1
|
Shahzad KA, Wang Z, Li X, Li J, Xu M, Tan F. Immunomodulatory effect of PLGA-encapsulated mesenchymal stem cells-derived exosomes for the treatment of allergic rhinitis. Front Immunol 2024; 15:1429442. [PMID: 39040099 PMCID: PMC11260627 DOI: 10.3389/fimmu.2024.1429442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Allergic rhinitis (AR) is an upper airway inflammatory disease of the nasal mucosa. Conventional treatments such as symptomatic pharmacotherapy and allergen-specific immunotherapy have considerable limitations and drawbacks. As an emerging therapy with regenerative potential and immunomodulatory effect, mesenchymal stem cell-derived exosomes (MSC-Exos) have recently been trialed for the treatment of various inflammatory and autoimmune diseases. Methods In order to achieve sustained and protected release of MSC-Exos for intranasal administration, we fabricated Poly(lactic-co-glycolic acid) (PLGA) micro and nanoparticles-encapsulated MSC-Exos (PLGA-Exos) using mechanical double emulsion for local treatment of AR. Preclinical in vivo imaging, ELISA, qPCR, flow cytometry, immunohistochemical staining, and multiomics sequencing were used for phenotypic and mechanistic evaluation of the therapeutic effect of PLGA-Exos in vitro and in vivo. Results The results showed that our PLGA platform could efficiently encapsulate and release the exosomes in a sustained manner. At protein level, PLGA-Exos treatment upregulated IL-2, IL-10 and IFN-γ, and downregulated IL-4, IL-17 and antigen-specific IgE in ovalbumin (OVA)-induced AR mice. At cellular level, exosomes treatment reduced Th2 cells, increased Tregs, and reestablished Th1/Th2 balance. At tissue level, PLGA-Exos significantly attenuated the infiltration of immune cells (e.g., eosinophils and goblet cells) in nasal mucosa. Finally, multiomics analysis discovered several signaling cascades, e.g., peroxisome proliferator-activated receptor (PPAR) pathway and glycolysis pathway, that might mechanistically support the immunomodulatory effect of PLGA-Exos. Discussion For the first time, we present a biomaterial-facilitated local delivery system for stem cell-derived exosomes as a novel and promising strategy for AR treatment.
Collapse
Affiliation(s)
- Khawar Ali Shahzad
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai, China
| | - Zhao Wang
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuran Li
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai, China
| | - Jiaojiao Li
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai, China
| | - Maoxiang Xu
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai, China
| | - Fei Tan
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai, China
- The Royal College of Surgeons in Ireland, Dublin, Ireland
- The Royal College of Surgeons of England, London, United Kingdom
| |
Collapse
|
2
|
Chen X, Moonshi SS, Nguyen NT, Ta HT. Preparation of protein-loaded nanoparticles based on poly(succinimide)-oleylamine for sustained protein release: a two-step nanoprecipitation method. NANOTECHNOLOGY 2023; 35:055101. [PMID: 37863070 DOI: 10.1088/1361-6528/ad0592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/20/2023] [Indexed: 10/22/2023]
Abstract
Currently, the treatment for acute disease encompasses the use of various biological drugs (BDs). However, the utilisation of BDs is limited due to their rapid clearance and non-specific accumulation in unwanted sites, resulting in a lack of therapeutic efficacy together with adverse effects. While nanoparticles are considered good candidates to resolve this problem, some available polymeric carriers for BDs were mainly designed for long-term sustained release. Thus, there is a need to explore new polymeric carriers for the acute disease phase that requires sustained release of BDs over a short period, for example for thrombolysis and infection. Poly(succinimide)-oleylamine (PSI-OA), a biocompatible polymer with a tuneable dissolution profile, represents a promising strategy for loading BDs for sustained release within a 48-h period. In this work, we developed a two-step nanoprecipitation method to load the model protein (e.g. bovine serum albumin and lipase) on PSI-OA. The characteristics of the nanoparticles were assessed based on various loading parameters, such as concentration, stirring rate, flow rate, volume ratio, dissolution and release of the protein. The optimised NPs displayed a size within 200 nm that is suitable for vasculature delivery to the target sites. These findings suggest that PSI-OA can be employed as a carrier for BDs for applications that require sustained release over a short period.
Collapse
Affiliation(s)
- Xiangxun Chen
- School of Environment and Science, Griffith University, Brisbane, Queensland 4111, Australia
- Queensland Micro- and Nanotechnology Centre, Griffith University, Brisbane, Queensland 4111, Australia
| | - Shehzahdi S Moonshi
- School of Environment and Science, Griffith University, Brisbane, Queensland 4111, Australia
- Queensland Micro- and Nanotechnology Centre, Griffith University, Brisbane, Queensland 4111, Australia
| | - Nam-Trung Nguyen
- School of Environment and Science, Griffith University, Brisbane, Queensland 4111, Australia
- Queensland Micro- and Nanotechnology Centre, Griffith University, Brisbane, Queensland 4111, Australia
| | - Hang Thu Ta
- School of Environment and Science, Griffith University, Brisbane, Queensland 4111, Australia
- Queensland Micro- and Nanotechnology Centre, Griffith University, Brisbane, Queensland 4111, Australia
| |
Collapse
|
3
|
Corpstein CD, Li T. A Perspective on Model-Informed IVIVC for Development of Subcutaneous Injectables. Pharm Res 2023; 40:1633-1639. [PMID: 37523013 DOI: 10.1007/s11095-023-03572-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023]
Abstract
Subcutaneously administered drugs are growing in popularity for both large and small molecule drugs. However, development of these systems - particularly generics - is slowed due to a lack of formal guidance regarding preclinical testing and in vitro - in vivo correlations (IVIVC). Many of these methods, while appropriate for oral drugs, may not be optimized for the complex injection site physiologies, and release rate and absorption mechanisms of subcutaneous drugs. Current limitations for formulation design and IVIVC can be supported by implementing mechanistic, computational methods. These methods can help to inform drug development by identifying key drug and formulation attributes, and their effects on drug release rates. This perspective, therefore, addresses current guidelines in place for oral IVIVC development, how they may differ for subcutaneously administered compounds, and how modeling and simulation can be implemented to inform design of these products. As such, integration of modeling and simulation with current IVIVC systems can help in driving the development of subcutaneous injectables.
Collapse
Affiliation(s)
- Clairissa D Corpstein
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana, USA
| | - Tonglei Li
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
4
|
Ahmad S, Mirza MU, Trant JF. Dock-able linear and homodetic di, tri, tetra and pentapeptide library from canonical amino acids: SARS-CoV-2 Mpro as a case study. J Pharm Anal 2023; 13:523-534. [PMID: 37275125 PMCID: PMC10104786 DOI: 10.1016/j.jpha.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/07/2023] [Accepted: 04/13/2023] [Indexed: 06/07/2023] Open
Abstract
Peptide-based therapeutics are increasingly pushing to the forefront of biomedicine with their promise of high specificity and low toxicity. Although noncanonical residues can always be used, employing only the natural 20 residues restricts the chemical space to a finite dimension allowing for comprehensive in silico screening. Towards this goal, the dataset comprising all possible di-, tri-, and tetra-peptide combinations of the canonical residues has been previously reported. However, with increasing computational power, the comprehensive set of pentapeptides is now also feasible for screening as the comprehensive set of cyclic peptides comprising four or five residues. Here, we provide both the complete and prefiltered libraries of all di-, tri-, tetra-, and penta-peptide sequences from 20 canonical amino acids and their homodetic (N-to-C-terminal) cyclic homologues. The FASTA, simplified molecular-input line-entry system (SMILES), and structure-data file (SDF)-three dimension (3D) libraries can be readily used for screening against protein targets. We also provide a simple method and tool for conducting identity-based filtering. Access to this dataset will accelerate small peptide screening workflows and encourage their use in drug discovery campaigns. As a case study, the developed library was screened against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease to identify potential small peptide inhibitors.
Collapse
Affiliation(s)
- Sarfraz Ahmad
- Department of Chemistry and Biochemistry, University of Windsor, Windsor N9B 3P4, Ontario, Canada
- Binary Star Research Services, LaSalle N9J 3X8, Ontario, Canada
| | - Muhammad Usman Mirza
- Department of Chemistry and Biochemistry, University of Windsor, Windsor N9B 3P4, Ontario, Canada
- Binary Star Research Services, LaSalle N9J 3X8, Ontario, Canada
| | - John F Trant
- Department of Chemistry and Biochemistry, University of Windsor, Windsor N9B 3P4, Ontario, Canada
- Binary Star Research Services, LaSalle N9J 3X8, Ontario, Canada
| |
Collapse
|
5
|
Cohen J, Shull D, Reed S. Co-delivery of an HIV prophylactic and contraceptive using PGSU as a long-acting multipurpose prevention technology. Expert Opin Drug Deliv 2023; 20:285-299. [PMID: 36654482 DOI: 10.1080/17425247.2023.2168642] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVES Poly(glycerol sebacate) urethane (PGSU) elastomers formulated with 4'-ethynyl-2-fluoro-2'-deoxyadenosine (EFdA), levonorgestrel (LNG), or a combination thereof can function as multipurpose prevention technology implants for prophylaxis against HIV and unintended pregnancies. For these public health challenges, long-acting drug delivery technologies may improve patient experience and adherence. Traditional polymers encounter challenges delivering multiple drugs with dissimilar physiochemical properties. PGSU offers an alternative option that successfully delivers hydrophilic EFdA alongside hydrophobic LNG. METHODS This article presents the formulation, design, and characterization of PGSU implants, highlighting the impact of API loading, dimensions, and individual- versus combination-loading on release rates. RESULTS Co-delivery of hydrophilic EFdA alongside hydrophobic LNG acted as a porogen to accelerate LNG release. Increasing the surface area of LNG-only implants increased LNG release. All EFdA-LNG, EFdA-only, and LNG-only formulated implants demonstrated low burst release and linear release kinetics over 245 or 122 days studied to date. CONCLUSION PGSU co-delivers two APIs for HIV prevention and contraception at therapeutically relevant concentrations in vitro from a single bioresorbable, elastomeric implant. A new long-acting polymer technology, PGSU demonstrates linear-release kinetics, dual delivery of APIs with disparate physiochemical properties, and biocompatibility through long-term subcutaneous implantation. PGSU can potentially meet the demands of complex MPT or fixed-dose combination products, where better solutions can serve and empower patients.
