone R, Roffler SR, Lai SK. Anti-PEG antibodies alter the mobility and biodistribution of densely PEGylated nanoparticles in mucus. Acta Biomater 2016; 43:61-70. [PMID: 27424083 DOI: 10.1016/j.actbio.2016.07.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/24/2016] [Accepted: 07/12/2016] [Indexed: 11/18/2022]
Abstract
UNLABELLED Antibodies that specifically bind polyethylene glycol (PEG) can lead to rapid elimination of PEGylated therapeutics from the systemic circulation. We have recently shown that virus-binding IgG can immobilize viruses in mucus via multiple low-affinity crosslinks between IgG and mucins. However, it remains unclear whether anti-PEG antibodies in mucus may also alter the penetration and consequently biodistribution of PEGylated nanoparticles delivered to mucosal surfaces. We found that both anti-PEG IgG and IgM can readily bind nanoparticles that were densely coated with PEG polymer to minimize adhesive interactions with mucus constituents. Addition of anti-PEG IgG and IgM into mouse cervicovaginal mucus resulted in extensive trapping of mucus-penetrating PEGylated nanoparticles, with the fraction of mobile particles reduced from over 95% to only 34% and 7% with anti-PEG IgG and IgM, respectively. Surprisingly, we did not observe significant agglutination induced by either antibody, suggesting that particle immobilization is caused by adhesive crosslinks between mucin fibers and IgG or IgM bound to individual nanoparticles. Importantly, addition of corresponding control antibodies did not slow the PEGylated nanoparticles, confirming anti-PEG antibodies specifically bound to and trapped the PEGylated nanoparticles. Finally, we showed that trapped PEGylated nanoparticles remained largely in the luminal mucus layer of the mouse vagina even when delivered in hypotonic formulations that caused untrapped particles to be drawn by the flow of water (advection) through mucus all the way to the epithelial surface. These results underscore the potential importance of elucidating mucosal anti-PEG immune responses for PEGylated therapeutics and biomaterials applied to mucosal surfaces. STATEMENT OF SIGNIFICANCE PEG, generally considered a 'stealth' polymer, is broadly used to improve the circulation times and therapeutic efficacy of nanomedicines. Nevertheless, there is increasing scientific evidence that demonstrates both animals and humans can generate PEG-specific antibodies. Here, we show that anti-PEG IgG and IgM can specifically immobilize otherwise freely diffusing PEG-coated nanoparticles in fresh vaginal mucus gel ex vivo by crosslinking nanoparticles to the mucin mesh, and consequently prevent PEG-coated nanoparticles from accessing the vaginal epithelium in vivo. Given the increasing use of PEG coatings to enhance nanoparticle penetration of mucosal barriers, our findings demonstrate that anti-PEG immunity may be a potential concern not only for systemic drug delivery but also for mucosal drug delivery.
Collapse
Affiliation(s)
- Christine E Henry
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina - Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Ying-Ying Wang
- Department of Biophysics, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Qi Yang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina - Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Thuy Hoang
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | - Sumon Chattopadhyay
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | - Timothy Hoen
- Department of Biophysics, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Laura M Ensign
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | - Kenetta L Nunn
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina - Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Holly Schroeder
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina - Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Justin McCallen
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina - Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Thomas Moench
- ReProtect, Inc., 703 Stags Head Road, Baltimore, MD 21286, USA
| | - Richard Cone
- Department of Biophysics, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, No. 128, Section 2, Academia Road, Taipei 11529, Taiwan
| | - Samuel K Lai
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina - Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC 27599, USA; UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina - Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC 27599, USA; Department of Microbiology & Immunology, University of North Carolina - Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC 27599, USA.
| |
Collapse
|
371
|
The Mucus Barrier to Inhaled Gene Therapy. Mol Ther 2016; 24:2043-2053. [PMID: 27646604 DOI: 10.1038/mt.2016.182] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 09/07/2016] [Indexed: 12/21/2022] Open
Abstract
Recent evidence suggests that the airway mucus gel layer may be impermeable to the viral and synthetic gene vectors used in past inhaled gene therapy clinical trials for diseases like cystic fibrosis. These findings support the logic that inhaled gene vectors that are incapable of penetrating the mucus barrier are unlikely to provide meaningful benefit to patients. In this review, we discuss the biochemical and biophysical features of mucus that contribute its barrier function, and how these barrier properties may be reinforced in patients with lung disease. We next review biophysical techniques used to assess the potential ability of gene vectors to penetrate airway mucus. Finally, we provide new data suggesting that fresh human airway mucus should be used to test the penetration rates of gene vectors. The physiological barrier properties of spontaneously expectorated CF sputum remained intact up to 24 hours after collection when refrigerated at 4 °C. Conversely, the barrier properties were significantly altered after freezing and thawing of sputum samples. Gene vectors capable of overcoming the airway mucus barrier hold promise as a means to provide the widespread gene transfer throughout the airway epithelium required to achieve meaningful patient outcomes in inhaled gene therapy clinical trials.
