151
|
Hahn D, Sonntag JM, Lück S, Maitz MF, Freudenberg U, Jordan R, Werner C. Poly(2-alkyl-2-oxazoline)-Heparin Hydrogels-Expanding the Physicochemical Parameter Space of Biohybrid Materials. Adv Healthc Mater 2021; 10:e2101327. [PMID: 34541827 PMCID: PMC11481032 DOI: 10.1002/adhm.202101327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/10/2021] [Indexed: 12/19/2022]
Abstract
Poly(ethylene glycol) (PEG)-glycosaminoglycan (GAG) hydrogel networks are established as very versatile biomaterials. Herein, the synthetic gel component of the biohybrid materials is systematically varied by combining different poly(2-alkyl-2-oxazolines) (POx) with heparin applying a Michael-type addition crosslinking scheme: POx of gradated hydrophilicity and temperature-responsiveness provides polymer networks of distinctly different stiffness and swelling. Adjusting the mechanical properties and the GAG concentration of the gels to similar values allows for modulating the release of GAG-binding growth factors (VEGF165 and PDGF-BB) by the choice of the POx and its temperature-dependent conformation. Adsorption of fibronectin, growth of fibroblasts, and bacterial adhesion scale with the hydrophobicity of the gel-incorporated POx. In vitro hemocompatibility tests with freshly drawn human whole blood show advantages of POx-based gels compared to the PEG-based reference materials. Biohybrid POx hydrogels can therefore enable biomedical technologies requiring GAG-based materials with customized and switchable physicochemical characteristics.
Collapse
Affiliation(s)
- Dominik Hahn
- Leibniz Institute of Polymer Research DresdenMax‐Bergmann Center of Biomaterials DresdenHohe Str. 601069DresdenGermany
| | - Jannick M. Sonntag
- Dresden Initiative for Bioactive Interfaces & MaterialsTechnische Universität DresdenMommsenstr. 401069DresdenGermany
- Professur für Makromolekulare ChemieFaculty of Chemistry and Food ChemistryTechnische Universität DresdenMommsenstr. 401069DresdenGermany
| | - Steffen Lück
- Dresden Initiative for Bioactive Interfaces & MaterialsTechnische Universität DresdenMommsenstr. 401069DresdenGermany
- Professur für Makromolekulare ChemieFaculty of Chemistry and Food ChemistryTechnische Universität DresdenMommsenstr. 401069DresdenGermany
| | - Manfred F. Maitz
- Leibniz Institute of Polymer Research DresdenMax‐Bergmann Center of Biomaterials DresdenHohe Str. 601069DresdenGermany
| | - Uwe Freudenberg
- Leibniz Institute of Polymer Research DresdenMax‐Bergmann Center of Biomaterials DresdenHohe Str. 601069DresdenGermany
| | - Rainer Jordan
- Dresden Initiative for Bioactive Interfaces & MaterialsTechnische Universität DresdenMommsenstr. 401069DresdenGermany
- Professur für Makromolekulare ChemieFaculty of Chemistry and Food ChemistryTechnische Universität DresdenMommsenstr. 401069DresdenGermany
| | - Carsten Werner
- Leibniz Institute of Polymer Research DresdenMax‐Bergmann Center of Biomaterials DresdenHohe Str. 601069DresdenGermany
- Center for Regenerative Therapies Dresden (CRTD)Fetscherstr. 10501307DresdenGermany
| |
Collapse
|
152
|
Hwang D, Vinod N, Skoczen SL, Ramsey JD, Snapp KS, Montgomery SA, Wang M, Lim C, Frank JE, Sokolsky-Papkov M, Li Z, Yuan H, Stern ST, Kabanov AV. Bioequivalence assessment of high-capacity polymeric micelle nanoformulation of paclitaxel and Abraxane® in rodent and non-human primate models using a stable isotope tracer assay. Biomaterials 2021; 278:121140. [PMID: 34634661 PMCID: PMC10726948 DOI: 10.1016/j.biomaterials.2021.121140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 02/06/2023]
Abstract
The in vivo fate of nanoformulated drugs is governed by the physicochemical properties of the drug and the functionality of nanocarriers. Nanoformulations such as polymeric micelles, which physically encapsulate poorly soluble drugs, release their payload into the bloodstream during systemic circulation. This results in three distinct fractions of the drug-nanomedicine: encapsulated, protein-bound, and free drug. Having a thorough understanding of the pharmacokinetic (PK) profiles of each fraction is essential to elucidate mechanisms of nanomedicine-driven changes in drug exposure and PK/PD relationships pharmacodynamic activity. Here, we present a comprehensive preclinical assessment of the poly (2-oxazoline)-based polymeric micelle of paclitaxel (PTX) (POXOL hl-PM), including bioequivalence comparison to the clinically approved paclitaxel nanomedicine, Abraxane®. Physicochemical characterization and toxicity analysis of POXOL hl-PM was conducted using standardized protocols by the Nanotechnology Characterization Laboratory (NCL). The bioequivalence of POXOL hl-PM to Abraxane® was evaluated in rats and rhesus macaques using the NCL's established stable isotope tracer ultrafiltration assay (SITUA) to delineate the plasma PK of each PTX fraction. The SITUA study revealed that POXOL hl-PM and Abraxane® had comparable PK profiles not only for total PTX but also for the distinct drug fractions, suggesting bioequivalence in given animal models. The comprehensive preclinical evaluation of POXOL hl-PM in this study showcases a series of widely applicable standardized studies by NCL for assessing nanoformulations prior to clinical investigation.
Collapse
Affiliation(s)
- Duhyeong Hwang
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Natasha Vinod
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States; Joint UNC/NC State Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, 27599, United States
| | - Sarah L Skoczen
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, MD, 21702, United States
| | - Jacob D Ramsey
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Kelsie S Snapp
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, MD, 21702, United States
| | - Stephanie A Montgomery
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Mengzhe Wang
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, United States
| | - Chaemin Lim
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Jonathan E Frank
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, United States
| | - Marina Sokolsky-Papkov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Zibo Li
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, United States
| | - Hong Yuan
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, United States
| | - Stephan T Stern
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, MD, 21702, United States
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M. V. Lomonosov Moscow State University, Moscow, 119992, Russia.
| |
Collapse
|
153
|
Won C, Kwon C, Park K, Seo J, Lee T. Electronic Drugs: Spatial and Temporal Medical Treatment of Human Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005930. [PMID: 33938022 DOI: 10.1002/adma.202005930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/11/2020] [Indexed: 06/12/2023]
Abstract
Recent advances in diagnostics and medicines emphasize the spatial and temporal aspects of monitoring and treating diseases. However, conventional therapeutics, including oral administration and injection, have difficulties meeting these aspects due to physiological and technological limitations, such as long-term implantation and a narrow therapeutic window. As an innovative approach to overcome these limitations, electronic devices known as electronic drugs (e-drugs) have been developed to monitor real-time body signals and deliver specific treatments to targeted tissues or organs. For example, ingestible and patch-type e-drugs could detect changes in biomarkers at the target sites, including the gastrointestinal (GI) tract and the skin, and deliver therapeutics to enhance healing in a spatiotemporal manner. However, medical treatments often require invasive surgical procedures and implantation of medical equipment for either short or long-term use. Therefore, approaches that could minimize implantation-associated side effects, such as inflammation and scar tissue formation, while maintaining high functionality of e-drugs, are highly needed. Herein, the importance of the spatial and temporal aspects of medical treatment is thoroughly reviewed along with how e-drugs use cutting-edge technological innovations to deal with unresolved medical challenges. Furthermore, diverse uses of e-drugs in clinical applications and the future perspectives of e-drugs are discussed.
Collapse
Affiliation(s)
- Chihyeong Won
- Nanobio Device Laboratory, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Chaebeen Kwon
- Nanobio Device Laboratory, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kijun Park
- Biological Interfaces and Sensor Systems Laboratory, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jungmok Seo
- Biological Interfaces and Sensor Systems Laboratory, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Taeyoon Lee
- Nanobio Device Laboratory, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| |
Collapse
|
154
|
Krieghoff J, Gronbach M, Schulz-Siegmund M, Hacker MC. Biodegradable macromers for implant bulk and surface engineering. Biol Chem 2021; 402:1357-1374. [PMID: 34433237 DOI: 10.1515/hsz-2021-0161] [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: 02/23/2021] [Accepted: 08/09/2021] [Indexed: 11/15/2022]
Abstract
Macromers, polymeric molecules with at least two functional groups for cross-polymerization, are interesting materials to tailor mechanical, biochemical and degradative bulk and surface properties of implants for tissue regeneration. In this review we focus on macromers with at least one biodegradable building block. Manifold design options, such as choice of polymeric block(s), optional core molecule and reactive groups, as well as cross-co-polymerization with suitable anchor or linker molecules, allow the adaptation of macromer-based biomaterials towards specific application requirements in both hard and soft tissue regeneration. Implants can be manufactured from macromers using additive manufacturing as well as molding and templating approaches. This review summarizes and discusses the overall concept of biodegradable macromers and recent approaches for macromer processing into implants as well as techniques for surface modification directed towards bone regeneration. These aspects are reviewed including a focus on the authors' contributions to the field through research within the collaborative research project Transregio 67.
Collapse
Affiliation(s)
- Jan Krieghoff
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany
| | - Mathis Gronbach
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany
| | - Michaela Schulz-Siegmund
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany
| | - Michael C Hacker
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany.,Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University, Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| |
Collapse
|
155
|
Subasic CN, Ardana A, Chan LJ, Huang F, Scoble JA, Butcher NJ, Meagher L, Chiefari J, Kaminskas LM, Williams CC. Poly(HPMA-co-NIPAM) copolymer as an alternative to polyethylene glycol-based pharmacokinetic modulation of therapeutic proteins. Int J Pharm 2021; 608:121075. [PMID: 34481889 DOI: 10.1016/j.ijpharm.2021.121075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022]
Abstract
PEGylation is the standard approach for prolonging the plasma exposure of protein therapeutics but has limitations. We explored whether polymers prepared by Reversible Addition-Fragmentation chain-Transfer (RAFT) may provide better alternatives to polyethylene glycol (PEG). Four RAFT polymers were synthesised with varying compositions, molar mass (Mn), and structures, including a homopolymer of N-(2-hydroxypropyl)methacrylamide, (pHPMA) and statistical copolymers of HPMA with poly(ethylene glycol methyl ether acrylate) p(HPMA-co-PEGA); HPMA and N-acryloylmorpholine, p(HPMA-co-NAM); and HPMA and N-isopropylacrylamide, p(HPMA-co-NIPAM). The intravenous pharmacokinetics of the polymers were then evaluated in rats. The in vitro activity and in vivo pharmacokinetics of p(HPMA-co-NIPAM)-conjugated trastuzumab Fab' and full length mAb were then evaluated. p(HPMA-co-NIPAM) prolonged plasma exposure more avidly compared to the other p(HPMA) polymers or PEG, irrespective of molecular weight. When conjugated to trastuzumab-Fab', p(HPMA-co-NIPAM) prolonged plasma exposure of the Fab' similar to PEG-Fab'. The generation of anti-PEG IgM in rats 7 days after intravenous and subcutaneous dosing of p(HPMA-co-NIPAM) conjugated trastuzumab mAb was also examined and was shown to exhibit lower immunogenicity than the PEGylated construct. These data suggest that p(HPMA-co-NIPAM) has potential as a promising copolymer for use as an alternative conjugation strategy to PEG, to prolong the plasma exposure of therapeutic proteins.
