1
|
Thoma JL, Little H, Duhamel J. Location of a Hydrophobic Load in Poly(oligo(ethylene glycol) methyl ether methacrylate)s (PEGMAs) Dissolved in Water and Probed by Fluorescence. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:5900-5912. [PMID: 38442036 DOI: 10.1021/acs.langmuir.3c03802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Two series of pyrene-labeled poly(oligo(ethylene glycol) methyl ether methacrylate)s referred to as PyEG5-PEGnMA and PyC4-PEGnMA were prepared to probe the region surrounding the polymethacrylate backbone by using the fluorescence of the dye pyrene. PyEG5-PEGnMA and PyC4-PEGnMA were prepared by copolymerizing the EGnMA methacrylate monomers with penta(ethylene glycol) 1-pyrenemethyl ether methacrylate or 1-pyrenebutyl methacrylate, respectively. In organic solvents, the much longer 18 non-hydrogen atom linker connecting the pyrene moieties to the polymethacrylate backbone in the PyEG5-PEGnMA samples enabled the deployment of the pyrenyl labels into the solution. In water, however, an excited pyrene for PyEG5-PEGnMA was found to probe a same volume as for the PyC4-PEGnMA samples where a much shorter 6 non-hydrogen atom spacer connected pyrene to the backbone. Another surprising observation, considering that the hydrophobicity of pyrene induces strong pyrene aggregation for many pyrene-labeled water-soluble polymers (Py-WSPs) in water, was the little pyrene aggregation found for the PyEG5-PEGnMA and PyC4-PEGnMA samples in water. These effects could be related to the organic-like domain (OLD) generated by the oligo(ethylene glycol) side chains densely arranged around the polymethacrylate backbone of the polymeric bottlebrush (PBB). Additional fluorescence experiments conducted with the penta(ethylene glycol) 1-pyrenemethyl ether derivative indicated that the cylindrical OLD surrounding the polymethacrylate backbone had a chemical composition similar to that of ethylene glycol. Binding of hydrophobic pyrene molecules to unlabeled PEGnMA bottlebrushes in water further supported the existence of the OLD. The demonstration, that PEGnMA samples form an OLD in water, which can host and protect hydrophobic cargoes like pyrene, should lead to the development of improved PEGnMA-based drug delivery systems.
Collapse
Affiliation(s)
- Janine L Thoma
- Department of Chemistry, Institute for Polymer Research, Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Hunter Little
- Department of Chemistry, Institute for Polymer Research, Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Jean Duhamel
- Department of Chemistry, Institute for Polymer Research, Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
2
|
Hamelmann NM, Paulusse JMJ. Single-chain polymer nanoparticles in biomedical applications. J Control Release 2023; 356:26-42. [PMID: 36804328 DOI: 10.1016/j.jconrel.2023.02.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/02/2023] [Accepted: 02/13/2023] [Indexed: 02/23/2023]
Abstract
Single-chain polymer nanoparticles (SCNPs) are a well-defined and uniquely sized class of polymer nanoparticles. The advances in polymer science over the past decades have enabled the development of a variety of intramolecular crosslinking systems, leading to particles in the 5-20 nm size regime. Which is aligned with the size regime of proteins and therefore making SCNPs an interesting class of NPs for biomedical applications. The high modularity of SCNP design and the ease of their functionalization have led to growing research interest. In this review, we describe different crosslinking systems, as well as the preparation of functional SCNPs and the variety of biomedical applications that have been explored.
Collapse
Affiliation(s)
- Naomi M Hamelmann
- Department of Molecules and Materials, MESA+ Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, the Netherlands
| | - Jos M J Paulusse
- Department of Molecules and Materials, MESA+ Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, the Netherlands.