Collapse
|
372
|
Feng Q, Tong R. Anticancer nanoparticulate polymer-drug conjugate. Bioeng Transl Med 2016; 1:277-296. [PMID: 29313017 PMCID: PMC5689533 DOI: 10.1002/btm2.10033] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/17/2016] [Accepted: 08/26/2016] [Indexed: 12/11/2022] Open
Abstract
We review recent progress in polymer-drug conjugate for cancer nanomedicine. Polymer-drug conjugates, including the nanoparticle prepared from these conjugates, are designed to release drug in tumor tissues or cells in order to improve drugs' therapeutic efficacy. We summarize general design principles for the polymer-drug conjugate, including the synthetic strategies, the design of the chemical linkers between the drug and polymer in the conjugate, and the in vivo drug delivery barriers for polymer-drug conjugates. Several new strategies, such as the synthesis of polymer-drug conjugates and supramolecular-drug conjugates, the use of stimulus-responsive delivery, and triggering the change of the nanoparticle physiochemical properties to over delivery barriers, are also highlighted.
Collapse
Affiliation(s)
- Quanyou Feng
- Dept. of Chemical EngineeringVirginia Polytechnic Institute and State University635 Prices Fork RoadBlacksburgVA24061
| | - Rong Tong
- Dept. of Chemical EngineeringVirginia Polytechnic Institute and State University635 Prices Fork RoadBlacksburgVA24061
| |
Collapse
|
373
|
Kim HJ, Kim A, Miyata K, Kataoka K. Recent progress in development of siRNA delivery vehicles for cancer therapy. Adv Drug Deliv Rev 2016; 104:61-77. [PMID: 27352638 DOI: 10.1016/j.addr.2016.06.011] [Citation(s) in RCA: 313] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 05/21/2016] [Accepted: 06/16/2016] [Indexed: 12/13/2022]
Abstract
Recent progress in RNA biology has broadened the scope of therapeutic targets of RNA drugs for cancer therapy. However, RNA drugs, typically small interfering RNAs (siRNAs), are rapidly degraded by RNases and filtrated in the kidney, thereby requiring a delivery vehicle for efficient transport to the target cells. To date, various delivery formulations have been developed from cationic lipids, polymers, and/or inorganic nanoparticles for systemic delivery of siRNA to solid tumors. This review describes the current status of clinical trials related to siRNA-based cancer therapy, as well as the remaining issues that need to be overcome to establish a successful therapy. It, then introduces various promising design strategies of delivery vehicles for stable and targeted siRNA delivery, including the prospects for future design.
Collapse
|
374
|
Curtis C, Zhang M, Liao R, Wood T, Nance E. Systems-level thinking for nanoparticle-mediated therapeutic delivery to neurological diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27562224 DOI: 10.1002/wnan.1422] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/27/2016] [Accepted: 07/17/2016] [Indexed: 12/27/2022]
Abstract
Neurological diseases account for 13% of the global burden of disease. As a result, treating these diseases costs $750 billion a year. Nanotechnology, which consists of small (~1-100 nm) but highly tailorable platforms, can provide significant opportunities for improving therapeutic delivery to the brain. Nanoparticles can increase drug solubility, overcome the blood-brain and brain penetration barriers, and provide timed release of a drug at a site of interest. Many researchers have successfully used nanotechnology to overcome individual barriers to therapeutic delivery to the brain, yet no platform has translated into a standard of care for any neurological disease. The challenge in translating nanotechnology platforms into clinical use for patients with neurological disease necessitates a new approach to: (1) collect information from the fields associated with understanding and treating brain diseases and (2) apply that information using scalable technologies in a clinically-relevant way. This approach requires systems-level thinking to integrate an understanding of biological barriers to therapeutic intervention in the brain with the engineering of nanoparticle material properties to overcome those barriers. To demonstrate how a systems perspective can tackle the challenge of treating neurological diseases using nanotechnology, this review will first present physiological barriers to drug delivery in the brain and common neurological disease hallmarks that influence these barriers. We will then analyze the design of nanotechnology platforms in preclinical in vivo efficacy studies for treatment of neurological disease, and map concepts for the interaction of nanoparticle physicochemical properties and pathophysiological hallmarks in the brain. WIREs Nanomed Nanobiotechnol 2017, 9:e1422. doi: 10.1002/wnan.1422 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Chad Curtis