Collapse
Affiliation(s)
- Christopher N Subasic
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Aditya Ardana
- CSIRO Manufacturing, 343 Royal Parade, Parkville, Victoria 3052, Australia
| | - Linda J Chan
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Fei Huang
- CSIRO Manufacturing, 343 Royal Parade, Parkville, Victoria 3052, Australia
| | - Judith A Scoble
- CSIRO Manufacturing, 343 Royal Parade, Parkville, Victoria 3052, Australia
| | - Neville J Butcher
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Laurence Meagher
- CSIRO Manufacturing, 343 Royal Parade, Parkville, Victoria 3052, Australia; Department of Materials Science and Engineering, Monash University, 20 Research Way, Clayton, Victoria 3168, Australia
| | - John Chiefari
- CSIRO Manufacturing, 343 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lisa M Kaminskas
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia; Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.
| | | |
Collapse
|
156
|
De D, Upadhyay P, Das A, Ghosh A, Adhikary A, Goswami MM. Studies on cancer cell death through delivery of dopamine as anti-cancer drug by a newly functionalized cobalt ferrite nano-carrier. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127202] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
157
|
Soni SS, Rodell CB. Polymeric materials for immune engineering: Molecular interaction to biomaterial design. Acta Biomater 2021; 133:139-152. [PMID: 33484909 DOI: 10.1016/j.actbio.2021.01.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 12/15/2022]
Abstract
Biomaterials continue to evolve as complex engineered tools for interactively instructing biological systems, aiding in the understanding and treatment of various disease states through intimate biological interaction. The immune response to polymeric materials is a critical area of study, as it governs the body's response to biomaterial implants, drug delivery vehicles, and even therapeutic drug formulations. Importantly, the development of the immune response to polymeric biomaterials spans length scales - from single molecular interactions to the complex sensing of bulk biophysical properties, all of which coordinate a tissue- and systems-level response. In this review, we specifically discuss a bottom-up approach to designing biomaterials that use molecular-scale interactions to drive immune response to polymers and discuss how these interactions can be leveraged for biomaterial design. STATEMENT OF SIGNIFICANCE: The immune system is an integral controller of (patho)physiological processes, affecting nearly all aspects of human health and disease. Polymeric biomaterials, whether biologically derived or synthetically produced, can potentially alter the behavior of immune cells due to their molecular-scale interaction with individual cells, as well as their interpretation at the bulk scale. This article reviews common mechanisms by which immune cells interact with polymers at the molecular level and discusses how these interactions are being leveraged to produce the next generation of biocompatible and immunomodulatory materials.
Collapse
|
158
|
McSweeney MD, Mohan M, Commins SP, Lai SK. Anaphylaxis to Pfizer/BioNTech mRNA COVID-19 Vaccine in a Patient With Clinically Confirmed PEG Allergy. FRONTIERS IN ALLERGY 2021; 2:715844. [PMID: 35387046 PMCID: PMC8974707 DOI: 10.3389/falgy.2021.715844] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Although allergic responses to the mRNA COVID-19 vaccines are rare, recent reports have suggested that a small number of individuals with allergy to polyethylene glycol (PEG), a component of the mRNA lipid nanoshell, may be at increased risk of anaphylaxis following vaccination. In this report, we describe a case of a patient who received an mRNA COVID-19 vaccine, experienced anaphylaxis, and was subsequently confirmed to have anti-PEG allergy by skin prick testing. The patient had previously noticed urticaria after handling PEG powder for their occupation and had a history of severe allergic response to multiple other allergens. Importantly, as many as 70% of people possess detectable levels of anti-PEG antibodies, indicating that the detection of such antibodies does not imply high risk for an anaphylactic response to vaccination. However, in people with pre-existing anti-PEG antibodies, the administration of PEGylated liposomes may induce higher levels of antibodies, which may cause accelerated clearance of other PEGylated therapeutics a patient may be receiving. It is important to improve awareness of PEG allergy among patients and clinicians.
Collapse
Affiliation(s)
| | - Manoj Mohan
- Okemos Allergy Center, Okemos, MI, United States
| | - Scott P. Commins
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of North Carolina—Chapel Hill, Chapel Hill, NC, United States
| | - Samuel K. Lai
- Mucommune, LLC, Durham, NC, United States
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina—Chapel Hill, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, University of North Carolina—Chapel Hill, Chapel Hill, NC, United States
- Department of Biomedical Engineering, University of North Carolina—Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
159
|
Chen BM, Cheng TL, Roffler SR. Polyethylene Glycol Immunogenicity: Theoretical, Clinical, and Practical Aspects of Anti-Polyethylene Glycol Antibodies. ACS NANO 2021; 15:14022-14048. [PMID: 34469112 DOI: 10.1021/acsnano.1c05922] [Citation(s) in RCA: 205] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Polyethylene glycol (PEG) is a flexible, hydrophilic simple polymer that is physically attached to peptides, proteins, nucleic acids, liposomes, and nanoparticles to reduce renal clearance, block antibody and protein binding sites, and enhance the half-life and efficacy of therapeutic molecules. Some naïve individuals have pre-existing antibodies that can bind to PEG, and some PEG-modified compounds induce additional antibodies against PEG, which can adversely impact drug efficacy and safety. Here we provide a framework to better understand PEG immunogenicity and how antibodies against PEG affect pegylated drug and nanoparticles. Analysis of published studies reveals rules for predicting accelerated blood clearance of pegylated medicine and therapeutic liposomes. Experimental studies of anti-PEG antibody binding to different forms, sizes, and immobilization states of PEG are also provided. The widespread use of SARS-CoV-2 RNA vaccines that incorporate PEG in lipid nanoparticles make understanding possible effects of anti-PEG antibodies on pegylated medicines even more critical.
Collapse
Affiliation(s)
- Bing-Mae Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Tian-Lu Cheng
- Center for Biomarkers and Biotech Drugs, Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
160
|
Kawassaki RK, Romano M, Dietrich N, Araki K. Titanium and Iron Oxide Nanoparticles for Cancer Therapy: Surface Chemistry and Biological Implications. FRONTIERS IN NANOTECHNOLOGY 2021. [DOI: 10.3389/fnano.2021.735434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Currently, cancer is among the most challenging diseases due to its ability to continuously evolve into a more complex muldimentional system, in addition to its high capability to spread to other organs and tissues. In this context, the relevance of nanobiomaterials (NBMs) for the development of new more effective and less harmful treatments is increasing. NBMs provide the possibility of combining several functionalities on a single system, expectedly in a synergic way, to better perform the treatment and cure. However, the control of properties such as colloidal stability, circulation time, pharmacokinetics, and biodistribution, assuring the concentration in specific target tissues and organs, while keeping all desired properties, tends to be dependent on subtle changes in surface chemistry. Hence, the behavior of such materials in different media/environments is of uttermost relevance and concern since it can compromise their efficiency and safety on application. Given the bright perspectives, many efforts have been focused on the development of nanomaterials fulfilling the requirements for real application. These include robust and reproducible preparation methods to avoid aggregation while preserving the interaction properties. The possible impact of nanomaterials in different forms of diagnosis and therapy has been demonstrated in the past few years, given the perspectives on how revolutionary they can be in medicine and health. Considering the high biocompatibility and suitability, this review is focused on titanium dioxide– and iron oxide–based nanoagents highlighting the current trends and main advancements in the research for cancer therapies. The effects of phenomena, such as aggregation and agglomeration, the formation of the corona layer, and how they can compromise relevant properties of nanomaterials and their potential applicability, are also addressed. In short, this review summarizes the current understanding and perspectives on such smart nanobiomaterials for diagnostics, treatment, and theranostics of diseases.
Collapse
|
161
|
Huang X, Zhang J, Song Y, Zhang T, Wang B. Combating liver cancer through GO-targeted biomaterials. Biomed Mater 2021; 16:065003. [PMID: 34412048 DOI: 10.1088/1748-605x/ac1f72] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 08/19/2021] [Indexed: 11/12/2022]
Abstract
Hydroxycamptothecin (HCPT) is a topoisomerase I inhibitor, and it has been widely used clinically in the treatment of primary liver cancer, gastric cancer, and other tumors. The clinical application of HCPT is limited by its water solubility, and it has certain toxicity to patients with tumor. Therefore, the effective tumor site accumulation of HCPT is necessary. This work studied the inhibitory effect of HCPT on the proliferation and migration of human liver cancer cells (HepG-2) and used carboxymethyl chitosan (CMC) and hyaluronic acid (HA) to modify graphene oxide (GO) as nano-carrier materials, which load HCPT to achieve a drug delivery system for liver tumors with good biocompatibility and high drug loading. HCPT can significantly inhibit proliferation and migration of HepG-2, enhance the release of reactive oxygen species, reduce mitochondrial membrane potential, and induce apoptosis. The GO-CMC-HA/HCPT drug delivery system enabled HepG-2 to uptake more HCPT, thereby inhibiting its proliferation and improving the efficacy of HCPTin vivoandin vitro. This study explored a potential therapy strategy by preparing a GO-based tumor-targeted drug delivery system.
Collapse
Affiliation(s)
- Xing Huang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Jiaxin Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yijie Song
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Tong Zhang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Bing Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| |
Collapse
|
162
|
Xu X, Liu C, Wang Y, Koivisto O, Zhou J, Shu Y, Zhang H. Nanotechnology-based delivery of CRISPR/Cas9 for cancer treatment. Adv Drug Deliv Rev 2021; 176:113891. [PMID: 34324887 DOI: 10.1016/j.addr.2021.113891] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats-associated protein 9) is a potent technology for gene-editing. Owing to its high specificity and efficiency, CRISPR/Cas9 is extensity used for human diseases treatment, especially for cancer, which involves multiple genetic alterations. Different concepts of cancer treatment by CRISPR/Cas9 are established. However, significant challenges remain for its clinical applications. The greatest challenge for CRISPR/Cas9 therapy is how to safely and efficiently deliver it to target sites in vivo. Nanotechnology has greatly contributed to cancer drug delivery. Here, we present the action mechanisms of CRISPR/Cas9, its application in cancer therapy and especially focus on the nanotechnology-based delivery of CRISPR/Cas9 for cancer gene editing and immunotherapy to pave the way for its clinical translation. We detail the difficult barriers for CRISIR/Cas9 delivery in vivo and discuss the relative solutions for encapsulation, target delivery, controlled release, cellular internalization, and endosomal escape.