| |
Collapse
|
3
|
López CL, Brempelis KJ, Matthaei JF, Montgomery KS, Srinivasan S, Roy D, Huang F, Kreuser SA, Gardell JL, Blumenthal I, Chiefari J, Jensen MC, Crane CA, Stayton PS. Arming Immune Cell Therapeutics with Polymeric Prodrugs. Adv Healthc Mater 2022; 11:e2101944. [PMID: 34889072 PMCID: PMC9847575 DOI: 10.1002/adhm.202101944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/16/2021] [Indexed: 01/21/2023]
Abstract
Engineered immune cells are an exciting therapeutic modality, which survey and attack tumors. Backpacking strategies exploit cell targeting capabilities for delivery of drugs to combat tumors and their immune-suppressive environments. Here, a new platform for arming cell therapeutics through dual receptor and polymeric prodrug engineering is developed. Macrophage and T cell therapeutics are engineered to express a bioorthogonal single chain variable fragment receptor. The receptor binds a fluorescein ligand that directs cell loading with ligand-tagged polymeric prodrugs, termed "drugamers." The fluorescein ligand facilitates stable binding of drugamer to engineered macrophages over 10 days with 80% surface retention. Drugamers also incorporate prodrug monomers of the phosphoinositide-3-kinase inhibitor, PI-103. The extended release of PI-103 from the drugamer sustains antiproliferative activity against a glioblastoma cell line compared to the parent drug. The versatility and modularity of this cell arming system is demonstrated by loading T cells with a second fluorescein-drugamer. This drugamer incorporates a small molecule estrogen analog, CMP8, which stabilizes a degron-tagged transgene to provide temporal regulation of protein activity in engineered T cells. These results demonstrate that this bioorthogonal receptor and drugamer system can be used to arm multiple immune cell classes with both antitumor and transgene-activating small molecule prodrugs.
Collapse
Affiliation(s)
- Ciana L López
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA,Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Katherine J Brempelis
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - James F Matthaei
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Kate S Montgomery
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA
| | - Selvi Srinivasan
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA
| | - Debashish Roy
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA
| | - Fei Huang
- CSIRO Manufacturing, Bag 10, Bayview Avenue, Clayton, VIC. 3168, Australia
| | - Shannon A Kreuser
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Jennifer L Gardell
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Ian Blumenthal
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA,Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - John Chiefari
- CSIRO Manufacturing, Bag 10, Bayview Avenue, Clayton, VIC. 3168, Australia
| | - Michael C Jensen
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA,Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, WA 98101, USA,Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Courtney A Crane
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA,Department of Neurological Surgery, University of Washington, Seattle WA 98195, USA
| | - Patrick S Stayton
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA
| |
Collapse
|
4
|
Priester A, Waters R, Abbott A, Hilmas K, Woelk K, Miller HA, Tarudji AW, Gee CC, McDonald B, Kievit FM, Convertine AJ. Theranostic Copolymers Neutralize Reactive Oxygen Species and Lipid Peroxidation Products for the Combined Treatment of Traumatic Brain Injury. Biomacromolecules 2022; 23:1703-1712. [PMID: 35316025 PMCID: PMC9031337 DOI: 10.1021/acs.biomac.1c01635] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Traumatic brain injury (TBI) results in the generation of reactive oxygen species (ROS) and lipid peroxidation product (LPOx), including acrolein and 4-hydroxynonenal (4HNE). The presence of these biochemical derangements results in neurodegeneration during the secondary phase of the injury. The ability to rapidly neutralize multiple species could significantly improve outcomes for TBI patients. However, the difficulty in creating therapies that target multiple biochemical derangements simultaneously has greatly limited therapeutic efficacy. Therefore, our goal was to design a material that could rapidly bind and neutralize both ROS and LPOx following TBI. To do this, a series of thiol-functionalized biocompatible copolymers based on lipoic acid methacrylate and polyethylene glycol monomethyl ether methacrylate (FW ∼ 950 Da) (O950) were prepared. A polymerizable gadolinium-DOTA methacrylate monomer (Gd-MA) was also synthesized starting from cyclen to facilitate direct magnetic resonance imaging and in vivo tracking of accumulation. These neuroprotective copolymers (NPCs) were shown to rapidly and effectively neutralize both ROS and LPOx. Horseradish peroxidase absorbance assays showed that the NPCs efficiently neutralized H2O2, while R-phycoerythrin protection assays demonstrated their ability to protect the fluorescent protein from oxidative damage. 1H NMR studies indicated that the thiol-functional NPCs rapidly form covalent bonds with acrolein, efficiently removing it from solution. In vitro cell studies with SH-SY5Y-differentiated neurons showed that NPCs provide unique protection against toxic concentrations of both H2O2 and acrolein. NPCs rapidly accumulate and are retained in the injured brain in controlled cortical impact mice and reduce post-traumatic oxidative stress. Therefore, these materials show promise for improved target engagement of multiple biochemical derangements in hopes of improving TBI therapeutic outcomes.