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Mengying Zhang
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
| | - Rick Liao
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Thomas Wood
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA.,Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA.,Department of Radiology, University of Washington, Seattle, WA, USA.,Center on Human Development and Disability, University of Washington, Seattle, WA, USA
| |
Collapse
|
375
|
Wang H, Cheng F, Shen W, Cheng G, Zhao J, Peng W, Qu J. Amino acid-based anti-fouling functionalization of silica nanoparticles using divinyl sulfone. Acta Biomater 2016; 40:273-281. [PMID: 27032480 DOI: 10.1016/j.actbio.2016.03.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/17/2016] [Accepted: 03/22/2016] [Indexed: 01/03/2023]
Abstract
UNLABELLED Natural amino acids are zwitterionic molecules and the good biocompatibility promises them potential candidates as anti-fouling materials. Here, we developed a new method to functionalize silica nanoparticles with a natural amino acid-based anti-fouling layer. Amino acids were covalently immobilized on 3-aminopropyltriethoxysilane modified silica nanoparticles using divinyl sulfone through a two-step reaction in aqueous solution at room temperature. The progress was monitored with NMR, X-ray photoelectron spectroscopy (XPS), transmission electron microscope (TEM) and zeta potential measurements. A library of amino acids was screened and the nonspecific protein adsorption of bovine serum albumin (BSA) and fetal bovine serum (FBS) were investigated using dynamic light scattering method. The results showed that cysteine, lysine and arginine functionalized silica nanoparticles can effectively resist protein adsorption due to the zwitterionic structure. Among them, lysine functionalized silica nanoparticles had the best anti-fouling performance, which showed hydrodynamic diameter increases of only 10% after incubated in BSA solution and 20% after incubated in FBS solution for 24h. The neat aqueous modification process can conveniently create a thin zwitterionic layer on silica particles, and it has a great potential in biomolecule immobilization and biofunctional surface preparation. STATEMENT OF SIGNIFICANCE Zwitterionic polymer is an outstanding class of anti-fouling material; but the difficulty in synthesis is challenging its spread utilization. In this study, we developed a new method to create an amino acid-based zwitterionic layer on APTES functionalized silica nanoparticles through a two-step reaction in aqueous solution at room temperature. The surface chemistry was monitored with NMR, XPS, TEM and zeta potential measurements. With this method, a library of amino acid conjugated-silica nanoparticles was synthesized and their anti-fouling performance was evaluated using dynamic light scattering method. The results showed that the cysteine, lysine and arginine conjugated nanoparticles all can effectively resist nonspecific protein adsorption. Among them, lysine conjugated nanoparticles show the best anti-fouling performance, which showed hydrodynamic diameter increases of only 10% after incubated in BSA solution and 20% after incubated in FBS solution for 24 hours. These results indicates that the anti-fouling silica nanoparticles are of great potential in many biomedical applications, especially biosensing and diagnose imaging. The modification reactions in aqueous solution at room temperature are easily conducted in laboratory, indicating high potential in the functionalization of silica particles/surfaces with other biomolecules.