Collapse
|
6
|
Guo X, Lou J, Wang F, Fan D, Qin Z. Recent Advances in Nano-Therapeutic Strategies for Osteoarthritis. Front Pharmacol 2022; 13:924387. [PMID: 35800449 PMCID: PMC9253376 DOI: 10.3389/fphar.2022.924387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/30/2022] [Indexed: 01/08/2023] Open
Abstract
Osteoarthritis (OA) is the most common type of arthritis and the leading cause of disability globally. It tends to occur in middle age or due to an injury or obesity. OA occurs with the onset of symptoms, including joint swelling, joint effusion, and limited movement at a late stage of the disease, which leads to teratogenesis and loss of joint function. During the pathogenesis of this degenerative joint lesion, several local inflammatory responses are activated, resulting in synovial proliferation and pannus formation that facilitates the destruction of the bone and the articular cartilage. The commonly used drugs for the clinical diagnosis and treatment of OA have limitations such as low bioavailability, short half-life, poor targeting, and high systemic toxicity. With the application of nanomaterials and intelligent nanomedicines, novel nanotherapeutic strategies have shown more specific targeting, prolonged half-life, refined bioavailability, and reduced systemic toxicity, compared to the existing medications. In this review, we summarized the recent advancements in new nanotherapeutic strategies for OA and provided suggestions for improving the treatment of OA.
Collapse
Affiliation(s)
- Xinjing Guo
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jia Lou
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Fazhan Wang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- *Correspondence: Daoyang Fan, ; Fazhan Wang, ; Zhihai Qin,
| | - Daoyang Fan
- Department of Orthopedic, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- *Correspondence: Daoyang Fan, ; Fazhan Wang, ; Zhihai Qin,
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Daoyang Fan, ; Fazhan Wang, ; Zhihai Qin,
| |
Collapse
|
7
|
Three-Dimensional Printing of a Container Tablet: A New Paradigm for Multi-Drug-Containing Bioactive Self-Nanoemulsifying Drug-Delivery Systems (Bio-SNEDDSs). Pharmaceutics 2022; 14:pharmaceutics14051082. [PMID: 35631668 PMCID: PMC9147480 DOI: 10.3390/pharmaceutics14051082] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
This research demonstrates the use of fused deposition modeling (FDM) 3D printing to control the delivery of multiple drugs containing bioactive self-nano emulsifying drug-delivery systems (SNEDDSs). Around two-thirds of the new chemical entities being introduced in the market are associated with some inherent issues, such as poor solubility and high lipophilicity. SNEDDSs provide for an innovative and easy way to develop a delivery platform for such drugs. Combining this platform with FDM 3D printing would further aid in developing new strategies for delivering poorly soluble drugs and personalized drug-delivery systems with added therapeutic benefits. This study evaluates the performance of a 3D-printed container system containing curcumin (CUR)- and lansoprazole (LNS)-loaded SNEDDS. The SNEDDS showed 50% antioxidant activity (IC50) at concentrations of around 330.1 µg/mL and 393.3 µg/mL in the DPPH and ABTS radical scavenging assay, respectively. These SNEDDSs were loaded with no degradation and leakage from the 3D-printed container. We were able to delay the release of the SNEDDS from the hollow prints while controlling the print wall thickness to achieve lag phases of 30 min and 60 min before the release from the 0.4 mm and 1 mm wall thicknesses, respectively. Combining these two innovative drug-delivery strategies demonstrates a novel option for tackling the problems associated with multi-drug delivery and delivery of drugs susceptible to degradation in, i.e., gastric pH for targeting disease conditions throughout the gastrointestinal tract (GIT). It is also envisaged that such delivery systems reported herein can be an ideal solution to deliver many challenging molecules, such as biologics, orally or near the target site in the future, thus opening a new paradigm for multi-drug-delivery systems.
Collapse
|
8
|
Kunde SS, Ghosh R, Wairkar S. Emerging trends in pulmonary delivery of biopharmaceuticals. Drug Discov Today 2022; 27:1474-1482. [PMID: 35143963 DOI: 10.1016/j.drudis.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/21/2021] [Accepted: 02/03/2022] [Indexed: 11/03/2022]
Abstract
Over the years, a tendency toward biopharmaceutical products as therapeutics has been witnessed compared with small molecular drugs. Biopharmaceuticals possess greater specificity, selectivity and potency with fewer side effects. The pulmonary route is a potential noninvasive route studied for the delivery of various molecules, including biopharmaceuticals. It directly delivers drugs to the lungs in higher concentrations and provides greater bioavailability than other noninvasive routes. This review focuses on the pulmonary route for the delivery of biopharmaceuticals. We have covered various biopharmaceuticals, including peptides, recombinant proteins, enzymes, monoclonal antibodies and nucleic acids, administered via a pulmonary route and discussed their rewards and drawbacks.
Collapse
Affiliation(s)
- Shalvi Sinai Kunde
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India
| | - Ritushree Ghosh
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India.
| |
Collapse
|
9
|
An Overview of Nanotechnologies for Drug Delivery to the Brain. Pharmaceutics 2022; 14:pharmaceutics14020224. [PMID: 35213957 PMCID: PMC8875260 DOI: 10.3390/pharmaceutics14020224] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/12/2022] Open
Abstract
Drug delivery to the brain has been one of the toughest challenges researchers have faced to develop effective treatments for brain diseases. Owing to the blood–brain barrier (BBB), only a small portion of administered drug can reach the brain. A consequence of that is the need to administer a higher dose of the drug, which, expectedly, leads to a variety of unwanted side effects. Research in a variety of different fields has been underway for the past couple of decades to address this very serious and frequently lethal problem. One area of research that has produced optimistic results in recent years is nanomedicine. Nanomedicine is the science birthed by fusing the fields of nanotechnology, chemistry and medicine into one. Many different types of nanomedicine-based drug-delivery systems are currently being studied for the sole purpose of improved drug delivery to the brain. This review puts together and briefly summarizes some of the major breakthroughs in this crusade. Inorganic nanoparticle-based drug-delivery systems, such as gold nanoparticles and magnetic nanoparticles, are discussed, as well as some organic nanoparticulate systems. Amongst the organic drug-delivery nanosystems, polymeric micelles and dendrimers are discussed briefly and solid polymeric nanoparticles are explored in detail.
Collapse
|
10
|
Miao YB, Lin YJ, Chen KH, Luo PK, Chuang SH, Yu YT, Tai HM, Chen CT, Lin KJ, Sung HW. Engineering Nano- and Microparticles as Oral Delivery Vehicles to Promote Intestinal Lymphatic Drug Transport. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2104139. [PMID: 34596293 DOI: 10.1002/adma.202104139] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/20/2021] [Indexed: 06/13/2023]
Abstract
Targeted oral delivery of a drug via the intestinal lymphatic system (ILS) has the advantages of protecting against hepatic first-pass metabolism of the drug and improving its pharmacokinetic performance. It is also a promising route for the oral delivery of vaccines and therapeutic agents to induce mucosal immune responses and treat lymphatic diseases, respectively. This article describes the anatomical structures and physiological characteristics of the ILS, with an emphasis on enterocytes and microfold (M) cells, which are the main gateways for the transport of particulate delivery vehicles across the intestinal epithelium into the lymphatics. A comprehensive overview of recent advances in the rational engineering of particulate vehicles, along with the challenges and opportunities that they present for improving ILS drug delivery, is provided, and the mechanisms by which such vehicles target and transport through enterocytes or M cells are discussed. The use of naturally sourced materials, such as yeast microcapsules and their derived polymeric β-glucans, as novel ILS-targeting delivery vehicles is also reviewed. Such use is the focus of an emerging field of research. Their potential use in the oral delivery of nucleic acids, such as mRNA vaccines, is proposed.
Collapse
Affiliation(s)
- Yang-Bao Miao
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Yu-Jung Lin
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Kuan-Hung Chen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Po-Kai Luo
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Shun-Hao Chuang
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Yu-Tzu Yu
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Hsien-Meng Tai
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan, Republic of China
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Molecular Imaging Center, Linkou Chang Gung Memorial Hospital, and Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| |
Collapse
|
11
|
Sarker S, Wankum B, Perey T, Mau MM, Shimizu J, Jones R, Terry B. A Novel Capsule-Delivered Enteric Drug-Injection Device for Delivery of Systemic Biologics: A Pilot Study in a Porcine Model. IEEE Trans Biomed Eng 2021; 69:1870-1879. [PMID: 34807818 DOI: 10.1109/tbme.2021.3129653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Innovative swallowable capsule technologies such as drug-loaded, dissolvable microneedles, mucoadhesive patches, and various microdevices present unique drug-carrying capabilities to overcome challenges regarding oral delivery of biologics. Here, we report a swallowable capsule for intestinal drug delivery (SCIDD) with the potential of directly injecting biological therapeutics into the insensate small intestine wall. The design, optimization, and validation of the SCIDD's primary subsystems were performed both ex-vivo and in-vivo. The assembled capsule was further tested in vivo to validate the actuation sequence and showed a 70% (n=17) success rate in an animal model. Additionally, a drug delivery study indicated systemic uptake of adalimumab via SCIDD compared with luminal delivery in the small intestine. The pilot study presented here establishes that the novel platform could be used to orally deliver systemic biologics.