Collapse
Affiliation(s)
- Aaron Priester
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Richard Waters
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Ashleigh Abbott
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Krista Hilmas
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Klaus Woelk
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Hunter A Miller
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68583-0900, United States
| | - Aria W Tarudji
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68583-0900, United States
| | - Connor C Gee
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68583-0900, United States
| | - Brandon McDonald
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68583-0900, United States
| | - Forrest M Kievit
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68583-0900, United States
| | - Anthony J Convertine
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| |
Collapse
|
5
|
Prossnitz AN, Pun SH. Modulating Boronic Ester Stability in Block Copolymer Micelles via the Neighbor Effect of Copolymerized Tertiary Amines for Controlled Release of Polyphenolic Drugs. ACS Macro Lett 2022; 11:276-283. [PMID: 35575376 DOI: 10.1021/acsmacrolett.1c00751] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The traceless and pH-sensitive properties of boronic esters are attractive for the synthesis of polymer-drug conjugates, but current platforms suffer from both low stability under physiologically relevant conditions and synthetically demanding optimization to tune drug release profiles. We hypothesized that the high catechol affinity and stability of Wulff-type boronic acids could be mimicked by copolymerization of phenyl boronic acid with a tertiary amine and subsequent micellization. This strategy yielded a versatile platform for the preparation of reversible polymer-drug conjugates, which more than doubled the oxidative stability of encapsulated polyphenolic drug cargo at physiologically relevant pH and enabled simple and incremental tuning of drug release kinetics. Moreover, we validated, with 19F NMR, that these copolymers exhibit uniquely high catechol affinity that could not be replicated by combinations of similarly functionalized small molecules. Overall, this report demonstrates that copolymerization of boronic acid and tertiary amine monomers is a powerful and modular approach to improving boronic ester chemistry for drug delivery applications.
Collapse
Affiliation(s)
- Alexander N. Prossnitz
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
6
|
Shin S, Kwon S, Yeo Y. Meta-Analysis of Drug Delivery Approaches for Treating Intracellular Infections. Pharm Res 2022; 39:1085-1114. [PMID: 35146592 PMCID: PMC8830998 DOI: 10.1007/s11095-022-03188-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/01/2022] [Indexed: 12/20/2022]
Abstract
This meta-analysis aims to evaluate the trend, methodological quality and completeness of studies on intracellular delivery of antimicrobial agents. PubMed, Embase, and reference lists of related reviews were searched to identify original articles that evaluated carrier-mediated intracellular delivery and pharmacodynamics (PD) of antimicrobial therapeutics against intracellular pathogens in vitro and/or in vivo. A total of 99 studies were included in the analysis. The most commonly targeted intracellular pathogens were bacteria (62.6%), followed by viruses (16.2%) and parasites (15.2%). Twenty-one out of 99 (21.2%) studies performed neither microscopic imaging nor flow cytometric analysis to verify that the carrier particles are present in the infected cells. Only 31.3% of studies provided comparative inhibitory concentrations against a free drug control. Approximately 8% of studies, albeit claimed for intracellular delivery of antimicrobial therapeutics, did not provide any experimental data such as microscopic imaging, flow cytometry, and in vitro PD. Future research on intracellular delivery of antimicrobial agents needs to improve the methodological quality and completeness of supporting data in order to facilitate clinical translation of intracellular delivery platforms for antimicrobial therapeutics.
Collapse
Affiliation(s)
- Sooyoung Shin
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, 16499, Republic of Korea. .,Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, Gyeonggi-do, 16499, Republic of Korea.
| | - Soonbum Kwon
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Dr., West Lafayette, IN, 47906, USA
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Dr., West Lafayette, IN, 47906, USA. .,Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr., West Lafayette, IN, 47907, USA.
| |
Collapse
|
7
|
Park JR, Verderosa AD, Totsika M, Hoogenboom R, Dargaville TR. Thermoresponsive Polymer-Antibiotic Conjugates Based on Gradient Copolymers of 2-Oxazoline and 2-Oxazine. Biomacromolecules 2021; 22:5185-5194. [PMID: 34726387 DOI: 10.1021/acs.biomac.1c01133] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A polymer-antibiotic conjugate with thermoresponsive properties near body temperature is presented. The backbone polymer is a copolymer of 2-n-propyl-2-oxazine (PropOzi) and methoxycarbonylethyl-2-oxazoline (C2MestOx) which is conjugated with the broad-spectrum antibiotic, cefazolin, via modification of the methyl ester group of C2MestOx. The resulting polymer-antibiotic conjugate has a cloud point temperature near body temperature, meaning that it can form a homogenous solution if cooled, but when injected into a skin-mimic at 37 °C, it forms a drug depot precipitate. Cleavage of the ester linker leads to quantitative release of the pristine cefazolin (with some antibiotic degradation observed) and redissolution of the polymer. When Escherichia coli were treated with polymer-antibiotic conjugate total clearance is observed within 12 h. The power of this approach is the potential for localized antibiotic delivery, for example, at a specific tissue site or into infected phagocytic cells.