Collapse
|
376
|
Abd Ellah NH, Abouelmagd SA. Surface functionalization of polymeric nanoparticles for tumor drug delivery: approaches and challenges. Expert Opin Drug Deliv 2016; 14:201-214. [DOI: 10.1080/17425247.2016.1213238] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Noura H. Abd Ellah
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Sara A. Abouelmagd
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
377
|
Zhang P, Sun F, Liu S, Jiang S. Anti-PEG antibodies in the clinic: Current issues and beyond PEGylation. J Control Release 2016; 244:184-193. [PMID: 27369864 DOI: 10.1016/j.jconrel.2016.06.040] [Citation(s) in RCA: 410] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/15/2016] [Accepted: 06/27/2016] [Indexed: 12/14/2022]
Abstract
The technique of attaching the polymer polyethylene glycol (PEG), or PEGylation, has brought more than ten protein drugs into market. The surface conjugation of PEG on proteins prolongs their blood circulation time and reduces immunogenicity by increasing their hydrodynamic size and masking surface epitopes. Despite this success, an emerging body of literature highlights the presence of antibodies produced by the immune system that specifically recognize and bind to PEG (anti-PEG Abs), including both pre-existing and treatment-induced Abs. More importantly, the existence of anti-PEG Abs has been correlated with loss of therapeutic efficacy and increase in adverse effects in several clinical reports examining different PEGylated therapeutics. To better understand the nature of anti-PEG immunity, we summarize a number of clinical reports and some critical animal studies regarding pre-existing and treatment-induced anti-PEG Abs. Various anti-PEG detection methods used in different studies were provided. Several protein modification technologies beyond PEGylation were also highlighted.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, United States
| | - Fang Sun
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, United States
| | - Sijun Liu
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Shaoyi Jiang
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, United States; Department of Bioengineering, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
378
|
Pham TTH, Rombouts WH, Fokkink R, Stuart MCA, Cohen Stuart MA, Kleijn JM. Nanoparticle-Templated Formation and Growth Mechanism of Curved Protein Polymer Fibrils. Biomacromolecules 2016; 17:2392-8. [PMID: 27250876 DOI: 10.1021/acs.biomac.6b00486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We investigated the growth of biosynthetic protein polymers with templated curvature on pluronic nanospheres. The protein has a central silk-like block containing glutamic residues (S(E)) and collagen-like end-blocks (C). The S(E) blocks stack into filaments when their charge is removed (pH <5). Indeed, at low pH curved and circular fibers are formed at the surface of the nanospheres, which keep their shape after removal of the pluronics. The data reveal the mechanism of the templated fibril-growth: The growth of protein assemblies is nucleated in solution; small protein fibrils adsorb on the nanospheres, presumably due to hydrogen bond formation between the silk-like blocks and the pluronic PEO blocks. The surface of the pluronic particles templates further growth. At relatively low protein/pluronic weight ratios, only a fraction of the nanospheres bears protein fibers, pointing to a limiting amount of nuclei in solution. Because the nanospheres capture fibrils at an early stage of growth, they can be used to separate growth and nucleation rates in protein fibril formation. Moreover, the nanoparticle-templated growth of stable curved fibers opens ways to build proteinaceous nanocapsules from designed protein polymers.
Collapse
Affiliation(s)
- Thao T H Pham
- Physical Chemistry and Soft Matter, Wageningen University , P.O. Box 8038, NL-6700 EK Wageningen, The Netherlands
| | - Wolf H Rombouts
- Physical Chemistry and Soft Matter, Wageningen University , P.O. Box 8038, NL-6700 EK Wageningen, The Netherlands
| | - Remco Fokkink
- Physical Chemistry and Soft Matter, Wageningen University , P.O. Box 8038, NL-6700 EK Wageningen, The Netherlands
| | - Marc C A Stuart
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Nijenborgh 7, NL-9747 AG Groningen, The Netherlands
| | - Martien A Cohen Stuart
- Physical Chemistry and Soft Matter, Wageningen University , P.O. Box 8038, NL-6700 EK Wageningen, The Netherlands
| | - J Mieke Kleijn
- Physical Chemistry and Soft Matter, Wageningen University , P.O. Box 8038, NL-6700 EK Wageningen, The Netherlands
| |
Collapse
|
379
|
Nimjee SM, Povsic TJ, Sullenger BA, Becker RC. Translation and Clinical Development of Antithrombotic Aptamers. Nucleic Acid Ther 2016; 26:147-55. [PMID: 26882082 PMCID: PMC4900189 DOI: 10.1089/nat.2015.0581] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 01/12/2016] [Indexed: 01/02/2023] Open
Abstract
Thrombosis is a necessary physiological process to protect the body from uncontrolled bleeding. Pathological thrombus formation can lead to devastating clinical events including heart attack, stroke, deep vein thrombosis, pulmonary embolism, and disseminated intravascular coagulation. Numerous drugs have been developed to inhibit thrombosis. These have been targeted to coagulation factors along with proteins and receptors that activate platelets. While these drugs are effective at preventing blood clotting, their major side effect is inadvertent hemorrhage that can result in significant morbidity and mortality. There exists a need for anticoagulants that are not only effective at preventing thrombosis but can also be readily reversed. Aptamers offer a potential solution, representing a new class of drug agents that can be isolated to any protein and where antidote oligonucleotides can be designed based on the sequence of the aptamer. We present a summary of the anticoagulant and antithrombotic aptamers that have been identified and their stage of development and comment on the future of aptamer-based drug development to treat thrombosis.