Collapse
|
12
|
McNulty MJ, Berliner AJ, Negulescu PG, McKee L, Hart O, Yates K, Arkin AP, Nandi S, McDonald KA. Evaluating the Cost of Pharmaceutical Purification for a Long-Duration Space Exploration Medical Foundry. Front Microbiol 2021; 12:700863. [PMID: 34707576 PMCID: PMC8542935 DOI: 10.3389/fmicb.2021.700863] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/20/2021] [Indexed: 01/10/2023] Open
Abstract
There are medical treatment vulnerabilities in longer-duration space missions present in the current International Space Station crew health care system with risks, arising from spaceflight-accelerated pharmaceutical degradation and resupply lag times. Bioregenerative life support systems may be a way to close this risk gap by leveraging in situ resource utilization (ISRU) to perform pharmaceutical synthesis and purification. Recent literature has begun to consider biological ISRU using microbes and plants as the basis for pharmaceutical life support technologies. However, there has not yet been a rigorous analysis of the processing and quality systems required to implement biologically produced pharmaceuticals for human medical treatment. In this work, we use the equivalent system mass (ESM) metric to evaluate pharmaceutical purification processing strategies for longer-duration space exploration missions. Monoclonal antibodies, representing a diverse therapeutic platform capable of treating multiple space-relevant disease states, were selected as the target products for this analysis. We investigate the ESM resource costs (mass, volume, power, cooling, and crew time) of an affinity-based capture step for monoclonal antibody purification as a test case within a manned Mars mission architecture. We compare six technologies (three biotic capture methods and three abiotic capture methods), optimize scheduling to minimize ESM for each technology, and perform scenario analysis to consider a range of input stream compositions and pharmaceutical demand. We also compare the base case ESM to scenarios of alternative mission configuration, equipment models, and technology reusability. Throughout the analyses, we identify key areas for development of pharmaceutical life support technology and improvement of the ESM framework for assessment of bioregenerative life support technologies.
Collapse
Affiliation(s)
- Matthew J. McNulty
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, United States
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
| | - Aaron J. Berliner
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, United States
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, United States
| | - Patrick G. Negulescu
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, United States
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
| | - Liber McKee
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
| | - Olivia Hart
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
| | - Kevin Yates
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, United States
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
| | - Adam P. Arkin
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, United States
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, United States
| | - Somen Nandi
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, United States
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
- Global HealthShare Initiative, University of California, Davis, Davis, CA, United States
| | - Karen A. McDonald
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, United States
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
- Global HealthShare Initiative, University of California, Davis, Davis, CA, United States
| |
Collapse
|
13
|
Ultrasound-Guided Knee Injections Are More Accurate Than Blind Injections: A Systematic Review of Randomized Controlled Trials. Arthrosc Sports Med Rehabil 2021; 3:e1177-e1187. [PMID: 34430899 PMCID: PMC8365196 DOI: 10.1016/j.asmr.2021.01.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 01/30/2021] [Indexed: 11/21/2022] Open
Abstract
Purpose To review the current literature to determine which injection technique and needle portal placement provide the greatest accuracy for intra-articular access to the knee. Methods This study followed Preferred Reporting Items and Systematic Reviews and Meta-Analyses guidelines. A comprehensive literature search was conducted in March 2020 and repeated in May 2020 using electronic databases PubMed, MEDLINE, and the Cochrane Library. Data on the accuracy of intra-articular knee injection (successful injections/total number of injections) were collected. Only Level I studies were included. Study design, demographic variables, needle sizes, and method of validating accuracy were recorded. The Jadad score was used to assess methodologic quality, and a risk-of-bias assessment was performed. Results A total of 12 Level I human studies (1431 patients, 1315 knees) were included in this review. Seven of the studies did a direct comparison between ultrasound-guided and blind knee injections. Ultrasound-guided injections were more accurate compared with blinded knee injections in every study. The most accurate anatomical approach was an isometric quadricep contraction method with the superolateral approach. Conclusions This study showed that ultrasound-guided knee injections were more accurate across every anatomical needle injection site compared with blind injections. Injections made by a blind/anatomically guided method had inconsistent accuracy rates that seemed highly dependent on the portal of entry. Level of Evidence Level I, systematic review of Level I studies.
Collapse
|
14
|
Carneiro DC, Fernandez LG, Monteiro-Cunha JP, Benevides RG, Cunha Lima ST. A patent review of the antimicrobial applications of lectins: Perspectives on therapy of infectious diseases. J Appl Microbiol 2021; 132:841-854. [PMID: 34416098 DOI: 10.1111/jam.15263] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022]
Abstract
Patents of lectins with antiviral, antibacterial and antifungal applications were searched and reviewed. Lectins are proteins that reversibly bind to specific carbohydrates and have the potential for therapy of infectious diseases as biopharmaceuticals, biomedical tools or in drug design. Given the rising concerns over drug resistance and epidemics, our patent review aims to add information, open horizons and indicate our view of the future perspectives about the antimicrobial applications of lectins. Patents with publications until December 2020 were retrieved from Espacenet using defined search terms and Boolean operators. The documents were used to identify the geographical and temporal distribution of the patents, characterize their lectins, and classify and summarize their antiviral, antibiotic and antifungal applications. Lectins are promising antiviral agents against viruses with epidemics and drug resistance concerns. Mannose-binding lectins were the most suggested antiviral agents since glycans with mannose residues are commonly involved in viral entry mechanisms. They were also immobilized onto surfaces to trap viral particles and inhibit their spread and replication. Many patents described the extraction, isolation, amino acid and nucleotide sequences, and expression vectors of lectins with antibiotic and/or antifungal activities in terms of MIC and IC50 for in vitro assays. The inventions also included lectins as biological tools in nanosensors for antibiotics susceptibility tests, drug-delivery systems for the treatment of resistant bacteria, diagnostics of viral diseases and as a vaccine adjuvant. Although research and development of new medicines is highly expensive, antimicrobial lectins may be worth investments given the emergence of epidemics and drug resistance. For this purpose, less invasive routes should be developed as alternatives to the parenteral administration of biologics. While anti-glycan neutralizing antibodies are difficult to develop due to the low immunogenicity of carbohydrates, lectins can be produced more easily and have a broad-spectrum activity. Protein engineering technologies may make the antimicrobial applications of lectins more successful.
Collapse
Affiliation(s)
- Diego C Carneiro
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Brazil
| | - Luzimar G Fernandez
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Brazil
| | - Joana P Monteiro-Cunha
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Brazil
| | - Raquel G Benevides
- Department of Biological Sciences, State University of Feira de Santana, Feira de Santana, Brazil
| | | |
Collapse
|
15
|
Sarker S, Wankum B, Shimizu J, Jones R, Terry B. A Factorial Approach for Optimizing the Design Parameters of a Tissue Attachment Mechanism for Drug Delivery. IEEE Trans Biomed Eng 2021; 69:32-41. [PMID: 34097601 DOI: 10.1109/tbme.2021.3086975] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Biological macromolecule drugs or biologics are not suited for commonly preferred oral delivery due to their intrinsic instability and physical, chemical, or immunological barriers to the gastrointestinal tract. Ingestible capsule robots (ICR) have become a versatile platform, including use for drug delivery applications for various gastrointestinal pathologies with future potential for systemic drug delivery. In this work, a tissue attachment mechanism (TAM) for a drug delivery ICR is introduced that can facilitate a non-invasive systemic delivery of unaltered biologics via direct injection through the insensate layers of the small intestine. The main prerequisite for achieving systemic drug delivery via this device is to have strong tissue attachment of the TAM. This study aimed to optimize the attachment success rate for drug delivery and characterize attachment duration in vivo. A fractional factorial approach was used in vivo to identify and optimize factors that most influence attachment of the TAM to maximize attachment rate. Multiple in vivo optimization levels were performed using the small intestine of anesthetized pigs, and an attachment success rate of 92% was achieved. Optimal TAMs were surgically placed in vivo to determine the duration of attachment following anesthetization and surgery recovery. The average in vivo attachment duration was 32.29.4 hours. This work establishes a device for consistent and reliable attachment duration, making the TAM a suitable candidate for a 24-hour systemic drug delivery platform.