Collapse
Affiliation(s)
- Jong-Ryul Park
- Centre for Materials Science, School of Chemistry and Physics, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - Anthony D Verderosa
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland 4006, Australia
| | - Makrina Totsika
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland 4006, Australia
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, Ghent B-9000, Belgium
| | - Tim R Dargaville
- Centre for Materials Science, School of Chemistry and Physics, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| |
Collapse
|
8
|
Zhang Z, Jones MM, Sabatini C, Vanyo ST, Yang M, Kumar A, Jiang Y, Swihart MT, Visser MB, Cheng C. Synthesis and antibacterial activity of polymer-antibiotic conjugates incorporated into a resin-based dental adhesive. Biomater Sci 2021; 9:2043-2052. [PMID: 33464241 PMCID: PMC7990707 DOI: 10.1039/d0bm01910k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This work reports on polymer-antibiotic conjugates (PACs) as additives to resin-based restorative dental materials as a new strategy to convey sustained antibacterial character to these materials. Such antibacterial performance is expected to improve their longevity in the oral cavity. Using the previously reported ciprofloxacin (Cip)-based PAC as a control, a penicillin V (PV)-based PAC was investigated. The monomer-antibiotic conjugate (MAC) containing a methacrylate monomer group and a PV moiety was prepared via nucleophilic substitution between 2-chloroethyl methacrylate (CEMA) and penicillin V potassium (PVK). The PV-based PAC was synthesized by reversible addition-fragmentation chain transfer (RAFT) polymerization of the MAC with hydroxyethyl methacrylate (HEMA), and further characterized by 1H NMR and gel permeation chromatography (GPC) analysis. Antibiotic resistance was investigated by passaging bacteria in low concentrations of the antibiotic for 19 days, followed by a 48 h challenge at higher concentrations. Our results suggest that the development of antibiotic resistance is unlikely. Zone of inhibition (ZOI) assays revealed no clearing zones around PV-containing resins indicating minimal antibiotic leakage from the material. Similarly, MTT assay demonstrated that the antibiotic-containing specimens did not release cytotoxic byproducts that may inhibit human gingival fibroblast growth. Counting of colony-forming units in an S. mutans biofilm model was used to assess bacterial survival at baseline and after subjecting the antibiotic-containing resin specimens to an enzymatic challenge for 30 days. Significantly reduced bacterial counts were observed as the biofilm aged from 24 to 72 h, and salivary enzymatic exposure did not reduce the antibacterial efficacy of the discs, suggesting that PV-resin will be effective in reducing the re-incidence of dental caries.
Collapse
Affiliation(s)
- Ziwen Zhang
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, New York 14260, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Miyazaki T, Nakagawa Y, Cabral H. Strategies for ligand-installed nanocarriers. HANDBOOK OF NANOTECHNOLOGY APPLICATIONS 2021:633-655. [DOI: 10.1016/b978-0-12-821506-7.00024-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
10
|
Ho DK, LeGuyader C, Srinivasan S, Roy D, Vlaskin V, Chavas TEJ, Lopez CL, Snyder JM, Postma A, Chiefari J, Stayton PS. Fully synthetic injectable depots with high drug content and tunable pharmacokinetics for long-acting drug delivery. J Control Release 2020; 329:257-269. [PMID: 33217474 DOI: 10.1016/j.jconrel.2020.11.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/06/2020] [Accepted: 11/15/2020] [Indexed: 12/27/2022]
Abstract
Clinical studies have validated that antiretroviral (ARV) drugs can serve as an HIV pre-exposure prophylactic (PrEP) strategy. Dosing adherence remains a crucial factor determining the final efficacy outcomes, and both long-acting implants and injectable depot systems are being developed to improve patient adherence. Here, we describe an injectable depot platform that exploits a new mechanism for both formation and controlled release. The depot is a polymeric prodrug synthesized from monomers that incorporate an ARV drug tenofovir alafenamide (TAF) with degradable linkers that can be designed to control release rates. The prodrug monomers are synthetically incorporated into homopolymer or block designs that exhibit high drug weight percent (wt%) and also are hydrophobized in these prodrug segments to drive depot formation upon injection. Drug release converts those monomers to more hydrophilic pendant groups via linker cleavage, and as this drug release proceeds, the polymer chains losing hydrophobicity are then disassociated from the depot and released over time to provide a depot dissolution mechanism. We show that long-acting TAF depots can be designed as block copolymers or as homopolymers. They can also be designed with different linkers, for example with faster or slower degrading p-hydroxybenzyloxycarbonyl (Benzyl) and ethyloxycarbonyl (Alkyl) linkers, respectively. Diblock designs of p(glycerol monomethacrylate)-b-p(Alkyl-TAF-methacrylate) and p(glycerol monomethacrylate)-b-p(Benzyl-TAF-methacrylate) were first characterized in a mouse subcutaneous injection model. The alkylcarbamate linker design (TAF 51 wt%) showed excellent sustained release profiles of the key metabolite tenofovir (TFV) in skin and plasma over a 50-day period. Next, the homopolymer design with a high TAF drug wt% of 73% was characterized in the same model. The homopolymer depots with p(Alkyl-TAFMA) exhibited sustained TFV and TAF release profiles in skin and blood over 60 days, and TFV-DP concentrations in peripheral blood mononuclear cells (PBMC) were found to be at least 10-fold higher than the clinically suggested minimally EC90 protective concentration of 24 fmol/106 cells. These are the first reports of sustained parent TAF dosing observed in mouse and TFV-DP in mouse PBMC. IVIS imaging of rhodamine labeled homopolymer depots showed that degradation and release of the depot coincided with the sustained TAF release. Finally, these polymers showed excellent stability in accelerated stability studies over a six-month time period, and exceptional solubility of over 700 mg/mL in the DMSO formulation solvent. The homopolymer designs have a drug reservoir potential of well over a year at mg/day dosing and may not require cold chain storage for global health and developed world long-acting drug delivery applications.