Collapse
Affiliation(s)
- Shahid M. Nimjee
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Thomas J. Povsic
- Duke Clinical Research Institute, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Bruce A. Sullenger
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Richard C. Becker
- Department of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio
| |
Collapse
|
380
|
Bienk K, Hvam ML, Pakula MM, Dagnæs-Hansen F, Wengel J, Malle BM, Kragh-Hansen U, Cameron J, Bukrinski JT, Howard KA. An albumin-mediated cholesterol design-based strategy for tuning siRNA pharmacokinetics and gene silencing. J Control Release 2016; 232:143-51. [DOI: 10.1016/j.jconrel.2016.04.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/30/2016] [Accepted: 04/06/2016] [Indexed: 11/30/2022]
|
381
|
Mehdizadeh M, Rouhani H, Sepehri N, Varshochian R, Ghahremani MH, Amini M, Gharghabi M, Ostad SN, Atyabi F, Baharian A, Dinarvand R. Biotin decorated PLGA nanoparticles containing SN-38 designed for cancer therapy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:495-504. [DOI: 10.1080/21691401.2016.1178130] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Mozhdeh Mehdizadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Hasti Rouhani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
- Nano Alvand Co., Avicenna Tech Park, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Nima Sepehri
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
- Nano Alvand Co., Avicenna Tech Park, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Reyhaneh Varshochian
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Mohammad Hossein Ghahremani
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Mehdi Gharghabi
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Seyed Nasser Ostad
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Azin Baharian
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| |
Collapse
|
382
|
Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev 2016. [DOI: '10.1016/j.addr.2015.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
|
383
|
PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev 2016; 99:28-51. [PMID: 26456916 DOI: 10.1016/j.addr.2015.09.012] [Citation(s) in RCA: 2432] [Impact Index Per Article: 304.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 09/21/2015] [Accepted: 09/26/2015] [Indexed: 12/12/2022]
Abstract
Coating the surface of nanoparticles with polyethylene glycol (PEG), or "PEGylation", is a commonly used approach for improving the efficiency of drug and gene delivery to target cells and tissues. Building from the success of PEGylating proteins to improve systemic circulation time and decrease immunogenicity, the impact of PEG coatings on the fate of systemically administered nanoparticle formulations has, and continues to be, widely studied. PEG coatings on nanoparticles shield the surface from aggregation, opsonization, and phagocytosis, prolonging systemic circulation time. Here, we briefly describe the history of the development of PEGylated nanoparticle formulations for systemic administration, including how factors such as PEG molecular weight, PEG surface density, nanoparticle core properties, and repeated administration impact circulation time. A less frequently discussed topic, we then describe how PEG coatings on nanoparticles have also been utilized for overcoming various biological barriers to efficient drug and gene delivery associated with other modes of administration, ranging from gastrointestinal to ocular. Finally, we describe both methods for PEGylating nanoparticles and methods for characterizing PEG surface density, a key factor in the effectiveness of the PEG surface coating for improving drug and gene delivery.
Collapse
|
384
|
Kamaly N, Yameen B, Wu J, Farokhzad OC. Degradable Controlled-Release Polymers and Polymeric Nanoparticles: Mechanisms of Controlling Drug Release. Chem Rev 2016; 116:2602-63. [PMID: 26854975 PMCID: PMC5509216 DOI: 10.1021/acs.chemrev.5b00346] [Citation(s) in RCA: 1600] [Impact Index Per Article: 200.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nazila Kamaly
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Basit Yameen
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jun Wu
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Omid C. Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
385
|
Herzberger J, Niederer K, Pohlit H, Seiwert J, Worm M, Wurm FR, Frey H. Polymerization of Ethylene Oxide, Propylene Oxide, and Other Alkylene Oxides: Synthesis, Novel Polymer Architectures, and Bioconjugation. Chem Rev 2015; 116:2170-243. [PMID: 26713458 DOI: 10.1021/acs.chemrev.5b00441] [Citation(s) in RCA: 456] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The review summarizes current trends and developments in the polymerization of alkylene oxides in the last two decades since 1995, with a particular focus on the most important epoxide monomers ethylene oxide (EO), propylene oxide (PO), and butylene oxide (BO). Classical synthetic pathways, i.e., anionic polymerization, coordination polymerization, and cationic polymerization of epoxides (oxiranes), are briefly reviewed. The main focus of the review lies on more recent and in some cases metal-free methods for epoxide polymerization, i.e., the activated monomer strategy, the use of organocatalysts, such as N-heterocyclic carbenes (NHCs) and N-heterocyclic olefins (NHOs) as well as phosphazene bases. In addition, the commercially relevant double-metal cyanide (DMC) catalyst systems are discussed. Besides the synthetic progress, new types of multifunctional linear PEG (mf-PEG) and PPO structures accessible by copolymerization of EO or PO with functional epoxide comonomers are presented as well as complex branched, hyperbranched, and dendrimer like polyethers. Amphiphilic block copolymers based on PEO and PPO (Poloxamers and Pluronics) and advances in the area of PEGylation as the most important bioconjugation strategy are also summarized. With the ever growing toolbox for epoxide polymerization, a "polyether universe" may be envisaged that in its structural diversity parallels the immense variety of structural options available for polymers based on vinyl monomers with a purely carbon-based backbone.