Collapse
|
16
|
Shah V, Jobanputra A, Saxena B, Nivsarkar M. Development and Characterization of Saturated Fatty Acid-Engineered, Silica-Coated Lipid Vesicular System for Effective Oral Delivery of Alfa-Choriogonadotropin. AAPS PharmSciTech 2021; 22:118. [PMID: 33782790 DOI: 10.1208/s12249-021-01985-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/08/2021] [Indexed: 11/30/2022] Open
Abstract
The present study was designed to develop an efficient, safe, and patient-friendly dosage form, for oral delivery of alfa-choriogonadotropin, used in the treatment of female reproductive infertility. Silica-coated, saturated fatty acid (dipalmitoylphosphatidylcholine (DPPC))-engineered, nanolipidic vesicular (NLVs) system was developed for systemic delivery of therapeutic peptide, alfa-choriogonadotropin, through oral route. DPPC-based NLVs were formulated using the technique of thin-film hydration and were coated with silica to form a homogeneous surface silica shell. The formulated silica-coated NLVs were evaluated for physicochemical and physiologic stability under simulated conditions and were optimized based on physicochemical parameters like particle size, zeta potential, polydispersity index (PDI), entrapment efficiency, and in vitro release profile. Silica-coated, DPPC-based NLVs imparted physicochemical stability to entrapped alfa-choriogonadotropin against the biological environment prevailing in the human gastrointestinal tract (GIT). In vivo, subchronic animal toxicity studies were performed to assess the safety of the designed dosage form. Results of in vitro characterization and in vivo pharmacokinetic studies of fabricated formulation revealed that the silica-coated, DPPC-based NLV formulation was not only stable in human GIT but was also as efficacious as a marketed parenteral formulation for the systemic delivery of alfa-choriogonadotropin. In vivo toxicity studies revealed that silica-coated NLVs did not alter hematological and serum biochemical parameters. The histopathological studies also depicted no macroscopic changes in major organs; thus, the developed formulation was proven to be nontoxic and equally efficient as a marketed parenteral formulation for the delivery of alfa-choriogonadotropin with added benefits of possible self-medication, more patient acceptability, and no chances of infection.
Collapse
|
17
|
Sanabria R. Nanotechnological Improvement of Veterinary Anthelmintics. Pharm Nanotechnol 2021; 9:5-14. [PMID: 32448112 DOI: 10.2174/2211738508666200524233724] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/31/2020] [Accepted: 04/27/2020] [Indexed: 11/22/2022]
Abstract
Helminths infections are among the most important problems in animal health and husbandry. Moreover, zoonotic helminths endanger rural communities, particularly in developing countries. Helminthiasis are not only important in relation to the harmful effects of parasites; additional issues like anthelmintic resistance spread became more important over time. As new anthelmintic development takes many years and millions of dollars of investment, some strategies are currently focused on the modification of already available drugs, in order to improve their efficacy and overcome their limitations. In this field, nanotechnology has brought a novel approach, showing advantages like the regulation of the drug's delivery and kinetics, reaching of specific targets, and possibilities to avoid the systemic spread and side effects. Taking this into account, the present review aims to introduce some of the current knowledge in anthelmintic improvement based on nanotechnology, and how researchers could benefit from this technology in order to overcome the drugs limitations. Finally, some insights into potential field applications are discussed, based on the most important concerns of current anthelmintic therapy.
Collapse
Affiliation(s)
- Rodrigo Sanabria
- Instituto Tecnologico Chascomus (INTECH)-CONICET-UNSAM. Av. Marino KM 8.2, (7130), Chascomús, Argentina
| |
Collapse
|
18
|
Ferrer‐Tasies L, Santana H, Cabrera‐Puig I, González‐Mira E, Ballell‐Hosa L, Castellar‐Álvarez C, Córdoba A, Merlo‐Mas J, Gerónimo H, Chinea G, Falcón V, Moreno‐Calvo E, Pedersen JS, Romero J, Navarro‐Requena C, Valdés C, Limonta M, Berlanga J, Sala S, Martínez E, Veciana J, Ventosa N. Recombinant Human Epidermal Growth Factor/Quatsome Nanoconjugates: A Robust Topical Delivery System for Complex Wound Healing. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000260] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Lidia Ferrer‐Tasies
- Institut de Ciència de Materials de Barcelona ICMAB‐CSIC/CIBER‐BBN Campus Universitari Bellaterra 08193 Spain
- Nanomol Technologies S.L. Campus UAB Bellaterra 08193 Spain
| | - Hector Santana
- Center for Genetic Engineering and Biotechnology (CIGB) 31st Avenue between 158 and 190 Streets, Cubanacán, Playa Havana 10600 Cuba
| | - Ingrid Cabrera‐Puig
- Institut de Ciència de Materials de Barcelona ICMAB‐CSIC/CIBER‐BBN Campus Universitari Bellaterra 08193 Spain
| | - Elisabet González‐Mira
- Institut de Ciència de Materials de Barcelona ICMAB‐CSIC/CIBER‐BBN Campus Universitari Bellaterra 08193 Spain
| | - Lídia Ballell‐Hosa
- Institut de Ciència de Materials de Barcelona ICMAB‐CSIC/CIBER‐BBN Campus Universitari Bellaterra 08193 Spain
- Nanomol Technologies S.L. Campus UAB Bellaterra 08193 Spain
| | | | - Alba Córdoba
- Nanomol Technologies S.L. Campus UAB Bellaterra 08193 Spain
| | | | - Haydee Gerónimo
- Center for Genetic Engineering and Biotechnology (CIGB) 31st Avenue between 158 and 190 Streets, Cubanacán, Playa Havana 10600 Cuba
| | - Glay Chinea
- Center for Genetic Engineering and Biotechnology (CIGB) 31st Avenue between 158 and 190 Streets, Cubanacán, Playa Havana 10600 Cuba
| | - Viviana Falcón
- Center for Genetic Engineering and Biotechnology (CIGB) 31st Avenue between 158 and 190 Streets, Cubanacán, Playa Havana 10600 Cuba
| | - Evelyn Moreno‐Calvo
- Institut de Ciència de Materials de Barcelona ICMAB‐CSIC/CIBER‐BBN Campus Universitari Bellaterra 08193 Spain
| | - Jan Skov Pedersen
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO) Aarhus University Gustav Wieds Vej 14 Aarhus C DK‐8000 Denmark
| | - Jessica Romero
- Health and Biomedicine Unit LEITAT Technological Center C/ de la Innovació, 2 Terrassa Barcelona 08225 Spain
| | - Claudia Navarro‐Requena
- Health and Biomedicine Unit LEITAT Technological Center C/ de la Innovació, 2 Terrassa Barcelona 08225 Spain
| | - Calixto Valdés
- National Institute for Angiology and Vascular Surgery 1551 Calzada del Cerro, Cerro Havana 12000 Cuba
| | - Miladys Limonta
- Center for Genetic Engineering and Biotechnology (CIGB) 31st Avenue between 158 and 190 Streets, Cubanacán, Playa Havana 10600 Cuba
| | - Jorge Berlanga
- Center for Genetic Engineering and Biotechnology (CIGB) 31st Avenue between 158 and 190 Streets, Cubanacán, Playa Havana 10600 Cuba
| | - Santiago Sala
- Nanomol Technologies S.L. Campus UAB Bellaterra 08193 Spain
| | - Eduardo Martínez
- Center for Genetic Engineering and Biotechnology (CIGB) 31st Avenue between 158 and 190 Streets, Cubanacán, Playa Havana 10600 Cuba
| | - Jaume Veciana
- Institut de Ciència de Materials de Barcelona ICMAB‐CSIC/CIBER‐BBN Campus Universitari Bellaterra 08193 Spain
| | - Nora Ventosa
- Institut de Ciència de Materials de Barcelona ICMAB‐CSIC/CIBER‐BBN Campus Universitari Bellaterra 08193 Spain
| |
Collapse
|
19
|
Farinha S, Moura C, Afonso MD, Henriques J. Production of Lysozyme-PLGA-Loaded Microparticles for Controlled Release Using Hot-Melt Extrusion. AAPS PharmSciTech 2020; 21:274. [PMID: 33033873 DOI: 10.1208/s12249-020-01816-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/08/2020] [Indexed: 01/03/2023] Open
Abstract
Biopharmaceuticals are usually administered intravenously with frequent dosing regimens which may decrease patient compliance. Controlled-release formulations allow to reduce the frequency of injections while providing a constant dosing of the biopharmaceutical over extended periods. These formulations are typically produced by emulsions, requiring high amounts of organic solvents and have limited productivity. Hot-melt extrusion (HME) is an alternative technology to produce controlled drug delivery systems. It is a continuous solvent-free process, leading to a small ecological footprint and higher productivity. However, it may induce thermolabile compounds' degradation. In this work, the impact of the formulation and extrusion temperature on lysozyme's bioactivity and release profile of poly(lactic-co-glycolic acid) (PLGA)-based extended release formulations were evaluated using a design-of-experiments (DoE) approach. The lysozyme-loaded PLGA microparticles were produced by HME followed by milling. It was observed that the in vitro release (IVR) profile was mainly affected by the drug load; higher drug load led to higher burst and total lysozyme release after 14 days. HME temperature seemed to decrease lysozyme's activity although this correlation was not statistically significant (p value = 0.0490). Adding polyethylene glycol 400 (PEG 400) as a plasticizer to the formulation had no significant impact on the lysozyme release profile. The burst release was effectively mitigated with the inclusion of a washing step. Washing the microparticles with water reduced the burst release by 80% whereas washing them with a poly(vinyl alcohol) (PVA) aqueous solution eliminated it. In conclusion, HME is demonstrated to be suitable in producing controlled-release microparticles of small biopharmaceuticals. Graphical abstract.