Collapse
Affiliation(s)
- Xiaoyu Xu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200031, China; Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| | - Chang Liu
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| | - Yonghui Wang
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| | - Oliver Koivisto
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| | - Junnian Zhou
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland; Experimental Hematology and Biochemistry Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Yilai Shu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200031, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland.
| |
Collapse
|
163
|
Jiang Z, Feng X, Zou H, Xu W, Zhuang X. Poly(l-glutamic acid)-cisplatin nanoformulations with detachable PEGylation for prolonged circulation half-life and enhanced cell internalization. Bioact Mater 2021; 6:2688-2697. [PMID: 33665501 PMCID: PMC7895728 DOI: 10.1016/j.bioactmat.2021.01.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022] Open
Abstract
PEGylation has been widely applied to prolong the circulation times of nanomedicines via the steric shielding effect, which consequently improves the intratumoral accumulation. However, cell uptake of PEGylated nanoformulations is always blocked by the steric repulsion of PEG, which limits their therapeutic effect. To this end, we designed and prepared two kinds of poly(l-glutamic acid)-cisplatin (PLG-CDDP) nanoformulations with detachable PEG, which is responsive to specific tumor tissue microenvironments for prolonged circulation time and enhanced cell internalization. The extracellular pH (pHe)-responsive cleavage 2-propionic-3-methylmaleic anhydride (CDM)-derived amide bond and matrix metalloproteinases-2/9 (MMP-2/9)-sensitive degradable peptide PLGLAG were utilized to link PLG and PEG, yielding pHe-responsive PEG-pH e-PLG and MMP-sensitive PEG-MMP-PLG. The corresponding smart nanoformulations PEG-pH e-PLG-Pt and PEG-MMP-PLG-Pt were then prepared by the complexation of polypeptides and cisplatin (CDDP). The circulation half-lives of PEG-pH e-PLG-Pt and PEG-MMP-PLG-Pt were about 4.6 and 4.2 times higher than that of the control PLG-Pt, respectively. Upon reaching tumor tissue, PEG on the surface of nanomedicines was detached as triggered by pHe or MMP, which increased intratumoral CDDP retention, enhanced cell uptake, and improved antitumor efficacy toward a fatal high-grade serous ovarian cancer (HGSOC) mouse model, indicating the promising prospects for clinical application of detachable PEGylated nanoformulations.
Collapse
Affiliation(s)
- Zhongyu Jiang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, P. R. China
| | - Xiangru Feng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Haoyang Zou
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Weiguo Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Xiuli Zhuang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, P. R. China
| |
Collapse
|
164
|
The evolution of commercial drug delivery technologies. Nat Biomed Eng 2021; 5:951-967. [PMID: 33795852 DOI: 10.1038/s41551-021-00698-w] [Citation(s) in RCA: 498] [Impact Index Per Article: 166.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Drug delivery technologies have enabled the development of many pharmaceutical products that improve patient health by enhancing the delivery of a therapeutic to its target site, minimizing off-target accumulation and facilitating patient compliance. As therapeutic modalities expanded beyond small molecules to include nucleic acids, peptides, proteins and antibodies, drug delivery technologies were adapted to address the challenges that emerged. In this Review Article, we discuss seminal approaches that led to the development of successful therapeutic products involving small molecules and macromolecules, identify three drug delivery paradigms that form the basis of contemporary drug delivery and discuss how they have aided the initial clinical successes of each class of therapeutic. We also outline how the paradigms will contribute to the delivery of live-cell therapies.
Collapse
|
165
|
Elalouf A. Immune response against the biomaterials used in 3D bioprinting of organs. Transpl Immunol 2021; 69:101446. [PMID: 34389430 DOI: 10.1016/j.trim.2021.101446] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/26/2022]
Abstract
Regenerative medicine has developed promising approaches for healing and replacing defective and damaged organs or tissues with functional ones. Three-dimensional (3D) bioprinting innovation has integrated a potential to design organs or tissues specific to the patient with the capability of rapid construction to fulfill the storage of organs and the need for transplantation. 3D bioprinting of organs has the main goal to develop a structural and functional organ or tissue mimic to the original one. The highly complex fabrication of tissue engineering scaffolds containing biomaterials, tissue models, and biomedical devices has made it possible to print small blood vessels to mimic organs to reduce organ or tissue rejection. 3D bioprinting has the concept of bioinks containing biomaterials that may trigger the immune responses in the body. Nevertheless, foreign body response (FBR) is mediated by various cell types such as B-cells, dendritic cells, macrophages, natural killer cells, neutrophils, and T-cells, and molecular signals such as antibodies (Abs), cytokines, and reactive radical species. Typically, the biomaterial is shielded by the fibrous encapsulation that is regulated by molecular signals. This review explored the progress in 3D bioprinting of vital organs and basic immune response against the biomaterials used in this approach. Thus, evaluating immune response against biomaterials used in 3D printed organs is necessary to mitigate tissue rejection after the transplantation.
Collapse
Affiliation(s)
- Amir Elalouf
- Bar-Ilan University, Department of Management, Ramat Gan 5290002, Israel.
| |
Collapse
|
166
|
Pires IS, Hammond PT, Irvine DJ. Engineering Strategies for Immunomodulatory Cytokine Therapies - Challenges and Clinical Progress. ADVANCED THERAPEUTICS 2021; 4:2100035. [PMID: 34734110 PMCID: PMC8562465 DOI: 10.1002/adtp.202100035] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Indexed: 12/15/2022]
Abstract
Cytokines are immunoregulatory proteins involved in many pathological states with promising potential as therapeutic agents. A diverse array of cytokines have been studied in preclinical disease models since the 1950s, some of which became successful biopharmaceutical products with the advancement of recombinant protein technology in the 1980s. However, following these early approvals, clinical translation of these natural immune signaling molecules has been limited due to their pleiotropic action in many cell types, and the fact that they have evolved to act primarily locally in tissues. These characteristics, combined with poor pharmacokinetics, have hindered the delivery of cytokines via systemic administration routes due to dose-limiting toxicities. However, given their clinical potential and recent clinical successes in cancer immunotherapy, cytokines continue to be extensively pursued in preclinical and clinical studies, and a range of molecular and formulation engineering strategies are being applied to reduce treatment toxicity while maintaining or enhancing therapeutic efficacy. This review provides a brief background on the characteristics of cytokines and their history as clinical therapeutics, followed by a deeper discussion on the engineering strategies developed for cytokine therapies with a focus on the translational relevance of these approaches.
Collapse
Affiliation(s)
- Ivan S Pires
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
167
|
Peters LJF, Jans A, Bartneck M, van der Vorst EPC. Immunomodulatory Nanomedicine for the Treatment of Atherosclerosis. J Clin Med 2021; 10:3185. [PMID: 34300351 PMCID: PMC8306310 DOI: 10.3390/jcm10143185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/09/2021] [Accepted: 07/16/2021] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis is the main underlying cause of cardiovascular diseases (CVDs), which remain the number one contributor to mortality worldwide. Although current therapies can slow down disease progression, no treatment is available that can fully cure or reverse atherosclerosis. Nanomedicine, which is the application of nanotechnology in medicine, is an emerging field in the treatment of many pathologies, including CVDs. It enables the production of drugs that interact with cellular receptors, and allows for controlling cellular processes after entering these cells. Nanomedicine aims to repair, control and monitor biological and physiological systems via nanoparticles (NPs), which have been shown to be efficient drug carriers. In this review we will, after a general introduction, highlight the advantages and limitations of the use of such nano-based medicine, the potential applications and targeting strategies via NPs. For example, we will provide a detailed discussion on NPs that can target relevant cellular receptors, such as integrins, or cellular processes related to atherogenesis, such as vascular smooth muscle cell proliferation. Furthermore, we will underline the (ongoing) clinical trials focusing on NPs in CVDs, which might bring new insights into this research field.
Collapse
Affiliation(s)
- Linsey J. F. Peters
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany;
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Alexander Jans
- Department of Medicine III, University Hospital Aachen, 52074 Aachen, Germany; (A.J.); (M.B.)
| | - Matthias Bartneck
- Department of Medicine III, University Hospital Aachen, 52074 Aachen, Germany; (A.J.); (M.B.)
| | - Emiel P. C. van der Vorst
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany;
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
168
|
Tieu T, Wei Y, Cifuentes‐Rius A, Voelcker NH. Overcoming Barriers: Clinical Translation of siRNA Nanomedicines. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100108] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Terence Tieu
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton VIC 3168 Australia
| | - Yingkai Wei
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
| | - Anna Cifuentes‐Rius
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
| | - Nicolas H. Voelcker
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton VIC 3168 Australia
- Melbourne Centre for Nanofabrication 151 Wellington Road Victorian Node of the Australian National Fabrication Facility Clayton VIC 3168 Australia
| |
Collapse
|
169
|
Li H, Wang Y, Tang Q, Yin D, Tang C, He E, Zou L, Peng Q. The protein corona and its effects on nanoparticle-based drug delivery systems. Acta Biomater 2021; 129:57-72. [PMID: 34048973 DOI: 10.1016/j.actbio.2021.05.019] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/25/2021] [Accepted: 05/18/2021] [Indexed: 02/04/2023]
Abstract
In most cases, once nanoparticles (NPs) enter the blood, their surface is covered by biological molecules, especially proteins, forming a so-called protein corona (PC). As a result, what the cells of the body "see" is not the NPs as formulated by the chemists, but the PC. In this way, the PC can influence the effects of the NPs and even mask the desired effects of the NP components. While this can argue for trying to inhibit protein-nanomaterial interactions, encapsulating NPs in an endogenous PC may increase their clinical usefulness. In this review, we briefly introduce the concept of the PC, its formation and its effects on the behavior of NPs. We also discuss how to reduce the formation of PCs or exploit them to enhance NP functions. Studying the interactions between proteins and NPs will provide insights into their clinical activity in health and disease. STATEMENT OF SIGNIFICANCE: The formation of protein corona (PC) will affect the operation of nanoparticles (NPs) in vivo. Since there are many proteins in the blood, it is impossible to completely overcome the formation of PC. Therefore, the use of PCs to deliver drug is the best choice. De-opsonins adsorbed on NPs can reduce macrophage phagocytosis and cytotoxicity of NPs, and prolong their circulation in blood. Albumin, apolipoprotein and transferrin are typical de-opsonins. In present review, we mainly discuss how to optimize the delivery of nanoparticles through the formation of albumin corona, transferrin corona and apolipoprotein corona in vivo or in vitro.