Collapse
Affiliation(s)
- Duy-Khiet Ho
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Clare LeGuyader
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Selvi Srinivasan
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Debashish Roy
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Vladimir Vlaskin
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Thomas E J Chavas
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Ciana L Lopez
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Jessica M Snyder
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA 98195, United States
| | - Almar Postma
- CSIRO Manufacturing, Bag 10, Clayton South MDC, Victoria 3169, Australia
| | - John Chiefari
- CSIRO Manufacturing, Bag 10, Clayton South MDC, Victoria 3169, Australia
| | - Patrick S Stayton
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
11
|
Limqueco E, Passos Da Silva D, Reichhardt C, Su FY, Das D, Chen J, Srinivasan S, Convertine A, Skerrett SJ, Parsek MR, Stayton PS, Ratner DM. Mannose Conjugated Polymer Targeting P. aeruginosa Biofilms. ACS Infect Dis 2020; 6:2866-2871. [PMID: 33074651 DOI: 10.1021/acsinfecdis.0c00407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Biofilms are one of the most challenging obstacles in bacterial infections. By providing protection against immune responses and antibiotic therapies, biofilms enable chronic colonization and the development of antibiotic resistance. As previous clinical observations and studies have shown, traditional antibiotic therapy alone cannot effectively treat and eliminate biofilm forming infections due to the protection conferred by the biofilm. A new strategy specifically targeting biofilms must be developed. Here, we specifically target and bind to the PAO1 biofilm and elucidate the molecular mechanism behind the interaction between a glycan targeted polymer and biofilm using a continuous flow biofilm model. The incubation of biofilms with fluorescent glycan targeted polymers demonstrated strong and persistent interactions with the mannose-containing polymer even after 24 h of continuous flow. To evaluate the role of major biofilm proteins LecB and CdrA, loss of function experiments with knockout variants established the dual involvement of both proteins in mannose targeted polymer retention. These results identify a persistent and specific targeting strategy to the biofilm, emphasizing its potential value as a delivery strategy and encouraging further exploration of biofilm targeted delivery.
Collapse
|
12
|
Su Y, Wang H, Mishra B, Lakshmaiah Narayana J, Jiang J, Reilly DA, Hollins RR, Carlson MA, Wang G, Xie J. Nanofiber Dressings Topically Delivering Molecularly Engineered Human Cathelicidin Peptides for the Treatment of Biofilms in Chronic Wounds. Mol Pharm 2019; 16:2011-2020. [PMID: 30916573 DOI: 10.1021/acs.molpharmaceut.8b01345] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Biofilms of multidrug-resistant bacteria in chronic wounds pose a great challenge in wound care. Herein, we report the topical delivery of molecularly engineered antimicrobial peptides using electrospun nanofiber dressings as a carrier for the treatment of biofilms of multidrug-resistant bacteria in diabetic wounds. Molecularly engineered human cathelicidin peptide 17BIPHE2 was successfully encapsulated in the core of pluronic F127/17BIPHE2-PCL core-shell nanofibers. The in vitro release profiles of 17BIPHE2 showed an in initial burst followed by a sustained release over 4 weeks. The peptide nanofiber formulations effectively killed methicillin-resistant Staphylococcus aureus (MRSA) USA300. Similarly, the 17BIPHE2 peptide containing nanofibers could also effectively kill other bacteria including Klebsiella pneumoniae (104 to 106 CFU) and Acinetobacter baumannii (104 to 107 CFU) clinical strains in vitro without showing evident cytotoxicity to skin cells and monocytes. Importantly, 17BIPHE2-containing nanofiber dressings without debridement caused five-magnitude decreases of the MRSA USA300 CFU in a biofilm-containing chronic wound model based on type II diabetic mice. In combination with debridement, 17BIPHE2-containing nanofiber dressings could completely eliminate the biofilms, providing one possible solution to chronic wound treatment. Taken together, the biodegradable nanofiber-based wound dressings developed in this study can be utilized to effectively deliver molecularly engineered peptides to treat biofilm-containing chronic wounds.