Collapse
|
20
|
Swanson WB, Gong T, Zhang Z, Eberle M, Niemann D, Dong R, Rambhia KJ, Ma PX. Controlled release of odontogenic exosomes from a biodegradable vehicle mediates dentinogenesis as a novel biomimetic pulp capping therapy. J Control Release 2020; 324:679-694. [PMID: 32534011 PMCID: PMC7429296 DOI: 10.1016/j.jconrel.2020.06.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
Abstract
Mineralized enamel and dentin provide protection to the dental pulp, which is vital tissue rich with cells, vasculature, and nerves in the inner tooth. Dental caries left untreated threaten exposure of the dental pulp, providing facile access for bacteria to cause severe infection both in the pulp and systemically. Dental materials which stimulate the formation of a protective dentin bridge after insult are necessary to seal the pulp chamber in an effort to maintain natural dentition and prevent pulpal infection. Dental materials to date including calcium hydroxide paste, mineral trioxide aggregate, and glass ionomer resin, are used with mixed results. Herein we exploited the cell-cell communicative properties of exosomes, extracellular vesicles derived from both mineralizing primary human dental pulp stem cells (hDPSCs) and an immortalized murine odontoblast cell line (MDPC-23), to catalyze the formation of a reactionary dentin bridge by recruiting endogenous stem cells of the dental pulp, through an easy-to-handle delivery vehicle which allows for their therapeutic controlled delivery at the pulp interface. Exosomes derived from both hDPSCs and MDPCs upregulated odontogenic gene expression and increased mineralization in vitro. We designed an amphiphilic synthetic polymeric vehicle from a triblock copolymer which encapsulates exosomes by polymeric self-assembly and maintains their biologic integrity throughout release up to 8-12 weeks. The controlled release of odontogenic exosomes resulted in a reparative dentin bridge formation, superior to glass-ionomer cement alone in vivo, in a rat molar pulpotomy model after six weeks. We have developed a platform for the encapsulation and controlled, tunable release of cell-derived exosomes, which maintains their advantageous physiologic properties reflective of the donor cells. This platform is used to modulate downstream recipient cells towards a designed dentinogenic trajectory in vitro and in vivo. Additionally, we have demonstrated the utility of an immortalized cell line to produce a high yield of exosomes with cross-species efficacy.
Collapse
Affiliation(s)
- W Benton Swanson
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, United States of America
| | - Ting Gong
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, United States of America
| | - Zhen Zhang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, United States of America
| | - Miranda Eberle
- Department of Chemistry, College of Literature, Science, and the Arts, University of Michigan, United States of America
| | - David Niemann
- Department of Chemistry, College of Literature, Science, and the Arts, University of Michigan, United States of America
| | - Ruonan Dong
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, United States of America
| | - Kunal J Rambhia
- Department of Biomedical Engineering, School of Medicine and College of Engineering, University of Michigan, United States of America
| | - Peter X Ma
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, United States of America; Macromolecular Science and Engineering Center, College of Engineering, University of Michigan, United States of America; Department of Biomedical Engineering, School of Medicine and College of Engineering, University of Michigan, United States of America; Department of Materials Science and Engineering, College of Engineering, University of Michigan, United States of America.
| |
Collapse
|
21
|
Yang M, Byrn SR, Clase KL. An Analytic Investigation of the Drug Formulation-Based Recalls in the USA: See More Beyond the Literal. AAPS PharmSciTech 2020; 21:198. [PMID: 32676955 DOI: 10.1208/s12249-020-01726-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 06/08/2020] [Indexed: 11/30/2022] Open
Abstract
High numbers of drug recalls persist despite the tremendous time and effort invested by pharmaceutical organizations and regulatory bodies such as the Food and Drug Administration (FDA) to ensure the quality of safe and effective medicines for the patient. It is imperative to better understand the underlying risk factors of drug formulation-based recalls to best protect the patient from poor quality drugs. Increased knowledge of underlying factors of formulation risk can also help inform the future design and development of drugs. In this study, we used a text mining technique with Python to parse the data and examine drug recalls from the aspect of administration route, dosage form, release mechanism, market type, pharmacologic class, and excipients. Observational analysis of the recalls revealed both high- and low-risk factors for the formulation-based recalls. Higher risk, or an increased probability of a formulation-based recall, was associated with factors such as extended release mechanism, capsule dosage form, oral route of administration, and an increased number of excipients, while lower risk of formulation-based recalls was associated with other factors including the new drug application market type, immediate release mechanism, and solution dosage form. In addition, the factors did not work independently, and we observed interactions among variables. For example, the release mechanism modified the effect of market type, administration route, and dosage form. This study will help inform the future design of quality drug products by pharmaceutical organizations and assist risk-based oversight by regulatory organizations, such as FDA, to ensure patient safety.
Collapse
|
22
|
Young CC, Vedadghavami A, Bajpayee AG. Bioelectricity for Drug Delivery: The Promise of Cationic Therapeutics. Bioelectricity 2020; 2:68-81. [PMID: 32803148 DOI: 10.1089/bioe.2020.0012] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Biological systems overwhelmingly comprise charged entities generating electrical activity that can have significant impact on biological structure and function. This intrinsic bio-electrical activity can also be harnessed for overcoming the tissue matrix and cell membrane barriers, which have been outstanding challenges for targeted drug delivery, by using rationally designed cationic carriers. The weak and reversible long-range electrostatic interactions with fixed negatively charged groups facilitate electro-diffusive transport of cationic therapeutics through full-tissue thickness to effectively reach intra-tissue, cellular, and intracellular target sites. This article presents a perspective on the promise of using rationally designed cationic biomaterials in targeted drug delivery, the underlying charge-based mechanisms, and bio-transport phenomena while addressing outstanding concerns around toxicity and methods to mitigate them. We also discuss electrically charged drugs that are currently being evaluated in clinical trials and identify areas of further development that have the potential to usher in new treatments.
Collapse
Affiliation(s)
- Cameron C Young
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Armin Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA.,Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Targeted drug-delivery systems in the treatment of rheumatoid arthritis: recent advancement and clinical status. Ther Deliv 2020; 11:269-284. [DOI: 10.4155/tde-2020-0029] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease that is characterized by synovial inflammation, cellular infiltration in joints which leads to progressive joint destruction and bone erosion. RA is associated with many comorbidities including pulmonary disease, rheumatoid nodules and can have a pessimistic impact on quality of life. The current therapies of RA treatment comprise conventional, small molecule and biological antirheumatic drugs. Their utility as therapeutic agents is limited because of poor absorption, rapid metabolism and adverse effects (dose-escalation, systemic toxicity, lack of selectivity and safety). To overcome these limitations, the novel drug delivery systems are being investigated. This review has compiled currently approved therapies along with emerging advanced drug-delivery systems for RA treatment. Further, active targeting of therapeutic agents to inflamed joints via folate receptor, CD44, angiogenesis, integrins and other provided an improved therapeutic efficacy in the treatment of RA.
Collapse
|
24
|
Evans BC, Fletcher RB, Kilchrist KV, Dailing EA, Mukalel AJ, Colazo JM, Oliver M, Cheung-Flynn J, Brophy CM, Tierney JW, Isenberg JS, Hankenson KD, Ghimire K, Lander C, Gersbach CA, Duvall CL. An anionic, endosome-escaping polymer to potentiate intracellular delivery of cationic peptides, biomacromolecules, and nanoparticles. Nat Commun 2019; 10:5012. [PMID: 31676764 PMCID: PMC6825215 DOI: 10.1038/s41467-019-12906-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 10/07/2019] [Indexed: 12/21/2022] Open
Abstract
Peptides and biologics provide unique opportunities to modulate intracellular targets not druggable by conventional small molecules. Most peptides and biologics are fused with cationic uptake moieties or formulated into nanoparticles to facilitate delivery, but these systems typically lack potency due to low uptake and/or entrapment and degradation in endolysosomal compartments. Because most delivery reagents comprise cationic lipids or polymers, there is a lack of reagents specifically optimized to deliver cationic cargo. Herein, we demonstrate the utility of the cytocompatible polymer poly(propylacrylic acid) (PPAA) to potentiate intracellular delivery of cationic biomacromolecules and nano-formulations. This approach demonstrates superior efficacy over all marketed peptide delivery reagents and enhances delivery of nucleic acids and gene editing ribonucleoproteins (RNPs) formulated with both commercially-available and our own custom-synthesized cationic polymer delivery reagents. These results demonstrate the broad potential of PPAA to serve as a platform reagent for the intracellular delivery of cationic cargo.
Collapse
Affiliation(s)
- Brian C Evans
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB351826, Nashville, TN, 37235, USA.
| | - R Brock Fletcher
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB351826, Nashville, TN, 37235, USA
| | - Kameron V Kilchrist
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB351826, Nashville, TN, 37235, USA
| | - Eric A Dailing
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB351826, Nashville, TN, 37235, USA
| | - Alvin J Mukalel
- Department of Bioengineering, University of Pennsylvania, 2301 240 Skirkanich Hall, 210S. 33rd Street, Philadelphia, PA, 19104-6321, USA
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB351826, Nashville, TN, 37235, USA
- Vanderbilt University School of Medicine, Vanderbilt University, 1161 21st Avenue South # D3300, Nashville, TN, 37232, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Matthew Oliver
- Program in Cell and Molecular Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, D-5237 Medical Center North, 1161 22nd Avenue South, Nashville, TN, 37232, USA
| | - Colleen M Brophy
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, D-5237 Medical Center North, 1161 22nd Avenue South, Nashville, TN, 37232, USA
- Veterans Affairs Medical Center, VA Tennessee Valley Healthcare System, 1310 24th Avenue South, Nashville, TN, 37212, USA
| | - John W Tierney
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB351826, Nashville, TN, 37235, USA
| | - Jeffrey S Isenberg
- Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, 4200 Fifth Avenue, Pittsburgh, PA, 15260, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Kedar Ghimire
- University of Pittsburgh School of Medicine, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Cynthia Lander
- Moerae Matrix Inc., 55 Madison Avenue, Suite 400, Morristown, NJ, 07960, USA
| | - Charles A Gersbach
- Center for Genomic and Computational Biology, Duke University, Durham, NC, 27710, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, 27710, USA
- Department of Orthopaedic Surgery, Duke University, Durham, NC, 27710, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB351826, Nashville, TN, 37235, USA.