Collapse
Affiliation(s)
- Hanmei Li
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China
| | - Yao Wang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Qi Tang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Dan Yin
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Chuane Tang
- School of Mechanical Engineering, Chengdu university, Chengdu 610106, China
| | - En He
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
170
|
Nishio A, Bolte FJ, Takeda K, Park N, Yu ZX, Park H, Valdez K, Ghany MG, Rehermann B. Clearance of pegylated interferon by Kupffer cells limits NK cell activation and therapy response of patients with HBV infection. Sci Transl Med 2021; 13:13/587/eaba6322. [PMID: 33790025 DOI: 10.1126/scitranslmed.aba6322] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022]
Abstract
Pegylated interferon-α (PEG-IFN-α), where IFN-α is attached to polyethylene glycol (PEG), is an approved treatment for chronic hepatitis B virus (HBV) infection, a disease that causes liver-related morbidity and mortality in 257 million people worldwide. It is unknown why only a minority of patients respond to PEG-IFN-α. Using sequential blood samples and liver biopsies of patients with chronic HBV infection before, during, and after PEG-IFN-α treatment, we find that patients with early natural killer (NK) cell activation after PEG-IFN-α injection experienced greater liver inflammation, lysis of HBV-infected hepatocytes, and hepatitis B surface antigen (HBsAg) decline than those without. NK cell activation was associated with induction of interferon-stimulated genes and determined by PEG-IFN-α pharmacokinetics. Patients with delayed increases in PEG-IFN-α concentrations had greater amounts of PEG-specific immunoglobulin M (IgM) immune complexes in the blood and more PEG and IgM detected in the liver than patients with rapid increase in PEG-IFN-α concentration. This was associated with reduced NK cell activation. These results indicate that the immunomodulatory functions of PEG-IFN-α, particularly activation of NK cells, play a pivotal role in the response to treatment and further demonstrate that these functions are affected by PEG-IFN-α pharmacokinetics. Accelerated clearance of antibody-complexed pegylated drugs by Kupffer cells may be important beyond the field of HBV therapeutics. Thus, these findings may contribute to improving the efficacy of pegylated drugs that are now being developed for other chronic diseases and cancer.
Collapse
Affiliation(s)
- Akira Nishio
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Fabian J Bolte
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Kazuyo Takeda
- Pathology Core, National Heart, Lung and Blood Institute, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Nana Park
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Zu-Xi Yu
- Pathology Core, National Heart, Lung and Blood Institute, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Heiyoung Park
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Kristin Valdez
- Clinical Research Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Marc G Ghany
- Clinical Research Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA.
| |
Collapse
|
171
|
Xiong W, Lan Q, Liang X, Zhao J, Huang H, Zhan Y, Qin Z, Jiang X, Zheng L. Cartilage-targeting poly(ethylene glycol) (PEG)-formononetin (FMN) nanodrug for the treatment of osteoarthritis. J Nanobiotechnology 2021; 19:197. [PMID: 34217311 PMCID: PMC8254262 DOI: 10.1186/s12951-021-00945-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 06/24/2021] [Indexed: 11/10/2022] Open
Abstract
Intra-articular (IA) injection is an efficient treatment for osteoarthritis, which will minimize systemic side effects. However, the joint experiences rapid clearance of therapeutics after intra-articular injection. Delivering system modified through active targeting strategies to facilitate localization within specific joint tissues such as cartilage is hopeful to increase the therapeutic effects. In this study, we designed a nanoscaled amphiphilic and cartilage-targeting polymer-drug delivery system by using formononetin (FMN)-poly(ethylene glycol) (PEG) (denoted as PCFMN), which was prepared by PEGylation of FMN followed by coupling with cartilage-targeting peptide (CollBP). Our results showed that PCFMN was approximately regular spherical with an average diameter about 218 nm. The in vitro test using IL-1β stimulated chondrocytes indicated that PCFMN was biocompatible and upregulated anabolic genes while simultaneously downregulated catabolic genes of the articular cartilage. The therapeutic effects in vivo indicated that PCFMN could effectively attenuate the progression of OA as evidenced by immunohistochemical staining and histological analysis. In addition, PCFMN showed higher intention time in joints and better anti-inflammatory effects than FMN, indicating the efficacy of cartilage targeting nanodrug on OA. This study may provide a reference for clinical OA therapy.
Collapse
Affiliation(s)
- Wei Xiong
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Qiumei Lan
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Xiaonan Liang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Hanji Huang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Yanting Zhan
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Zainen Qin
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Xianfang Jiang
- Department of Oral Radiology, Guangxi Medical University College of Stomatology, Nanning, 530021, China.
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
172
|
Boztepe T, Castro GR, León IE. Lipid, polymeric, inorganic-based drug delivery applications for platinum-based anticancer drugs. Int J Pharm 2021; 605:120788. [PMID: 34116182 DOI: 10.1016/j.ijpharm.2021.120788] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 12/13/2022]
Abstract
The three main FDA-approved platinum drugs in chemotherapy such as carboplatin, cisplatin, and oxaliplatin are extensively applied in cancer treatments. Although the clinical applications of platinum-based drugs are extremely effective, their toxicity profile restricts their extensive application. Therefore, recent studies focus on developing new platinum drug formulations, expanding the therapeutic aspect. In this sense, recent advances in the development of novel drug delivery carriers will help with the increase of drug stability and biodisponibility, concomitantly with the reduction of drug efflux and undesirable secondary toxic effects of platinum compounds. The present review describes the state of the art of platinum drugs with their biological effects, pre- and clinical studies, and novel drug delivery nanodevices based on lipids, polymers, and inorganic.
Collapse
Affiliation(s)
- Tugce Boztepe
- Laboratorio de Nanobiomateriales, CINDEFI - Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata-CONICET (CCT La Plata), Calle 47 y 115, B1900AJL La Plata, Argentina
| | - Guillermo R Castro
- Laboratorio de Nanobiomateriales, CINDEFI - Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata-CONICET (CCT La Plata), Calle 47 y 115, B1900AJL La Plata, Argentina; Max Planck Laboratory for Structural Biology, Chemistry and Molecular Biophysics of Rosario (MPLbioR, UNR-MPIbpC), Partner Laboratory of the Max Planck Institute for Biophysical Chemistry (MPIbpC, MPG), Centro de Estudios Interdisciplinarios (CEI), Universidad Nacional de Rosario, Maipú 1065, S2000 Rosario, Santa Fe, Argentina.
| | - Ignacio E León
- Centro de Química Inorgánica, CEQUINOR (CONICET-UNLP), Bv. 120 1465, La Plata, Argentina.
| |
Collapse
|
173
|
Chen L, Hong W, Ren W, Xu T, Qian Z, He Z. Recent progress in targeted delivery vectors based on biomimetic nanoparticles. Signal Transduct Target Ther 2021; 6:225. [PMID: 34099630 PMCID: PMC8182741 DOI: 10.1038/s41392-021-00631-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 02/05/2023] Open
Abstract
Over the past decades, great interest has been given to biomimetic nanoparticles (BNPs) since the rise of targeted drug delivery systems and biomimetic nanotechnology. Biological vectors including cell membranes, extracellular vesicles (EVs), and viruses are considered promising candidates for targeted delivery owing to their biocompatibility and biodegradability. BNPs, the integration of biological vectors and functional agents, are anticipated to load cargos or camouflage synthetic nanoparticles to achieve targeted delivery. Despite their excellent intrinsic properties, natural vectors are deliberately modified to endow multiple functions such as good permeability, improved loading capability, and high specificity. Through structural modification and transformation of the vectors, they are pervasively utilized as more effective vehicles that can deliver contrast agents, chemotherapy drugs, nucleic acids, and genes to target sites for refractory disease therapy. This review summarizes recent advances in targeted delivery vectors based on cell membranes, EVs, and viruses, highlighting the potential applications of BNPs in the fields of biomedical imaging and therapy industry, as well as discussing the possibility of clinical translation and exploitation trend of these BNPs.
Collapse
Affiliation(s)
- Li Chen
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weiqi Hong
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenyan Ren
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyong Qian
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
174
|
Risma KA, Edwards KM, Hummell DS, Little FF, Norton AE, Stallings A, Wood RA, Milner JD. Potential mechanisms of anaphylaxis to COVID-19 mRNA vaccines. J Allergy Clin Immunol 2021; 147:2075-2082.e2. [PMID: 33857566 PMCID: PMC8056854 DOI: 10.1016/j.jaci.2021.04.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Anaphylaxis to vaccines is historically a rare event. The coronavirus disease 2019 pandemic drove the need for rapid vaccine production applying a novel antigen delivery system: messenger RNA vaccines packaged in lipid nanoparticles. Unexpectedly, public vaccine administration led to a small number of severe allergic reactions, with resultant substantial public concern, especially within atopic individuals. We reviewed the constituents of the messenger RNA lipid nanoparticle vaccine and considered several contributors to these reactions: (1) contact system activation by nucleic acid, (2) complement recognition of the vaccine-activating allergic effector cells, (3) preexisting antibody recognition of polyethylene glycol, a lipid nanoparticle surface hydrophilic polymer, and (4) direct mast cell activation, coupled with potential genetic or environmental predispositions to hypersensitivity. Unfortunately, measurement of anti-polyethylene glycol antibodies in vitro is not clinically available, and the predictive value of skin testing to polyethylene glycol components as a coronavirus disease 2019 messenger RNA vaccine-specific anaphylaxis marker is unknown. Even less is known regarding the applicability of vaccine use for testing (in vitro/vivo) to ascertain pathogenesis or predict reactivity risk. Expedient and thorough research-based evaluation of patients who have suffered anaphylactic vaccine reactions and prospective clinical trials in putative at-risk individuals are needed to address these concerns during a public health crisis.