Collapse
Affiliation(s)
- Jana Herzberger
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Graduate School Materials Science in Mainz , Staudingerweg 9, D-55128 Mainz, Germany
| | - Kerstin Niederer
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Hannah Pohlit
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Graduate School Materials Science in Mainz , Staudingerweg 9, D-55128 Mainz, Germany.,Max Planck Graduate Center , Staudingerweg 6, D-55128 Mainz, Germany.,Department of Dermatology, University Medical Center , Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Jan Seiwert
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Matthias Worm
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Max Planck Graduate Center , Staudingerweg 6, D-55128 Mainz, Germany
| | - Frederik R Wurm
- Max Planck Graduate Center , Staudingerweg 6, D-55128 Mainz, Germany.,Max Planck Institute for Polymer Research , Ackermannweg 10, D-55128 Mainz, Germany
| | - Holger Frey
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Graduate School Materials Science in Mainz , Staudingerweg 9, D-55128 Mainz, Germany
| |
Collapse
|
386
|
Pre-existing anti-polyethylene glycol antibody linked to first-exposure allergic reactions to pegnivacogin, a PEGylated RNA aptamer. J Allergy Clin Immunol 2015; 137:1610-1613.e7. [PMID: 26688515 PMCID: PMC5819876 DOI: 10.1016/j.jaci.2015.10.034] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/22/2015] [Accepted: 10/12/2015] [Indexed: 12/25/2022]
|
387
|
Seo Y, Schulz A, Han Y, He Z, Bludau H, Wan X, Tong J, Bronich TK, Sokolsky M, Luxenhofer R, Jordan R, Kabanov AV. Poly(2-oxazoline) block copolymer based formulations of taxanes: effect of copolymer and drug structure, concentration, and environmental factors. POLYM ADVAN TECHNOL 2015. [DOI: 10.1002/pat.3556] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Youngee Seo
- Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599-7362 USA
| | - Anita Schulz
- Department Chemie; Technische Universität Dresden; Zellescher Weg 19 01069 Dresden Germany
| | - Yingchao Han
- Center for Drug Delivery and Nanomedicine and Department of Pharmaceutical Sciences, College of Pharmacy; University of Nebraska Medical Center; Omaha NE 68198-5830 USA
| | - Zhijian He
- Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599-7362 USA
| | - Herdis Bludau
- Department Chemie; Technische Universität Dresden; Zellescher Weg 19 01069 Dresden Germany
| | - Xiaomeng Wan
- Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599-7362 USA
| | - Jing Tong
- Center for Drug Delivery and Nanomedicine and Department of Pharmaceutical Sciences, College of Pharmacy; University of Nebraska Medical Center; Omaha NE 68198-5830 USA
| | - Tatiana K. Bronich
- Center for Drug Delivery and Nanomedicine and Department of Pharmaceutical Sciences, College of Pharmacy; University of Nebraska Medical Center; Omaha NE 68198-5830 USA
| | - Marina Sokolsky
- Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599-7362 USA
| | - Robert Luxenhofer
- Functional Polymer Materials, Chair for Chemical Technology of Materials Synthesis, Department Chemistry and Pharmacy; Julius-Maximilians-Universität Würzburg; Röntgenring 11 97070 Würzburg Germany
| | - Rainer Jordan
- Department Chemie; Technische Universität Dresden; Zellescher Weg 19 01069 Dresden Germany
| | - Alexander V. Kabanov
- Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; University of North Carolina at Chapel Hill; Chapel Hill NC 27599-7362 USA
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry; M.V. Lomonosov Moscow State University; Moscow 119992 Russia
| |
Collapse
|