| |
Collapse
|
25
|
Lofchy LA, Vu VP, Banda NK, Ramirez JR, Smith WJ, Gifford G, Gaikwad H, Scheinman RI, Simberg D. Evaluation of Targeting Efficiency of Joints with Anticollagen II Antibodies. Mol Pharm 2019; 16:2445-2451. [PMID: 31091104 DOI: 10.1021/acs.molpharmaceut.9b00059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diseases of the joints affect over 10% of the world's population, resulting in significant morbidity. There is an unmet need in strategies for specific delivery of therapeutics to the joints. Collagen type II is synthesized by chondrocytes and is mainly restricted to the cartilage and tendons. Arthrogen-CIA is a commercially available anticollagen II antibody cocktail that reacts with 5 different epitopes on human, bovine, and mouse collagen II. Arthrogen has been used for induction of experimental rheumatoid arthritis (RA) in mice because of high complement activation on the cartilage surface. Native collagen II might serve as a useful target for potential delivery of therapeutics to the joint. To evaluate the efficiency and specificity of targeting collagen II, Arthrogen was labeled with near-infrared (NIR) dye IRDye 800 or IRDye 680. Using ex vivo NIR imaging, we demonstrate that Arthrogen efficiently and specifically accumulated in the limb joints regardless of the label dye or injection route (intravenous and subcutaneous). After subcutaneous injection, the mean fluorescence of the hind limb joints was 19 times higher than that of the heart, 8.7 times higher than that of the liver, and 3.7 times higher than that of the kidney. Control mouse IgG did not show appreciable accumulation. Microscopically, the antibody accumulated on the cartilage surface of joints and on endosteal surfaces. A monoclonal antibody against a single epitope of collagen II showed similar binding affinity and elimination half-life, but about three times lower targeting efficiency than Arthrogen in vitro and ex vivo, and about two times lower targeting efficiency in vivo. We suggest that an antibody against multiple epitopes of collagen II could be developed into a highly effective and specific targeting strategy for diseases of the joints or spine.
Collapse
Affiliation(s)
| | | | - Nirmal K Banda
- Division of Rheumatology, School of Medicine , University of Colorado Denver , Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Joseline Ramos Ramirez
- Division of Rheumatology, School of Medicine , University of Colorado Denver , Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | | | | | | | | | | |
Collapse
|
26
|
Enhancing bioactivity, physicochemical, and pharmacokinetic properties of a nano-sized, anti-VEGFR2 Adnectin, through PASylation technology. Sci Rep 2019; 9:2978. [PMID: 30814652 PMCID: PMC6393559 DOI: 10.1038/s41598-019-39776-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/01/2019] [Indexed: 02/07/2023] Open
Abstract
The crucial role of VEGF receptor 2 (VEGFR2) signaling in the angiogenesis and metastasis of solid tumors has prompted the development of inhibitors with minimal bystander effects. Recently, Adnectin C has attracted attention for cancer treatment. To overcome the problematic properties of Adnectin, a novel form of Adnectin C has been designed by its fusion to a biodegradable polymeric peptide containing Pro/Ala/Ser (PAS) repetitive residues. E. coli-expressed recombinant fused and unfused proteins were compared in terms of bioactivity, physicochemical, and pharmacokinetic properties using standard methods. Dynamic light scattering (DLS) analysis of PASylated adnectin C revealed an approximate 2-fold increase in particle size with a slight change in the net charge. Additionally, fusion of the PAS sequence improved its stability against the growth of thermo-induced aggregated forms. The high receptor-binding and improved binding kinetic parameters of PASylated Adnectin C was confirmed by ELISA and surface plasmon resonance assays, respectively. Pharmacokinetic studies showed a noticeable increase in the terminal half-life of Adnectin C-PAS#1(200) by a factor of 4.57 after single dose by intravenous injection into female BALB/c mice. The results suggest that PASylation could offer a superior delivery strategy for developing Adnectin-derived drugs with improved patient compliance.
Collapse
|
27
|
Inchaurraga L, Martínez-López AL, Abdulkarim M, Gumbleton M, Quincoces G, Peñuelas I, Martin-Arbella N, Irache JM. Modulation of the fate of zein nanoparticles by their coating with a Gantrez® AN-thiamine polymer conjugate. INTERNATIONAL JOURNAL OF PHARMACEUTICS-X 2019; 1:100006. [PMID: 31517271 PMCID: PMC6733281 DOI: 10.1016/j.ijpx.2019.100006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 01/01/2023]
Abstract
The aim of this work was to evaluate the mucus-permeating properties of nanocarriers using zein nanoparticles (NPZ) coated with a Gantrez® AN-thiamine conjugate (GT). NPZ were coated by incubation at different GT-to-zein ratios: 2.5% coating with GT (GT-NPZ1), 5% (GT-NPZ2) and 10% (GT-NPZ3). During the process, the GT conjugate formed a polymer layer around the surface of zein nanoparticles. For GT-NPZ2, the thickness of this corona was estimated between 15 and 20 nm. These nanocarriers displayed a more negative zeta potential than uncoated NPZ. The diffusivity of nanoparticles was evaluated in pig intestinal mucus by multiple particle tracking analysis. GT-NPZ2 displayed a 28-fold higher diffusion coefficient within the mucus layer than NPZ particles. These results align with in vivo biodistribution studies in which NPZ displayed a localisation restricted to the mucus layer, whereas GT-NPZ2 were capable of reaching the intestinal epithelium. The gastro-intestinal transit of mucoadhesive (NPZ) and mucus-permeating nanoparticles (GT-NPZ2) was also found to be different. Thus, mucoadhesive nanoparticles displayed a significant accumulation in the stomach of animals, whereas mucus-penetrating nanoparticles appeared to exit the stomach more rapidly to access the small intestine of animals.
Collapse
Affiliation(s)
- Laura Inchaurraga
- NANO-VAC Research Group, Department of Chemistry and Pharmaceutical Technology, University of Navarra, Spain
| | - Ana L Martínez-López
- NANO-VAC Research Group, Department of Chemistry and Pharmaceutical Technology, University of Navarra, Spain
| | - Muthanna Abdulkarim
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Gemma Quincoces
- Radiopharmacy Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, University of Navarra, Spain
| | - Ivan Peñuelas
- Radiopharmacy Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, University of Navarra, Spain
| | - Nekane Martin-Arbella
- NANO-VAC Research Group, Department of Chemistry and Pharmaceutical Technology, University of Navarra, Spain
| | - Juan M Irache
- NANO-VAC Research Group, Department of Chemistry and Pharmaceutical Technology, University of Navarra, Spain
| |
Collapse
|
28
|
Lu H, Chelvanambi S, Poirier C, Saliba J, March KL, Clauss M, Bogatcheva NV. EMAPII Monoclonal Antibody Ameliorates Influenza A Virus-Induced Lung Injury. Mol Ther 2018; 26:2060-2069. [PMID: 29910176 PMCID: PMC6094359 DOI: 10.1016/j.ymthe.2018.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/14/2018] [Accepted: 05/21/2018] [Indexed: 01/27/2023] Open
Abstract
Influenza A virus (IAV) remains a major worldwide health threat, especially to high-risk populations, including the young and elderly. There is an unmet clinical need for therapy that will protect the lungs from damage caused by lower respiratory infection. Here, we analyzed the role of EMAPII, a stress- and virus-induced pro-inflammatory and pro-apoptotic factor, in IAV-induced lung injury. First, we demonstrated that IAV induces EMAPII surface translocation, release, and apoptosis in cultured endothelial and epithelial cells. Next, we showed that IAV induces EMAPII surface translocation and release to bronchoalveolar lavage fluid (BALF) in mouse lungs, concomitant with increases in caspase 3 activity. Injection of monoclonal antibody (mAb) against EMAPII attenuated IAV-induced EMAPII levels, weight loss, reduction of blood oxygenation, lung edema, and increase of the pro-inflammatory cytokine TNF alpha. In accordance with the pro-apoptotic properties of EMAPII, levels of caspase 3 activity in BALF were also decreased by mAb treatment. Moreover, we detected EMAPII mAb-induced increase in lung levels of M2-like macrophage markers YM1 and CD206. All together, these data strongly suggest that EMAPII mAb ameliorates IAV-induced lung injury by limiting lung cell apoptosis and shifting the host inflammatory setting toward resolution of inflammation.