Collapse
Affiliation(s)
- Kimberly A Risma
- Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Kathryn M Edwards
- Division of Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tenn
| | - Donna S Hummell
- Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Frederic F Little
- Division of Pulmonary, Allergy, Sleep and Critical Care Medicine, Department of Pediatrics, Boston University School of Medicine, Boston, Mass
| | - Allison E Norton
- Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Amy Stallings
- Division of Pediatric Allergy and Immunology, Duke University Medical Center, Durham, NC
| | - Robert A Wood
- Division of Pediatric Allergy and Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
175
|
Zhang Y, He P, Zhang P, Yi X, Xiao C, Chen X. Polypeptides-Drug Conjugates for Anticancer Therapy. Adv Healthc Mater 2021; 10:e2001974. [PMID: 33929786 DOI: 10.1002/adhm.202001974] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/30/2021] [Indexed: 12/15/2022]
Abstract
Polypeptides are an important class of biodegradable polymers that have been widely used in drug delivery field. Owing to the controllable synthesis and robust side chain-functionalization ability, polypeptides have long been ideal candidates for conjugation with anticancer drugs. The chemical conjugation of anticancer drugs with polypeptides, termed polypeptides-drug conjugates, has demonstrated several advantages in improving pharmacokinetics, enhancing drug targeting, and controlling drug release, thereby leading to enhanced therapeutic outcomes with reduced side toxicities. This review focuses on the recent advances in the design and preparation of polypeptides-drug conjugates for enhanced anticancer therapy. Strategies for conjugation of different types of drugs, including small-molecule chemotherapeutic drugs, proteins, vascular disrupting agents, and gas molecules, onto polypeptides backbone are summarized. Finally, the challenges and future perspectives on the development of innovative polypeptides-drug conjugates for clinical cancer treatment are also presented.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Pan He
- School of Materials Science and Engineering Changchun University of Science and Technology Changchun 130022 P. R. China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Xuan Yi
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| |
Collapse
|
176
|
Islam Y, Leach AG, Smith J, Pluchino S, Coxon CR, Sivakumaran M, Downing J, Fatokun AA, Teixidò M, Ehtezazi T. Physiological and Pathological Factors Affecting Drug Delivery to the Brain by Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2002085. [PMID: 34105297 PMCID: PMC8188209 DOI: 10.1002/advs.202002085] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/06/2021] [Indexed: 05/04/2023]
Abstract
The prevalence of neurological/neurodegenerative diseases, such as Alzheimer's disease is known to be increasing due to an aging population and is anticipated to further grow in the decades ahead. The treatment of brain diseases is challenging partly due to the inaccessibility of therapeutic agents to the brain. An increasingly important observation is that the physiology of the brain alters during many brain diseases, and aging adds even more to the complexity of the disease. There is a notion that the permeability of the blood-brain barrier (BBB) increases with aging or disease, however, the body has a defense mechanism that still retains the separation of the brain from harmful chemicals in the blood. This makes drug delivery to the diseased brain, even more challenging and complex task. Here, the physiological changes to the diseased brain and aged brain are covered in the context of drug delivery to the brain using nanoparticles. Also, recent and novel approaches are discussed for the delivery of therapeutic agents to the diseased brain using nanoparticle based or magnetic resonance imaging guided systems. Furthermore, the complement activation, toxicity, and immunogenicity of brain targeting nanoparticles as well as novel in vitro BBB models are discussed.
Collapse
Affiliation(s)
- Yamir Islam
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Andrew G. Leach
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- Division of Pharmacy and OptometryThe University of ManchesterStopford Building, Oxford RoadManchesterM13 9PTUK
| | - Jayden Smith
- Cambridge Innovation Technologies Consulting (CITC) LimitedSt. John's Innovation CentreCowley RoadCambridgeCB4 0WSUK
| | - Stefano Pluchino
- Department of Clinical NeurosciencesClifford Allbutt Building – Cambridge Biosciences Campus and NIHR Biomedical Research CentreUniversity of CambridgeHills RoadCambridgeCB2 0HAUK
| | - Christopher R. Coxon
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- School of Engineering and Physical SciencesHeriot‐Watt UniversityWilliam Perkin BuildingEdinburghEH14 4ASUK
| | - Muttuswamy Sivakumaran
- Department of HaematologyPeterborough City HospitalEdith Cavell CampusBretton Gate PeterboroughPeterboroughPE3 9GZUK
| | - James Downing
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Amos A. Fatokun
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Meritxell Teixidò
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 10Barcelona08028Spain
| | - Touraj Ehtezazi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| |
Collapse
|
177
|
Bao J, Zhang Q, Duan T, Hu R, Tang J. The Fate of Nanoparticles In Vivo and the Strategy of Designing Stealth Nanoparticle for Drug Delivery. Curr Drug Targets 2021; 22:922-946. [PMID: 33461465 DOI: 10.2174/1389450122666210118105122] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 11/22/2022]
Abstract
Nano-drug delivery systems (Nano-DDS) offer powerful advantages in drug delivery and targeted therapy for diseases. Compared to the traditional drug formulations, Nano-DDS can increase solubility, biocompatibility, and reduce off-targeted side effects of free drugs. However, they still have some disadvantages that pose a limitation in reaching their full potential in clinical use. Protein adsorption in blood, activation of the complement system, and subsequent sequestration by the mononuclear phagocyte system (MPS) consequently result in nanoparticles (NPs) to be rapidly cleared from circulation. Therefore, NPs have low drug delivery efficiency. So, it is important to develop stealth NPs for reducing bio-nano interaction. In this review, we first conclude the interaction between NPs and biological environments, such as blood proteins and MPS, and factors influencing each other. Next, we will summarize the new strategies to reduce NPs protein adsorption and uptake by the MPS based on current knowledge of the bio-nano interaction. Further directions will also be highlighted for the development of biomimetic stealth nano-delivery systems by combining targeted strategies for a better therapeutic effect.
Collapse
Affiliation(s)
- Jianwei Bao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qianqian Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Tijie Duan
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Rongfeng Hu
- key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Anhui "115" Xin'an Medicine Research & Development Innovation Team, Anhui Academy of Chinese Medicine, Hefei 230038, China
| | - Jihui Tang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
178
|
Ghasemi S, Dashti M. Fight against COVID-19 with mRNA vaccines and interaction with Dermal fillers. Clin Exp Vaccine Res 2021; 10:151-153. [PMID: 34222128 PMCID: PMC8217583 DOI: 10.7774/cevr.2021.10.2.151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 11/17/2022] Open
Abstract
The current coronavirus disease 2019 (COVID-19) outbreak has created a huge demand for rapid and high-volume vaccine manufacturing. Several new platform technologies and traditional manufacturing approaches are being used to meet this demand. Among them, the mRNA vaccine platform technology holds great promise for obtaining an emergency use authorization to facilitate immunization against SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), which causes the respiratory illness COVID-19. However, this is a new technology, which means that there is significant uncertainty about possible production at a large scale and speed. Vaccines of mRNA work by providing our cells with the genetic code to make viral proteins. Once proteins that do not cause any disease are produced, the body triggers an immune response against the virus, allowing them to develop immunity. mRNA can be used to make any protein theoretically. Still, it is much easier to produce than the proteins themselves or the inactivated and attenuated versions of viruses commonly used in vaccines, which makes this technique attractive, says mRNA specialist Norbert Pardi, at the University of Pennsylvania.
Collapse
Affiliation(s)
- Shohreh Ghasemi
- Department of Oral and Maxillofacial Surgery, University of Augusta, Marietta, GA, USA
| | - Mahmood Dashti
- DDS, Dental School, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
179
|
Tretiakova D, Svirshchevskaya E, Onishchenko N, Alekseeva A, Boldyrev I, Kamyshinsky R, Natykan A, Lokhmotov A, Arantseva D, Shobolov D, Vodovozova E. Liposomal Formulation of a Melphalan Lipophilic Prodrug: Studies of Acute Toxicity, Tolerability, and Antitumor Efficacy. Curr Drug Deliv 2021; 17:312-323. [PMID: 32056524 DOI: 10.2174/1567201817666200214105357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/13/2019] [Accepted: 02/02/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Recently we developed a scalable scheme of synthesis of melphalan ester conjugate with 1,2-dioleoyl-sn-glycerol (MlphDG) and a protocol for the fabrication of its lyophilized liposomal formulation. OBJECTIVE Herein we compared this new convenient in use formulation of MlphDG with parent drug Alkeran® in rats concerning several toxicological parameters and evaluated its antitumor efficacy in the model of breast cancer in mice. METHOD Liposomes of approximately 100 nm in diameter, consisting of egg phosphatidylcholine, soybean phosphatidylinositol, and MlphDG, or placebo liposomes without the drug were produced by extrusion and lyophilized. Alkeran® or liposomes recovered by the addition of water were injected into the tail vein of animals. Clinical examination of rats consisted of detailed inspection of the behavior, general status, and hematological parameters. Mice with transplanted breast cancer WNT-1 were subjected to multiple treatments with the drugs; tumor growth inhibition was assessed, together with cellular immunity parameters. RESULTS Liposomes showed approximately two times lower acute toxicity and better tolerability than Alkeran® in terms of behavioral criteria. The toxic effects of liposomes on hemopoiesis were manifested at higher doses than in the case of Alkeran®, proportionally to the difference in LD50 values. The formulation inhibited tumor growth significantly more effectively than Alkeran®, delaying the start of the exponential growth phase and exhibiting no additional toxic effects toward bone marrow. CONCLUSION Lower toxicity of the liposomal formulation of MlphDG promises improved quality of life for cancer patients in need of treatment with melphalan. Presumably, the list of indications for melphalan therapy could be extended.
Collapse
Affiliation(s)
- Daria Tretiakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Elena Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Natalia Onishchenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Anna Alekseeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Ivan Boldyrev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Roman Kamyshinsky
- National Research Center "Kurchatov Institute", Moscow, Russian Federation
| | - Alexey Natykan
- Drugs Technology Ltd., Khimki, Мoscow Region, Russian Federation
| | - Anton Lokhmotov
- Drugs Technology Ltd., Khimki, Мoscow Region, Russian Federation
| | - Diana Arantseva
- Drugs Technology Ltd., Khimki, Мoscow Region, Russian Federation
| | - Dmitry Shobolov
- Drugs Technology Ltd., Khimki, Мoscow Region, Russian Federation
| | - Elena Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| |
Collapse
|
180
|
Raoufi E, Bahramimeimandi B, Salehi-Shadkami M, Chaosri P, Mozafari MR. Methodical Design of Viral Vaccines Based on Avant-Garde Nanocarriers: A Multi-Domain Narrative Review. Biomedicines 2021; 9:520. [PMID: 34066608 PMCID: PMC8148582 DOI: 10.3390/biomedicines9050520] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/27/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
The current health crisis caused by coronavirus 2019 (COVID-19) and associated pathogens emphasize the urgent need for vaccine systems that can generate protective and long-lasting immune responses. Vaccination, employing peptides, nucleic acids, and other molecules, or using pathogen-based strategies, in fact, is one of the most potent approaches in the management of viral diseases. However, the vaccine candidate requires protection from degradation and precise delivery to the target cells. This can be achieved by employing different types of drug and vaccine delivery strategies, among which, nanotechnology-based systems seem to be more promising. This entry aims to provide insight into major aspects of vaccine design and formulation to address different diseases, including the recent outbreak of SARS-CoV-2. Special emphasis of this review is on the technical and practical aspects of vaccine construction and theranostic approaches to precisely target and localize the active compounds.