Collapse
|
13
|
Mulas K, Stefanowicz Z, Oledzka E, Sobczak M. Current state of the polymeric delivery systems of fluoroquinolones – A review. J Control Release 2019; 294:195-215. [DOI: 10.1016/j.jconrel.2018.12.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 01/29/2023]
|
14
|
Zhang R, Jones MM, Moussa H, Keskar M, Huo N, Zhang Z, Visser MB, Sabatini C, Swihart MT, Cheng C. Polymer–antibiotic conjugates as antibacterial additives in dental resins. Biomater Sci 2019; 7:287-295. [DOI: 10.1039/c8bm01228h] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Dental resins containing polymer–antibiotic conjugates (PACs) demonstrate significant antibacterial properties.
Collapse
|
15
|
Freeman H, Srinivasan S, Das D, Stayton PS, Convertine AJ. Fully synthetic macromolecular prodrug chemotherapeutics with EGFR targeting and controlled camptothecin release kinetics. Polym Chem 2018; 9:5224-5233. [PMID: 36660314 PMCID: PMC9847574 DOI: 10.1039/c8py01047a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Herein, we developed a fully polymerizable, peptide-targeted, camptothecin polymeric prodrug system. Two prodrug monomers were synthesized via esterification of campothecin (20Cam) and 10-hydroxycamptothecin (10Cam) with mono-2-(methacryloyloxy)ethyl succinate (SMA) resulting in polymerizable forms of the aliphatic ester- and aromatic ester-linked drugs respectively. These monomers were then incorporated into zwitterionic polymers via RAFT copolymerization of the prodrug monomers with a tert-butyl ester protected carboxy betaine monomer. Subsequent deprotection of the tert-butyl residues with TFA yielded carboxy betaine methacrylate (CBM) scaffolds with controlled prodrug incorporation. Reverse phase HPLC was then employed to establish drug release kinetics in human serum at 37 oC for the resultant polymeric prodrugs. Copolymers containing 10Cam residues linked via aromatic esters showed faster hydrolysis rates with 59 % drug released at 7 days, while copolymers with Cam residues linked via aliphatic esters showed only 28 % drug release over the same time period. These differences in drug release kinetics were then shown to correlate with large differences in cytotoxic activity in SKOV3 ovarian cancer cell cultures. At 72 hours, the IC50s of aromatic- and aliphatic- ester linked prodrugs were 56 nM and 4776 nM, respectively. An EGFR-targeting peptide sequence, GE11, was then directly incorporated into the polymeric prodrugs via RAFT copolymerization of the polymeric prodrugs with a peptide macronomer. The GE11-targeted polymeric prodrugs showed enhanced targeting and cytotoxic activity in SKOV3 cell cultures relative to untargeted polymers containing the negative control sequence HW12. Following pulse-chase treatment (15 min, 37 °C), the 72 hour IC50 of GE11 targeted prodrug was determined to be 1597 nM, in contrast to 3399 nM for the non-targeted control.