Collapse
Affiliation(s)
- Hongyan Lu
- Division of Cardiology, Indiana University School of Medicine, Indianapolis, IN, USA; VC-CAST Signature Center, Indianapolis, IN, USA; Roudebush Veterans Affairs Medical Center, Indiana University, Indianapolis, IN, USA
| | - Sarvesh Chelvanambi
- VC-CAST Signature Center, Indianapolis, IN, USA; Roudebush Veterans Affairs Medical Center, Indiana University, Indianapolis, IN, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christophe Poirier
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jacob Saliba
- Division of Pulmonary and Critical Care Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Keith L March
- Division of Cardiology, Indiana University School of Medicine, Indianapolis, IN, USA; VC-CAST Signature Center, Indianapolis, IN, USA; Roudebush Veterans Affairs Medical Center, Indiana University, Indianapolis, IN, USA
| | - Matthias Clauss
- VC-CAST Signature Center, Indianapolis, IN, USA; Roudebush Veterans Affairs Medical Center, Indiana University, Indianapolis, IN, USA; Division of Pulmonary and Critical Care Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Natalia V Bogatcheva
- Division of Cardiology, Indiana University School of Medicine, Indianapolis, IN, USA; VC-CAST Signature Center, Indianapolis, IN, USA; Roudebush Veterans Affairs Medical Center, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
29
|
Le TMD, Duong HTT, Thambi T, Giang Phan V, Jeong JH, Lee DS. Bioinspired pH- and Temperature-Responsive Injectable Adhesive Hydrogels with Polyplexes Promotes Skin Wound Healing. Biomacromolecules 2018; 19:3536-3548. [DOI: 10.1021/acs.biomac.8b00819] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Thai Minh Duy Le
- School of Chemical Engineering, Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Huu Thuy Trang Duong
- School of Chemical Engineering, Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Thavasyappan Thambi
- School of Chemical Engineering, Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - V.H. Giang Phan
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 70000, Vietnam
| | - Ji Hoon Jeong
- School of Pharmacy, Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Doo Sung Lee
- School of Chemical Engineering, Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| |
Collapse
|
30
|
Vargason AM, Anselmo AC. Clinical translation of microbe-based therapies: Current clinical landscape and preclinical outlook. Bioeng Transl Med 2018; 3:124-137. [PMID: 30065967 PMCID: PMC6063871 DOI: 10.1002/btm2.10093] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 12/11/2022] Open
Abstract
Next generation microbe-based therapeutics, inspired by the success of fecal microbiota transplants, are being actively investigated in clinical trials to displace or eliminate pathogenic microbes to treat various diseases in the gastrointestinal tract, skin, and vagina. Genetically engineered microbes are also being investigated in the clinic as drug producing factories for biologic delivery, which can provide a constant local source of drugs. In either case, microbe-therapeutics have the opportunity to address unmet clinical needs and open new areas of research by reducing clinical side effects associated with current treatment modalities or by facilitating the delivery of biologics. This review will discuss examples of past and current clinical trials that are investigating microbe-therapeutics, both microbiome-modulating and drug-producing, for the treatment of a range of diseases. We then offer a perspective on how preclinical approaches, both those focused on developing advanced delivery systems and those that use in vitro microbiome model systems to inform formulation design, will lead to the realization of next-generation microbe-therapeutics.
Collapse
Affiliation(s)
- Ava M. Vargason
- Div. of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNC 27599
| | - Aaron C. Anselmo
- Div. of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNC 27599
| |
Collapse
|
31
|
Saraiva RG, Fang J, Kang S, Angleró-Rodríguez YI, Dong Y, Dimopoulos G. Aminopeptidase secreted by Chromobacterium sp. Panama inhibits dengue virus infection by degrading the E protein. PLoS Negl Trop Dis 2018; 12:e0006443. [PMID: 29694346 PMCID: PMC5937796 DOI: 10.1371/journal.pntd.0006443] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 05/07/2018] [Accepted: 04/10/2018] [Indexed: 12/14/2022] Open
Abstract
Dengue virus (DENV) is the most prevalent and burdensome arbovirus transmitted by Aedes mosquitoes, against which there is only a limited licensed vaccine and no approved drug treatment. A Chromobacterium species, C. sp. Panama, isolated from the midgut of A. aegypti is able to inhibit DENV replication within the mosquito and in vitro. Here we show that C. sp. Panama mediates its anti-DENV activity through secreted factors that are proteinous in nature. The inhibitory effect occurs prior to virus attachment to cells, and is attributed to a factor that destabilizes the virion by promoting the degradation of the viral envelope protein. Bioassay-guided fractionation, coupled with mass spectrometry, allowed for the identification of a C. sp. Panama-secreted neutral protease and an aminopeptidase that are co-expressed and appear to act synergistically to degrade the viral envelope (E) protein and thus prevent viral attachment and subsequent infection of cells. This is the first study characterizing the anti-DENV activity of a common soil and mosquito-associated bacterium, thereby contributing towards understanding how such bacteria may limit disease transmission, and providing new tools for dengue prevention and therapeutics.
Collapse
Affiliation(s)
- Raúl G. Saraiva
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Jingru Fang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Seokyoung Kang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Yesseinia I. Angleró-Rodríguez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- * E-mail:
| |
Collapse
|
32
|
Sharpe LA, Vela Ramirez JE, Haddadin OM, Ross KA, Narasimhan B, Peppas NA. pH-Responsive Microencapsulation Systems for the Oral Delivery of Polyanhydride Nanoparticles. Biomacromolecules 2018; 19:793-802. [PMID: 29443509 DOI: 10.1021/acs.biomac.7b01590] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Multicompartmental polymer carriers, referred to as Polyanhydride-Releasing Oral MicroParticle Technology (PROMPT), were formed by a pH-triggered antisolvent precipitation technique. Polyanhydride nanoparticles were encapsulated into anionic pH-responsive microparticle gels, allowing for nanoparticle encapsulation in acidic conditions and subsequent release in neutral pH conditions. The effects of varying the nanoparticle composition and feed ratio on the encapsulation efficiency were evaluated. Nanoparticle encapsulation was confirmed by confocal microscopy and infrared spectroscopy. pH-triggered protein delivery from PROMPT was explored using ovalbumin (ova) as a model drug. PROMPT microgels released ova in a pH-controlled manner. Increasing the feed ratio of nanoparticles into the microgels increased the total amount of ova delivered, as well as decreased the observed burst release. The cytocompatibility of the polymer materials were assessed using cells representative of the GI tract. Overall, these results suggest that pH-dependent microencapsulation is a viable platform to achieve targeted intestinal delivery of polyanhydride nanoparticles and their payload(s).
Collapse
Affiliation(s)
| | | | | | - Kathleen A Ross
- Department of Chemical and Biological Engineering , Iowa State University , Ames , Iowa 50011 , United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering , Iowa State University , Ames , Iowa 50011 , United States
| | | |
Collapse
|
33
|
Shelley MY, Selvan ME, Zhao J, Babin V, Liao C, Li J, Shelley JC. A New Mixed All-Atom/Coarse-Grained Model: Application to Melittin Aggregation in Aqueous Solution. J Chem Theory Comput 2017; 13:3881-3897. [PMID: 28636825 PMCID: PMC5551643 DOI: 10.1021/acs.jctc.7b00071] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Indexed: 11/28/2022]
Abstract
We introduce a new mixed resolution, all-atom/coarse-grained approach (AACG), for modeling peptides in aqueous solution and apply it to characterizing the aggregation of melittin. All of the atoms in peptidic components are represented, while a single site is used for each water molecule. With the full flexibility of the peptide retained, our AACG method achieves speedups by a factor of 3-4 for CPU time reduction and another factor of roughly 7 for diffusion. An Ewald treatment permits the inclusion of long-range electrostatic interactions. These characteristics fit well with the requirements for studying peptide association and aggregation, where the system sizes and time scales require considerable computational resources with all-atom models. In particular, AACG is well suited for biologics since changes in peptide shape and long-range electrostatics may play an important role. The application of AACG to melittin, a 26-residue peptide with a well-known propensity to aggregate in solution, serves as an initial demonstration of this technology for studying peptide aggregation. We observed the formation of melittin aggregates during our simulations and characterized the time-evolution of aggregate size distribution, buried surface areas, and residue contacts. Key interactions including π-cation and π-stacking involving TRP19 were also examined. Our AACG simulations demonstrated a clear salt effect and a moderate temperature effect on aggregation and support the molten globule model of melittin aggregates. As a showcase, this work illustrates the useful role for AACG in investigations of peptide aggregation and its potential to guide formulation and design of biologics.
Collapse
Affiliation(s)
- Mee Y. Shelley
- Schrödinger,
Inc., 101 SW Main Street,
Suite 1300, Portland, Oregon 97204, United States
| | - Myvizhi Esai Selvan
- Schrödinger,
Inc., 120 W. 45th Street,
17th Floor, New York, New
York 10036, United
States
| | - Jun Zhao
- Cancer
and Inflammation Program, National Cancer
Institute, Frederick, Maryland 21702, United
States
| | - Volodymyr Babin
- Schrödinger,
Inc., 101 SW Main Street,
Suite 1300, Portland, Oregon 97204, United States
| | - Chenyi Liao
- Department
of Chemistry, University of Vermont, Burlington, Vermont 05405, United States
| | - Jianing Li
- Department
of Chemistry, University of Vermont, Burlington, Vermont 05405, United States
| | - John C. Shelley
- Schrödinger,
Inc., 101 SW Main Street,
Suite 1300, Portland, Oregon 97204, United States
| |
Collapse
|
34
|
Yin L, Yuvienco C, Montclare JK. Protein based therapeutic delivery agents: Contemporary developments and challenges. Biomaterials 2017; 134:91-116. [PMID: 28458031 DOI: 10.1016/j.biomaterials.2017.04.036] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/18/2017] [Accepted: 04/21/2017] [Indexed: 12/15/2022]
Abstract
As unique biopolymers, proteins can be employed for therapeutic delivery. They bear important features such as bioavailability, biocompatibility, and biodegradability with low toxicity serving as a platform for delivery of various small molecule therapeutics, gene therapies, protein biologics and cells. Depending on size and characteristic of the therapeutic, a variety of natural and engineered proteins or peptides have been developed. This, coupled to recent advances in synthetic and chemical biology, has led to the creation of tailor-made protein materials for delivery. This review highlights strategies employing proteins to facilitate the delivery of therapeutic matter, addressing the challenges for small molecule, gene, protein and cell transport.