Collapse
Affiliation(s)
- Ehsan Raoufi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran; (E.R.); (B.B.)
| | - Bahar Bahramimeimandi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran; (E.R.); (B.B.)
| | - M. Salehi-Shadkami
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran;
| | - Patcharida Chaosri
- Supreme NanoBiotics Co. Ltd. and Supreme Pharmatech Co. Ltd., 399/90-95 Moo 13 Kingkaew Rd. Soi 25/1, T. Rachateva, A. Bangplee, Samutprakan 10540, Thailand;
| | - M. R. Mozafari
- Supreme NanoBiotics Co. Ltd. and Supreme Pharmatech Co. Ltd., 399/90-95 Moo 13 Kingkaew Rd. Soi 25/1, T. Rachateva, A. Bangplee, Samutprakan 10540, Thailand;
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
181
|
Emam SE, Elsadek NE, Abu Lila AS, Takata H, Kawaguchi Y, Shimizu T, Ando H, Ishima Y, Ishida T. Anti-PEG IgM production and accelerated blood clearance phenomenon after the administration of PEGylated exosomes in mice. J Control Release 2021; 334:327-334. [PMID: 33957196 DOI: 10.1016/j.jconrel.2021.05.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 12/18/2022]
Abstract
Recently, there is an increasing interest in exosomes or extracellular vesicles as potential candidates for delivering RNAs, proteins, genes, and anticancer agents. Engineering of exosome properties is rapidly evolving as a means of expanding exosome applications. PEGylation of exosomes is a technique used to improve their in vivo stability, circulation half-lives, and sometimes to allow the binding targeting ligands to the exosome exterior. According to FDA guidelines for the development of PEGylated proteins, immunological responses to PEGylated molecules and particles should be examined. In this study, we prepared PEGylated exosomes and investigated the production of anti-PEG IgM antibodies after single i.v. injections in mice. In addition, we monitored blood concentrations and tumor accumulation of a second dose of PEGylated exosomes administered after the initial dose. Single injections of PEGylated exosomes in mice induced anti-PEG IgM production in a T cell-dependent manner. The anti-PEG IgM production decreased when the injection dose of PEGylated exosomes was further increased. Anti-PEG IgM induced by injection of PEGylated exosomes decreased blood concentrations of a second dose of PEGylated exosomes and suppressed their tumor accumulation in a C26 murine colorectal cancer model. Initial injection doses of either PEGylated liposomes or PEGylated ovalbumin (PEG-OVA), both of them induced anti-PEG IgM production, also decreased the blood concentration of PEGylated exosomes. Interestingly, anti-PEG IgM induced by injection of PEGylated exosomes did not affect the blood concentration of PEG-OVA. These results imply the importance of monitoring anti-PEG IgM when repeat PEGylated exosome doses are required and/or when PEGylated exosomes are used together with other PEGylated therapeutics.
Collapse
Affiliation(s)
- Sherif E Emam
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Nehal E Elsadek
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Amr S Abu Lila
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; Department of Pharmaceutics, College of Pharmacy, Hail University, Hail 81442, Saudi Arabia
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Yoshino Kawaguchi
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan.
| |
Collapse
|
182
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
183
|
Kloos RQ, Mathôt R, Pieters R, van der Sluis IM. Individualized dosing guidelines for PEGasparaginase and factors influencing the clearance: a population pharmacokinetic model. Haematologica 2021; 106:1254-1261. [PMID: 32327497 PMCID: PMC8094082 DOI: 10.3324/haematol.2019.242289] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/09/2020] [Indexed: 12/24/2022] Open
Abstract
Considerable inter- and intra-patient variability exist in serum activity levels of PEGasparaginase, essential for pediatric acute lymphoblastic leukemia treatment. A population pharmacokinetic (popPK) model was developed, identifying patient characteristics explaining these variabilities. Patients (n=92) were treated according to the DCOG ALL-11 protocol, using therapeutic drug monitoring to individualize the PEGasparaginase doses. Non-linear mixed effects modeling (NONMEM) was used to analyze the popPK evaluating several covariates. The final model was validated using an independent database (n=28). Guidelines for starting doses and dose adjustments were developed. A one-compartment model with time-dependent clearance adequately described the popPK. Normalization of clearance and volume of distribution by body surface are (BSA) reduced inter-individual variability. Clearance was 0.084 L/day/m2 for 12.7 days, increasing with 0.082 L/day/m2/day thereafter. Clearance was 38% higher during an infection, and 11-19% higher during induction treatment than intensification and maintenance (p<0.001). Targeting an asparaginase activity level of 100 IU/L, a loading dose of 800 IU/m2 (induction) and 600 IU/m2 (intensification) is advised. In conclusion, variability of PEGasparaginase activity levels can be explained by BSA, treatment phase and the occurrence of an infection. With this popPK model, PEGasparaginase treatment can be individualized further, taking into account these covariates and the dosing guidelines provided.
Collapse
Affiliation(s)
- Robin Q.H. Kloos
- Department of Pediatric Oncology and Hematology, Sophia Children’s Hospital – Erasmus MC, Rotterdam
| | - Ron Mathôt
- Department of Hospital Pharmacy, Amsterdam University Medical Center, University of Amsterdam, Amsterdam
| | - Rob Pieters
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Inge M. van der Sluis
- Department of Pediatric Oncology and Hematology, Sophia Children’s Hospital – Erasmus MC, Rotterdam
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
184
|
Mechanistic investigation of thermosensitive liposome immunogenicity and understanding the drivers for circulation half-life: A polyethylene glycol versus 1,2-dipalmitoyl-sn-glycero-3-phosphodiglycerol study. J Control Release 2021; 333:1-15. [DOI: 10.1016/j.jconrel.2021.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
|
185
|
Bronzeri LB, Gauche C, Gudimard L, Courtial EJ, Marquette C, Felisberti MI. Amphiphilic and segmented polyurethanes based on poly(ε-caprolactone)diol and poly(2-ethyl-2-oxazoline)diol: Synthesis, properties, and a preliminary performance study of the 3D printing. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2021.110449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
186
|
Yeo ELL, Azman N'A, Kah JCY. Stealthiness and Hematocompatibility of Gold Nanoparticles with Pre-Formed Protein Corona. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:4913-4923. [PMID: 33861611 DOI: 10.1021/acs.langmuir.1c00151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Studies have established that a serum protein corona pre-formed around gold nanorods (NRs) could be exploited for loading photosensitizers and chemotherapeutics to result in efficient cell kill in vitro with an extremely low dose. In this study, we further demonstrated that pre-forming a serum protein corona (PC) around citrate-capped NRs (NR-Cit) to form NR-PC conferred them stealth property and high hematocompatibility similar to the common strategy of PEGylating NRs, which would otherwise not be able to evade the immune system. Specifically, the NR-PC caused minimal complement activation with significantly lower formation of the terminal complement complex SC5b-9 measured in human serum containing NR-PC, and this resulted in low uptake by phagocytic U937 monocytes of 5.9% of the initial gold dose compared to 55.8% of NR-Cit. In addition, NR-PC exhibited very low hemolytic activity of less than 0.2% hemolysis with no observable effect on RBC morphology as opposed to 0.6% for NR-Cit at the same concentration of 1 nM NRs. Furthermore, we showed that the high hematocompatibility and stealth property of NR-PC were maintained even after the loading of small molecules, photosensitizer Chlorine e6 (Ce6), into the protein corona, thus further establishing the potential clinical relevance of exploiting the inevitably formed serum protein corona on nanoparticles as an effective delivery vector for small molecular therapeutics.
Collapse
Affiliation(s)
- Eugenia Li Ling Yeo
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Block E4, #04-08, 117583 Singapore
| | - Nurul 'Ain Azman
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Block E4, #04-08, 117583 Singapore
| | - James Chen Yong Kah
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Block E4, #04-08, 117583 Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 21 Lower Kent Ridge Road, 119077 Singapore
| |
Collapse
|
187
|
Wang Y, Chen G, Zhang H, Zhao C, Sun L, Zhao Y. Emerging Functional Biomaterials as Medical Patches. ACS NANO 2021; 15:5977-6007. [PMID: 33856205 DOI: 10.1021/acsnano.0c10724] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Medical patches have been widely explored and applied in various medical fields, especially in wound healing, tissue engineering, and other biomedical areas. Benefiting from emerging biomaterials and advanced manufacturing technologies, great achievements have been made on medical patches to evolve them into a multifunctional medical device for diverse health-care purposes, thus attracting extensive attention and research interest. Here, we provide up-to-date research concerning emerging functional biomaterials as medical patches. An overview of the various approaches to construct patches with micro- and nanoarchitecture is presented and summarized. We then focus on the applications, especially the biomedical applications, of the medical patches, including wound healing, drug delivery, and real-time health monitoring. The challenges and prospects for the future development of the medical patches are also discussed.
Collapse
Affiliation(s)
- Yu Wang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Guopu Chen
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
| | - Han Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Cheng Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
188
|
Banerji A, Wickner PG, Saff R, Stone CA, Robinson LB, Long AA, Wolfson AR, Williams P, Khan DA, Phillips E, Blumenthal KG. mRNA Vaccines to Prevent COVID-19 Disease and Reported Allergic Reactions: Current Evidence and Suggested Approach. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2021; 9:1423-1437. [PMID: 33388478 PMCID: PMC7948517 DOI: 10.1016/j.jaip.2020.12.047] [Citation(s) in RCA: 294] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 12/31/2022]
Abstract
The U.S. Food and Drug Administration (FDA) has recently issued an Emergency Use Authorization (EUA) for 2 highly effective coronavirus disease 2019 (COVID-19) vaccines from Pfizer-BioNTech and Moderna. This has brought hope to millions of Americans in the midst of an ongoing global pandemic. The FDA EUA guidance for both vaccines is to not administer the vaccine to individuals with a known history of a severe allergic reaction (eg, anaphylaxis) to any component of the COVID-19 vaccine. The Centers for Disease Control and Prevention (CDC) additionally advises individuals with a history of an immediate allergic reaction to a vaccine or injectable or any history of anaphylaxis be observed for 30 minutes after COVID-19 vaccination. All other individuals should be observed for 15 minutes after COVID-19 vaccination. Staff at vaccine clinics must be able to identify and manage anaphylaxis. Post-FDA EUA, despite very strong safety signals in both phase 3 trials, reports of possible allergic reactions have raised public concern. To provide reassurance and support during widespread global vaccination, allergists must offer clear guidance to individuals based on the best information available, but also in accordance with the broader recommendations of regulatory agencies. This review summarizes vaccine allergy epidemiology and proposes drug and vaccine allergy expert opinion informed risk stratification for Allergy specialist use in conjunction with guidance of public health and regulatory authorities. The risk stratification schema guide care for (1) individuals with different allergy histories to safely receive their first mRNA COVID-19 vaccine and (2) individuals who develop a reaction to their first dose of mRNA COVID-19 vaccine.