Collapse
Affiliation(s)
- Hanna Freeman
- Molecular Engineering and Sciences Institute, department of BioEngineering, Box 355061, Seattle WA, 98195, USA
| | - Selvi Srinivasan
- Molecular Engineering and Sciences Institute, department of BioEngineering, Box 355061, Seattle WA, 98195, USA
| | - Debobrato Das
- Molecular Engineering and Sciences Institute, department of BioEngineering, Box 355061, Seattle WA, 98195, USA
| | - Patrick S Stayton
- Molecular Engineering and Sciences Institute, department of BioEngineering, Box 355061, Seattle WA, 98195, USA
| | - Anthony J Convertine
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla MO, 65401, USA
| |
Collapse
|
16
|
Chen J, Su FY, Das D, Srinivasan S, Son HN, Lee B, Radella F, Whittington D, Monroe-Jones T, West TE, Convertine AJ, Skerrett SJ, Stayton PS, Ratner DM. Glycan targeted polymeric antibiotic prodrugs for alveolar macrophage infections. Biomaterials 2018; 195:38-50. [PMID: 30610992 DOI: 10.1016/j.biomaterials.2018.10.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/03/2018] [Accepted: 10/15/2018] [Indexed: 10/28/2022]
Abstract
Alveolar macrophages resident in the lung are prominent phagocytic effector cells of the pulmonary innate immune response, and paradoxically, are attractive harbors for pathogens. Consequently, facultative intracellular bacteria, such as Francisella tularensis, can cause severe systemic disease and sepsis, with high morbidity and mortality associated with pulmonary infection. Current clinical treatment, which involves exhaustive oral or intravenous antibiotic therapy, has limitations such as systemic toxicity and off-target effects. Pulmonary administration represents a promising alternative to systemic dosing for delivering antibiotics directly to the lung. Here, we present synthesized mannosylated ciprofloxacin polymeric prodrugs for efficient pulmonary delivery, targeting, and subsequent internalization by alveolar macrophages. We demonstrate significant improvement in efficacy against intracellular infections in an otherwise uniformly lethal airborne Francisella murine model (F. novicida). When administered to the lungs of mice in a prophylactic regimen, the mannosylated ciprofloxacin polymeric prodrugs led to 50% survival. In a treatment regimen that was concurrent with infection, the survival of mice increased to 87.5%. Free ciprofloxacin antibiotic was ineffective in both cases. This significant difference in antibacterial efficacy demonstrates the impact of this delivery platform based on improved physiochemical, pharmacokinetic, and pharmacodynamic properties of ciprofloxacin administered via our glycan polymeric prodrug. This modular platform provides a route for overcoming the limitations of free drug and increasing efficacy in treatment of intracellular infection.
Collapse
Affiliation(s)
- Jasmin Chen
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Fang-Yi Su
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Debobrato Das
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Selvi Srinivasan
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Hye-Nam Son
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Brian Lee
- Division of Pulmonary, Critical Care & Sleep Medicine, Harborview Medical Center, University of Washington, Seattle, WA 98104, USA
| | - Frank Radella
- Division of Pulmonary, Critical Care & Sleep Medicine, Harborview Medical Center, University of Washington, Seattle, WA 98104, USA
| | - Dale Whittington
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Taylor Monroe-Jones
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - T Eoin West
- Division of Pulmonary, Critical Care & Sleep Medicine, Harborview Medical Center, University of Washington, Seattle, WA 98104, USA
| | | | - Shawn J Skerrett
- Division of Pulmonary, Critical Care & Sleep Medicine, Harborview Medical Center, University of Washington, Seattle, WA 98104, USA
| | - Patrick S Stayton
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
| | - Daniel M Ratner
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
17
|
Su FY, Srinivasan S, Lee B, Chen J, Convertine AJ, West TE, Ratner DM, Skerrett SJ, Stayton PS. Macrophage-targeted drugamers with enzyme-cleavable linkers deliver high intracellular drug dosing and sustained drug pharmacokinetics against alveolar pulmonary infections. J Control Release 2018; 287:1-11. [PMID: 30099019 PMCID: PMC6223132 DOI: 10.1016/j.jconrel.2018.08.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/30/2018] [Accepted: 08/07/2018] [Indexed: 12/18/2022]
Abstract
Intracellular bacterial infections localized to the lung alveolar macrophage (AM) remain one of the most challenging settings for antimicrobial therapy. Current systemic antibiotic treatment fails to deliver sustained doses to intracellular bacterial reservoirs, which necessitates prolonged treatment regimens. Herein, we demonstrate a new intracellular enzyme-cleavable polymeric prodrug with tailored ciprofloxacin release profiles in the lungs and AM. The targeted polymeric prodrug, termed "drugamers", incorporates (1) hydrophilic mannose residues to solubilize the antibiotic cargo and to target and enhance AM uptake and intracellular delivery, and (2) enzyme-cleavable linkage chemistry to provide high and sustained intracellular AM drug dosing. Prodrug monomers, derived from the antibiotic ciprofloxacin, were synthesized with either an intracellular protease cleavable dipeptide linker or a hydrolytic phenyl ester linker. RAFT polymerization was used to copolymerize the prodrug monomers and mannose monomer to synthesize well-defined drugamers without requiring a post-polymerization conjugation step. In addition to favorable in vivo safety profiles following intratracheal administration, a single dose of the drugamers sustained ciprofloxacin dosing in lungs and AMs above the minimum inhibitory concentration (MIC) over at least a 48 h period. The enzyme-cleavable therapeutic achieved a >10-fold increase in sustained ciprofloxacin in AM, and maintained a significantly higher whole lung PK as well. Ciprofloxacin dosed in identical fashion displayed rapid clearance with a half-life of approximately 30 min. Notably, inhalation of the mannose-targeted ciprofloxacin drugamers achieved full survival (100%) in a highly lethal mouse model of pneumonic tularemia, contrasted with 0% survival using free ciprofloxacin. These findings demonstrate the versatility of the drugamer platform for engineering the intracellular pharmacokinetic profiles and its strong therapeutic activity in treating pulmonary intracellular infections.