Collapse
Affiliation(s)
- Liming Yin
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY 11201, United States
| | - Carlo Yuvienco
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY 11201, United States
| | - Jin Kim Montclare
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY 11201, United States; Department of Chemistry, New York University, New York, NY 10003, United States; Department of Biomaterials, NYU College of Dentistry, New York, NY 10010, United States; Department of Biochemistry, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States.
| |
Collapse
|
35
|
Rayfield WJ, Kandula S, Khan H, Tugcu N. Impact of Freeze/Thaw Process on Drug Substance Storage of Therapeutics. J Pharm Sci 2017; 106:1944-1951. [PMID: 28343990 DOI: 10.1016/j.xphs.2017.03.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/19/2017] [Accepted: 03/13/2017] [Indexed: 10/19/2022]
Abstract
The storage of drug substance at subzero temperatures mitigates potential risks associated with liquid storage, such as degradation and shipping stress, making it the best solution for long-term storage. However, slower (generally uncontrolled) rates of freezing and thawing of drug substance in conventional large storage containers (>2L) can lead to greater cryoconcentration (exclusion of solute molecules) resulting in zones of higher protein and excipient concentrations and changes to the desired formulation pH and excipient concentration. These conditions can negatively impact product quality, thus changing the target product profile. Freeze/thaw studies can provide valuable knowledge on the molecule even when performed from an early formulation image. This study attempts to provide guidance and strategy for planning of drug substance freeze and thaw studies in early development using a scale-down model, evaluating the impact of the (1) freeze/thaw rate, (2) mode of freezing, (3) drug substance container, (4) drug substance concentration, and (5) formulation on the drug substance product quality. Data presented in this study showed no impact on drug substance product quality after undergoing the typical one freeze/thaw cycle process for the variables evaluated. These findings suggest that a qualified scale-down model is not required for early phases of process development and that existing small-scale models can be used for drug substance storage development studies. Based on our experience, a workflow is suggested with minimal experimental design to reduce the material requirement by >70% at early stages of product development to reduce constraints.
Collapse
Affiliation(s)
- William J Rayfield
- Process Development and Engineering, Biologics and Vaccines, MRL, Merck & Co., Inc., Kenilworth, New Jersey 07033.
| | - Sunitha Kandula
- Process Development and Engineering, Biologics and Vaccines, MRL, Merck & Co., Inc., Kenilworth, New Jersey 07033
| | - Heera Khan
- Mentrik Biotech, LLC, Two Lincoln Centre, Dallas, Texas 75240
| | - Nihal Tugcu
- Process Development and Engineering, Biologics and Vaccines, MRL, Merck & Co., Inc., Kenilworth, New Jersey 07033
| |
Collapse
|
36
|
Fox CB, Nemeth CL, Chevalier RW, Cantlon J, Bogdanoff DB, Hsiao JC, Desai TA. Picoliter-volume inkjet printing into planar microdevice reservoirs for low-waste, high-capacity drug loading. Bioeng Transl Med 2017; 2:9-16. [PMID: 28503662 PMCID: PMC5426811 DOI: 10.1002/btm2.10053] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Oral delivery of therapeutics is the preferred route for systemic drug administration due to ease of access and improved patient compliance. However, many therapeutics suffer from low oral bioavailability due to low pH and enzymatic conditions, poor cellular permeability, and low residence time. Microfabrication techniques have been used to create planar, asymmetric microdevices for oral drug delivery to address these limitations. The geometry of these microdevices facilitates prolonged drug exposure with unidirectional release of drug toward gastrointestinal epithelium. While these devices have significantly enhanced drug permeability in vitro and in vivo, loading drug into the micron-scale reservoirs of the devices in a low-waste, high-capacity manner remains challenging. Here, we use picoliter-volume inkjet printing to load topotecan and insulin into planar microdevices efficiently. Following a simple surface functionalization step, drug solution can be spotted into the microdevice reservoir. We show that relatively high capacities of both topotecan and insulin can be loaded into microdevices in a rapid, automated process with little to no drug waste.
Collapse
Affiliation(s)
- Cade B Fox
- Dept. of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158
| | - Cameron L Nemeth
- UC Berkeley and UCSF Graduate Program in Bioengineering, UCSF Mission Bay Campus, San Francisco, CA 94158
| | - Rachel W Chevalier
- Dept. of Pediatrics, Division of Pediatric Gastroenterology, School of Medicine, University of California, San Francisco, CA 94158
| | | | - Derek B Bogdanoff
- Center for Advanced Technology, University of California, San Francisco, CA, 94158
| | - Jeff C Hsiao
- Dept. of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158
| | - Tejal A Desai
- Dept. of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158.,UC Berkeley and UCSF Graduate Program in Bioengineering, UCSF Mission Bay Campus, San Francisco, CA 94158
| |
Collapse
|
37
|
Abstract
Macromolecules (proteins/peptides) have the potential for the development of new therapeutics. Due to their specific mechanism of action, macromolecules can be administered at relatively low doses compared with small-molecule drugs. Unfortunately, the therapeutic potential and clinical application of macromolecules is hampered by various obstacles including their large size, short in vivo half-life, phagocytic clearance, poor membrane permeability and structural instability. These challenges have encouraged researchers to develop novel strategies for effective delivery of macromolecules. In this review, various routes of macromolecule administration (invasive/noninvasive) are discussed. The advantages/limitations of novel delivery systems and the potential role of nanotechnology for the delivery of macromolecules are elaborated. In addition, fabrication approaches to make nanoformulations in different shapes and sizes are also summarized.
Collapse
|
38
|
Schoellhammer CM, Schroeder A, Maa R, Lauwers GY, Swiston A, Zervas M, Barman R, DiCiccio AM, Brugge WR, Anderson DG, Blankschtein D, Langer R, Traverso G. Ultrasound-mediated gastrointestinal drug delivery. Sci Transl Med 2016; 7:310ra168. [PMID: 26491078 DOI: 10.1126/scitranslmed.aaa5937] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
There is a significant clinical need for rapid and efficient delivery of drugs directly to the site of diseased tissues for the treatment of gastrointestinal (GI) pathologies, in particular, Crohn's and ulcerative colitis. However, complex therapeutic molecules cannot easily be delivered through the GI tract because of physiologic and structural barriers. We report the use of ultrasound as a modality for enhanced drug delivery to the GI tract, with an emphasis on rectal delivery. Ultrasound increased the absorption of model therapeutics inulin, hydrocortisone, and mesalamine two- to tenfold in ex vivo tissue, depending on location in the GI tract. In pigs, ultrasound induced transient cavitation with negligible heating, leading to an order of magnitude enhancement in the delivery of mesalamine, as well as successful systemic delivery of a macromolecule, insulin, with the expected hypoglycemic response. In a rodent model of chemically induced acute colitis, the addition of ultrasound to a daily mesalamine enema (compared to enema alone) resulted in superior clinical and histological scores of disease activity. In both animal models, ultrasound treatment was well tolerated and resulted in minimal tissue disruption, and in mice, there was no significant effect on histology, fecal score, or tissue inflammatory cytokine levels. The use of ultrasound to enhance GI drug delivery is safe in animals and could augment the efficacy of GI therapies and broaden the scope of agents that could be delivered locally and systemically through the GI tract for chronic conditions such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Carl M Schoellhammer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Avi Schroeder
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Faculty of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Ruby Maa
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gregory Yves Lauwers
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Albert Swiston
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael Zervas
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ross Barman
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Angela M DiCiccio
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - William R Brugge
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Daniel G Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel Blankschtein
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Giovanni Traverso
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
39
|
Rios S, Petersen MA, Arps JH. Tandem Delivery Platform for Small Molecules and Biologic Pharmaceuticals Via Silicone Foam1. J Med Device 2016. [DOI: 10.1115/1.4033218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Sasha Rios
- ProMed Pharma LLC, Minneapolis, MN 55447
| | | | | |
Collapse
|
40
|
Steinbach JM, Seo YE, Saltzman WM. Cell penetrating peptide-modified poly(lactic-co-glycolic acid) nanoparticles with enhanced cell internalization. Acta Biomater 2016; 30:49-61. [PMID: 26602822 PMCID: PMC4695306 DOI: 10.1016/j.actbio.2015.11.029] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 11/10/2015] [Accepted: 11/16/2015] [Indexed: 01/13/2023]
Abstract
The surface modification of nanoparticles (NPs) can enhance the intracellular delivery of drugs, proteins, and genetic agents. Here we studied the effect of different surface ligands, including cell penetrating peptides (CPPs), on the cell binding and internalization of poly(lactic-co-glycolic) (PLGA) NPs. Relative to unmodified NPs, we observed that surface-modified NPs greatly enhanced cell internalization. Using one CPP, MPG (unabbreviated notation), that achieved the highest degree of internalization at both low and high surface modification densities, we evaluated the effect of two different NP surface chemistries on cell internalization. After 2h, avidin-MPG NPs enhanced cellular internalization by 5 to 26-fold relative to DSPE-MPG NP formulations. Yet, despite a 5-fold increase in MPG density on DSPE compared to Avidin NPs, both formulations resulted in similar internalization levels (48 and 64-fold, respectively) after 24h. Regardless of surface modification, all NPs were internalized through an energy-dependent, clathrin-mediated process, and became dispersed throughout the cell. Overall both Avidin- and DSPE-CPP modified NPs significantly increased internalization and offer promising delivery options for applications in which internalization presents challenges to efficacious delivery.
Collapse
Affiliation(s)
- Jill M Steinbach
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA.
| | - Young-Eun Seo
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| |
Collapse
|