Collapse
Affiliation(s)
- Aleena Banerji
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass.
| | - Paige G Wickner
- Harvard Medical School, Boston, Mass; Division of Allergy and Immunology, Department of Medicine, Brigham and Women's Hospital, Boston, Mass
| | - Rebecca Saff
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Cosby A Stone
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn
| | - Lacey B Robinson
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Aidan A Long
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Anna R Wolfson
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Paul Williams
- Allergy Division, University of Washington School of Medicine, Seattle, Wash
| | - David A Khan
- Division of Allergy & Immunology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Elizabeth Phillips
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn
| | - Kimberly G Blumenthal
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Edward P. Lawrence Center for Quality and Safety, Massachusetts General Hospital, Boston, Mass
| |
Collapse
|
189
|
Novel engineering: Biomimicking erythrocyte as a revolutionary platform for drugs and vaccines delivery. Eur J Pharmacol 2021; 900:174009. [PMID: 33722591 DOI: 10.1016/j.ejphar.2021.174009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/25/2021] [Accepted: 02/28/2021] [Indexed: 02/07/2023]
Abstract
Over the years, extensive studies on erythrocytes, also known as red blood cells (RBCs), as a mechanism for drug delivery, have been explored mainly because the cell itself is the most abundant and has astonishing properties such as a long life span of 100-120 days, low immunogenicity, good biocompatibility, and flexibility. There are various types of RBC-based systems for drug delivery, including those that are genetically engineered, non-genetically engineered RBCs, as well as employing erythrocyte as nanocarriers for drug loading. Although promising, these systems are still in an early development stage. In this review, we aimed to highlight the development of biomimicking RBC-based drug and vaccine delivery systems, as well as the loading methods with illustrative examples. Drug-erythrocyte associations will also be discussed and highlighted in this review. We have highlighted the possibility of exploiting erythrocytes for the sustained delivery of drugs and vaccines, encapsulation of these biological agents within the erythrocyte or coupling to the surface of carrier erythrocytes, and provided insights on genetically- and non-genetically engineered erythrocytes-based strategies. Erythrocytes have been known as effective cellular carriers for therapeutic moieties for several years. Herein, we outline various loading methods that can be used to reap the benefits of these natural carriers. It has been shown that drugs and vaccines can be delivered via erythrocytes but it is important to select appropriate methods for increasing the drug encapsulated or conjugated on the surface of the erythrocyte membrane. The outlined examples will guide the selection of the most effective method as well as the impact of using erythrocytes as delivery systems for drugs and vaccines.
Collapse
|
190
|
Albuquerque LJC, Sincari V, Jäger A, Kucka J, Humajova J, Pankrac J, Paral P, Heizer T, Janouškova O, Davidovich I, Talmon Y, Pouckova P, Štěpánek P, Sefc L, Hruby M, Giacomelli FC, Jäger E. pH-responsive polymersome-mediated delivery of doxorubicin into tumor sites enhances the therapeutic efficacy and reduces cardiotoxic effects. J Control Release 2021; 332:529-538. [PMID: 33716094 DOI: 10.1016/j.jconrel.2021.03.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/11/2021] [Accepted: 03/09/2021] [Indexed: 12/14/2022]
Abstract
The delivery of therapeutics into sites of action by using cargo-delivery platforms potentially minimizes their premature degradation and fast clearance from the bloodstream. Additionally, drug-loaded stimuli-responsive supramolecular assemblies can be produced to respond to the inherent features of tumor microenvironments, such as extracellular acidosis. We report in this framework the use of pH-responsive polymersomes (PSs) manufactured using poly([N-(2-hydroxypropyl)] methacrylamide)35-b-poly[2-(diisopropylamino)ethyl methacrylate]75 as the building unit (PHPMA35-b-PDPA75). The self-assemblies were produced with desired size towards long circulation time and tumor accumulation (hydrodynamic diameter - DH ~ 100 nm), and they could be successfully loaded with 10% w/w DOX (doxorubicin), while maintaining colloidal stability. The DOX loaded amount is presumably mainly burst-released at the acidic microenvironment of tumors thanks to the pH-switchable property of PDPA (pKa ~ 6.8), while reduced drug leakage has been monitored in pH 7.4. Compared to the administration of free DOX, the drug-loaded supramolecular structures greatly enhanced the therapeutic efficacy with effective growth inhibition of EL4 lymphoma tumor model and 100% survival rate in female C57BL/6 black mice over 40 days. The approach also led to reduced cardiotoxic effect. These features highlight the potential application of such nanotechnology-based treatment in a variety of cancer therapies where low local pH is commonly found, and emphasize PHPMA-based nanomedicines as an alternative to PEGylated formulations.
Collapse
Affiliation(s)
- Lindomar J C Albuquerque
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovského nám. 2, 162 06 Prague 6, Czech Republic; Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Avenida dos Estados 5001, Santo André 09210-580, Brazil.
| | - Vladimir Sincari
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovského nám. 2, 162 06 Prague 6, Czech Republic
| | - Alessandro Jäger
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovského nám. 2, 162 06 Prague 6, Czech Republic
| | - Jan Kucka
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovského nám. 2, 162 06 Prague 6, Czech Republic
| | - Jana Humajova
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Salmovska, 1, 120 00 Prague, Czech Republic
| | - Jan Pankrac
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Salmovská 3, Prague 2, 120 00, Czech Republic
| | - Petr Paral
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Salmovská 3, Prague 2, 120 00, Czech Republic
| | - Tomas Heizer
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Salmovská 3, Prague 2, 120 00, Czech Republic
| | - Olga Janouškova
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovského nám. 2, 162 06 Prague 6, Czech Republic
| | - Irina Davidovich
- Department of Chemical Engineering and the Russell Berrie Nanotechnology Institute (RBNI), Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Yeshayahu Talmon
- Department of Chemical Engineering and the Russell Berrie Nanotechnology Institute (RBNI), Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Pavla Pouckova
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Salmovska, 1, 120 00 Prague, Czech Republic
| | - Petr Štěpánek
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovského nám. 2, 162 06 Prague 6, Czech Republic
| | - Ludek Sefc
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Salmovská 3, Prague 2, 120 00, Czech Republic
| | - Martin Hruby
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovského nám. 2, 162 06 Prague 6, Czech Republic
| | - Fernando C Giacomelli
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Avenida dos Estados 5001, Santo André 09210-580, Brazil
| | - Eliézer Jäger
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovského nám. 2, 162 06 Prague 6, Czech Republic.
| |
Collapse
|
191
|
Protein corona components of polyethylene glycol-conjugated organosilica nanoparticles modulates macrophage uptake. Colloids Surf B Biointerfaces 2021; 199:111527. [DOI: 10.1016/j.colsurfb.2020.111527] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/25/2022]
|
192
|
Martínez-Negro M, González-Rubio G, Aicart E, Landfester K, Guerrero-Martínez A, Junquera E. Insights into colloidal nanoparticle-protein corona interactions for nanomedicine applications. Adv Colloid Interface Sci 2021; 289:102366. [PMID: 33540289 DOI: 10.1016/j.cis.2021.102366] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 12/17/2022]
Abstract
Colloidal nanoparticles (NPs) have attracted significant attention due to their unique physicochemical properties suitable for diagnosing and treating different human diseases. Nevertheless, the successful implementation of NPs in medicine demands a proper understanding of their interactions with the different proteins found in biological fluids. Once introduced into the body, NPs are covered by a protein corona (PC) that determines the biological behavior of the NPs. The formation of the PC can eventually favor the rapid clearance of the NPs from the body before fulfilling the desired objective or lead to increased cytotoxicity. The PC nature varies as a function of the different repulsive and attractive forces that govern the NP-protein interaction and their colloidal stability. This review focuses on the phenomenon of PC formation on NPs from a physicochemical perspective, aiming to provide a general overview of this critical process. Main issues related to NP toxicity and clearance from the body as a result of protein adsorption are covered, including the most promising strategies to control PC formation and, thereby, ensure the successful application of NPs in nanomedicine.
Collapse
|
193
|
Effect of C-terminus Conjugation via Different Conjugation Chemistries on In Vivo Activity of Albumin-Conjugated Recombinant GLP-1. Pharmaceutics 2021; 13:pharmaceutics13020263. [PMID: 33672039 PMCID: PMC7919490 DOI: 10.3390/pharmaceutics13020263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/12/2021] [Accepted: 02/12/2021] [Indexed: 12/15/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is a peptide hormone with tremendous therapeutic potential for treating type 2 diabetes mellitus. However, the short half-life of its native form is a significant drawback. We previously prolonged the plasma half-life of GLP-1 via site-specific conjugation of human serum albumin (HSA) at position 16 of recombinant GLP-1 using site-specific incorporation of p-azido-phenylalanine (AzF) and strain-promoted azide-alkyne cycloaddition (SPAAC). However, the resulting conjugate GLP1_8G16AzF-HSA showed only moderate in vivo glucose-lowering activity, probably due to perturbed interactions with GLP-1 receptor (GLP-1R) caused by the albumin-linker. To identify albumin-conjugated GLP-1 variants with enhanced in vivo glucose-lowering activity, we investigated the conjugation of HSA to a C-terminal region of GLP-1 to reduce steric hindrance by the albumin-linker using two different conjugation chemistries. GLP-1 variants GLP1_8G37AzF-HSA and GLP1_8G37C-HSA were prepared using SPAAC and Michael addition, respectively. GLP1_8G37C-HSA exhibited a higher glucose-lowering activity in vivo than GLP1_8G16AzF-HSA, while GLP1_8G37AzF-HSA did not. Another GLP-1 variant, GLP1_8A37C-HSA, had a glycine to alanine mutation at position 8 and albumin at its C-terminus and exhibited in vivo glucose-lowering activity comparable to that of GLP1_8G37C-HSA, despite a moderately shorter plasma half-life. These results showed that site-specific HSA conjugation to the C-terminus of GLP-1 via Michael addition could be used to generate GLP-1 variants with enhanced glucose-lowering activity and prolonged plasma half-life in vivo.
Collapse
|
194
|
Systematic Studies of Gold Nanoparticles Functionalised with Thioglucose and its Cytotoxic Effect. ChemistrySelect 2021. [DOI: 10.1002/slct.202100034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
195
|
Differential Immunomodulatory Effect of Carbon Dots Influenced by the Type of Surface Passivation Agent. Inflammation 2021; 43:777-783. [PMID: 31873835 DOI: 10.1007/s10753-019-01165-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Carbon nanodots (CDs) are often synthesized from natural sources including honey, molasses, fruits, and foods, and plant extracts simply through caramelization. They have wide biological applications especially as drug delivery vehicles and bioimaging agent due to their small size and biocompatibility. This article details the synthesis of carbon dots from carob and its derivatives by surface passivation with polyethylene glycol (PEG), polyvinyl alcohol (PVA), and alginate (ALG). We investigated the immune response against CDs and evaluated the effect of surface passivation agents on their immunomodulatory functions. CDPVA had strong anti-inflammatory activities, whereas CDALG were pro-inflammatory. CDPEG had mild anti-inflammatory activities suggesting that these CDs can be used in the drug delivery studies as inert carriers. These results showed that depending on the type of activated groups dominated on the surface, CDs exerted differential effects on the inflammatory potential of the macrophages by changing the pro-inflammatory TNFα and IL6 production levels.