Collapse
Affiliation(s)
- Fang-Yi Su
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Selvi Srinivasan
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Brian Lee
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, WA 98104, United States
| | - Jasmin Chen
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Anthony J Convertine
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Timothy Eoin West
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, WA 98104, United States; Department of Global Health, University of Washington, Seattle, WA 98195, United States.
| | - Daniel M Ratner
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States.
| | - Shawn J Skerrett
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, WA 98104, United States.
| | - Patrick S Stayton
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
18
|
Goes A, Fuhrmann G. Biogenic and Biomimetic Carriers as Versatile Transporters To Treat Infections. ACS Infect Dis 2018; 4:881-892. [PMID: 29553240 DOI: 10.1021/acsinfecdis.8b00030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biogenic and biomimetic therapeutics are a relatively new class of systems that are of physiological origin and/or take advantage of natural pathways or aim at mimicking these to improve selective interaction with target tissue. The number of biogenic and bioengineered avenues for drug therapy and diagnostics has multiplied over the past years for many applications, indicating the high expectations associated with this biological route. Nevertheless, the use of "bio"-related approaches for treating or diagnosing infectious diseases is still rare. Given that infectious diseases, in particular bacterial resistances, are seriously on the rise, there is an urgent need to take advantage of biogenic and bioengineered systems to target these challenges. In this manuscript, we first give a definition of the various "bio" terms, including biogenic, biomimetic, bioinspired, and bioengineered and we highlight them using tangible applications in the field of infectious diseases. Our examples cover cell-derived systems, including bioengineered bacteria, virus-like particles, and different cell-mimetics. Moreover, we discuss natural and bioengineered particles such as extracellular vesicles from mammalian and bacterial sources and liposomes. A concluding section outlines the potential for biomaterial-related avenues to overcome challenges associated with difficult-to-treat infections. We critically discuss benefits and risks for these applications and give an outlook on the future of biogenic engineering.
Collapse
Affiliation(s)
- Adriely Goes
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Biogenic Nanotherapeutics group (BION), Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
| | - Gregor Fuhrmann
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Biogenic Nanotherapeutics group (BION), Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
19
|
Das D, Srinivasan S, Brown FD, Su FY, Burrell AL, Kollman JM, Postma A, Ratner DM, Stayton PS, Convertine AJ. Radiant star nanoparticle prodrugs for the treatment of intracellular alveolar infections. Polym Chem 2018. [DOI: 10.1039/c8py00202a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Radiant star nanoparticle prodrugs were synthesized in a two-step process by first homopolymerizing RAFT transmers followed by copolymerization from the hyperbranched polymer core.
Collapse
Affiliation(s)
- D. Das
- Molecular Engineering and Sciences Institute
- department of BioEngineering
- Seattle
- USA
| | - S. Srinivasan
- Molecular Engineering and Sciences Institute
- department of BioEngineering
- Seattle
- USA
| | - F. D. Brown
- Molecular Engineering and Sciences Institute
- department of BioEngineering
- Seattle
- USA
| | - F. Y. Su
- Molecular Engineering and Sciences Institute
- department of BioEngineering
- Seattle
- USA
| | - A. L. Burrell
- University of Washington
- Department of Biochemistry
- USA
| | - J. M. Kollman
- University of Washington
- Department of Biochemistry
- USA
| | - A. Postma
- The Commonwealth Scientific and Industrial Research Organization (CSIRO) Manufacturing
- Clayton
- Australia
| | - D. M. Ratner
- Molecular Engineering and Sciences Institute
- department of BioEngineering
- Seattle
- USA
| | - P. S. Stayton
- Molecular Engineering and Sciences Institute
- department of BioEngineering
- Seattle
- USA
| | - A. J. Convertine
- Molecular Engineering and Sciences Institute
- department of BioEngineering
- Seattle
- USA
| |
Collapse
|