Collapse
|
196
|
Zerrillo L, Gupta KBSS, Lefeber FA, Da Silva CG, Galli F, Chan A, Veltien A, Dou W, Censi R, Di Martino P, Srinivas M, Cruz L. Novel Fluorinated Poly (Lactic-Co-Glycolic acid) (PLGA) and Polyethylene Glycol (PEG) Nanoparticles for Monitoring and Imaging in Osteoarthritis. Pharmaceutics 2021; 13:pharmaceutics13020235. [PMID: 33562356 PMCID: PMC7914518 DOI: 10.3390/pharmaceutics13020235] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 01/28/2023] Open
Abstract
Polymeric nanoparticles (NPs) find many uses in nanomedicine, from drug delivery to imaging. In this regard, poly (lactic-co-glycolic acid) (PLGA) and polyethylene glycol (PEG) particles are the most widely applied types of nano-systems due to their biocompatibility and biodegradability. Here we developed novel fluorinated polymeric NPs as vectors for multi-modal nanoprobes. This approach involved modifying polymeric NPs with trifluoroacetamide (TFA) and loading them with a near-infrared (NIR) dye for different imaging modalities, such as magnetic resonance imaging (MRI) and optical imaging. The PLGA-PEG-TFA NPs generated were characterized in vitro using the C28/I2 human chondrocyte cell line and in vivo in a mouse model of osteoarthritis (OA). The NPs were well absorbed, as confirmed by confocal microscopy, and were non-toxic to cells. To test the NPs as a drug delivery system for contrast agents of OA, the nanomaterial was administered via the intra-articular (IA) administration method. The dye-loaded NPs were injected in the knee joint and then visualized and tracked in vivo by fluorine-19 nuclear magnetic resonance and fluorescence imaging. Here, we describe the development of novel intrinsically fluorinated polymeric NPs modality that can be used in various molecular imaging techniques to visualize and track OA treatments and their potential use in clinical trials.
Collapse
Affiliation(s)
- Luana Zerrillo
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Centrum, 2333 ZA Leiden, The Netherlands
- Percuros-Tarweveld 5, 6617 CD Bergharen, The Netherlands
| | | | - Fons A.W.M. Lefeber
- Institute of Chemistry, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Candido G. Da Silva
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Centrum, 2333 ZA Leiden, The Netherlands
| | - Federica Galli
- Leiden Institute of Physics, Niels Bohrweg 2, 2333 CA Leiden, the Netherlands
| | - Alan Chan
- Percuros-Tarweveld 5, 6617 CD Bergharen, The Netherlands
| | - Andor Veltien
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Weiqiang Dou
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Roberta Censi
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy
| | - Piera Di Martino
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud University Medical Center, 6525 XZ Nijmegen, The Netherlands
| | - Luis Cruz
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Centrum, 2333 ZA Leiden, The Netherlands
- Correspondence: ; Tel.: +31-71-5263025
| |
Collapse
|
197
|
Pędziwiatr-Werbicka E, Gorzkiewicz M, Horodecka K, Lach D, Barrios-Gumiel A, Sánchez-Nieves J, Gómez R, de la Mata FJ, Bryszewska M. PEGylation of Dendronized Gold Nanoparticles Affects Their Interaction with Thrombin and siRNA. J Phys Chem B 2021; 125:1196-1206. [PMID: 33481607 DOI: 10.1021/acs.jpcb.0c10177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The use of nonviral carriers based on nanomaterials is a promising strategy for modern gene therapy aimed at protecting the genetic material against degradation and enabling its efficient cellular uptake. To improve the effectiveness of nanocarriers in vivo, they are often modified with poly(ethylene glycol) (PEG) to reduce their toxicity, limit nonspecific binding by proteins in the bloodstream, and extend blood half-life. Thus, the selection of an appropriate degree of surface PEGylation is crucial to preserve the interaction of nanoparticles with the genetic material and to ensure its efficient transport to the site of action. Our research focuses on the use of innovative gold nanoparticles (AuNPs) coated with cationic carbosilane dendrons as carriers of siRNA. In this study, using dynamic light scattering and zeta potential measurements, circular dichroism, and gel electrophoresis, we investigated dendronized AuNPs modified to varying degrees with PEG in terms of their interactions with siRNA and thrombin to select the most promising PEGylated carrier for further research.
Collapse
Affiliation(s)
- Elżbieta Pędziwiatr-Werbicka
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska Street, 90-236 Lodz, Poland
| | - Michał Gorzkiewicz
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska Street, 90-236 Lodz, Poland
| | - Katarzyna Horodecka
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska Street, 90-236 Lodz, Poland
| | - Dominika Lach
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska Street, 90-236 Lodz, Poland
| | - Andrea Barrios-Gumiel
- Department of Organic and Inorganic Chemistry and Research Chemistry Institute "Andrés M. del Río" (IQAR), University of Alcalá, 28871 Alcalá de Henares, Spain.,Networking Research Center for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain.,Institute "Ramón y Cajal" for Health Research (IRYCIS), 28034 Madrid, Spain
| | - Javier Sánchez-Nieves
- Department of Organic and Inorganic Chemistry and Research Chemistry Institute "Andrés M. del Río" (IQAR), University of Alcalá, 28871 Alcalá de Henares, Spain.,Networking Research Center for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain.,Institute "Ramón y Cajal" for Health Research (IRYCIS), 28034 Madrid, Spain
| | - Rafael Gómez
- Department of Organic and Inorganic Chemistry and Research Chemistry Institute "Andrés M. del Río" (IQAR), University of Alcalá, 28871 Alcalá de Henares, Spain.,Networking Research Center for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain.,Institute "Ramón y Cajal" for Health Research (IRYCIS), 28034 Madrid, Spain
| | - Francisco Javier de la Mata
- Department of Organic and Inorganic Chemistry and Research Chemistry Institute "Andrés M. del Río" (IQAR), University of Alcalá, 28871 Alcalá de Henares, Spain.,Networking Research Center for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain.,Institute "Ramón y Cajal" for Health Research (IRYCIS), 28034 Madrid, Spain
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska Street, 90-236 Lodz, Poland
| |
Collapse
|
198
|
Gagliardi A, Giuliano E, Venkateswararao E, Fresta M, Bulotta S, Awasthi V, Cosco D. Biodegradable Polymeric Nanoparticles for Drug Delivery to Solid Tumors. Front Pharmacol 2021; 12:601626. [PMID: 33613290 PMCID: PMC7887387 DOI: 10.3389/fphar.2021.601626] [Citation(s) in RCA: 196] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/04/2021] [Indexed: 12/24/2022] Open
Abstract
Advances in nanotechnology have favored the development of novel colloidal formulations able to modulate the pharmacological and biopharmaceutical properties of drugs. The peculiar physico-chemical and technological properties of nanomaterial-based therapeutics have allowed for several successful applications in the treatment of cancer. The size, shape, charge and patterning of nanoscale therapeutic molecules are parameters that need to be investigated and modulated in order to promote and optimize cell and tissue interaction. In this review, the use of polymeric nanoparticles as drug delivery systems of anticancer compounds, their physico-chemical properties and their ability to be efficiently localized in specific tumor tissues have been described. The nanoencapsulation of antitumor active compounds in polymeric systems is a promising approach to improve the efficacy of various tumor treatments.
Collapse
Affiliation(s)
- Agnese Gagliardi
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Elena Giuliano
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Eeda Venkateswararao
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Massimo Fresta
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Stefania Bulotta
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Vibhudutta Awasthi
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Donato Cosco
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
199
|
Arora S, Layek B, Singh J. Design and Validation of Liposomal ApoE2 Gene Delivery System to Evade Blood-Brain Barrier for Effective Treatment of Alzheimer's Disease. Mol Pharm 2021; 18:714-725. [PMID: 32787268 PMCID: PMC10292003 DOI: 10.1021/acs.molpharmaceut.0c00461] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Targeting gene-based therapeutics to the brain is a strategy actively sought to treat Alzheimer's disease (AD). Recent findings discovered the role of apolipoprotein E (ApoE) isoforms in the clearance of toxic amyloid beta proteins from the brain. ApoE2 isoform is beneficial for preventing AD development, whereas ApoE4 is a major contributing factor to the disease. In this paper, we demonstrated efficient brain-targeted delivery of ApoE2 encoding plasmid DNA (pApoE2) using glucose transporter-1 (glut-1) targeted liposomes. Liposomes were surface-functionalized with a glut-1 targeting ligand mannose (MAN) and a cell-penetrating peptide (CPP) to enhance brain-targeting and cellular internalization, respectively. Among various CPPs, rabies virus glycoprotein peptide (RVG) or penetratin (Pen) was selected as a cell-penetration enhancer. Dual (RVGMAN and PenMAN)-functionalized liposomes were cytocompatible at 100 nM phospholipid concentration and demonstrated significantly higher expression of ApoE2 in bEnd.3 cells, primary neurons, and astrocytes compared to monofunctionalized and unmodified (plain) liposomes. Dual-modified liposomes also showed ∼2 times higher protein expression than other formulation controls in neurons cultured below the in vitro BBB model. These results translated well to in vivo efficacy study with significantly higher transfection of pApoE2 in the C57BL/6 mice brain following single tail vein administration of RVGMAN and PenMAN functionalized liposomes without any noticeable signs of toxicity. These results illustrate the potential of surface-modified liposomes for safe and brain-targeted delivery of the pApoE2 gene for effective AD therapy.
Collapse
Affiliation(s)
- Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, 58105 North Dakota, United States
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, 58105 North Dakota, United States
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, 58105 North Dakota, United States
| |
Collapse
|
200
|
Li B, Chu F, Lu Q, Wang Y, Lane LA. Alternating stealth polymer coatings between administrations minimizes toxic and antibody immune responses towards nanomedicine treatment regimens. Acta Biomater 2021; 121:527-540. [PMID: 33285326 DOI: 10.1016/j.actbio.2020.11.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 02/08/2023]
Abstract
In efforts to achieve minimal systemic toxicity and high tumor delivery efficiencies in cancer therapy, various nanomedicine formulations having stealth polymer coatings have been developed for minimizing immune cell uptake and off-target macrophage phagocyte system (MPS) organ accumulation. Despite an initial reduction in immune cell uptake, stealth nanoparticles still initiate an antibody immune response. This response acts on subsequent administrations in treatment regimens resulting in accelerated blood clearance of particles into MPS organs, particularly the liver, where they are retained for prolonged periods. Consequently, doses after the first administration in treatment regimens have diminished tumor accumulation and increased MPS toxicity. Here, we present a strategy reducing antibody responses to each dose in a treatment regimen by alternating between polyethylene-glycol and polymethyloxazoline polymers as the nanoparticle coating between administrations. In a weekly dosing regimen, we find that the first dose of particles having either coating display similar favorable pharmacokinetics and biodistributions, thus allowing the polymers to be used interchangeably. However, when maintaining the same coating in subsequent administrations, we find that particles are in circulation at the height of the antibody immune response resulting in 50-60% decreases of circulation half-lives and tumor accumulation along with 50% increases in liver accumulation. By alternating the polymers used in the nanoparticle coating between administrations, we find each dose maintains favorable in vivo behaviors at the height of the antibody immune response to the previous administration. Furthermore, our strategy increases the clearance of particles uptaken by macrophages and hepatocytes, resulting in marked decreases in hepatotoxicity.
Collapse
|