301
|
Suzuki Y, Ishihara H. Difference in the lipid nanoparticle technology employed in three approved siRNA (Patisiran) and mRNA (COVID-19 vaccine) drugs. Drug Metab Pharmacokinet 2021; 41:100424. [PMID: 34757287 PMCID: PMC8502116 DOI: 10.1016/j.dmpk.2021.100424] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/04/2021] [Accepted: 10/08/2021] [Indexed: 12/31/2022]
Abstract
Nucleic acid therapeutics are developing into precise medicines that can manipulate specific genes. However, the development of safe and effective delivery system for the target cells has remained a challenge. Lipid nanoparticles (LNPs) have provided a revolutionary delivery system that can ensure multiple clinical translation of RNA-based candidates. In 2018, Patisiran (Onpattro) was first approved as an LNP-based siRNA drug. In 2020, during the coronavirus disease 2019 (COVID-19) outbreak, LNPs have enabled the development of two SARS-CoV-2 mRNA vaccines, Tozinameran (Comirnaty or Pfizer-BioNTech COVID-19 vaccine) and Elasomeran (Spikevax or COVID-19 vaccine Moderna) for conditional approval. Here, we reviewed the state-of-the-art LNP technology employed in three approved drugs (one siRNA-based and two mRNA-based drugs) and discussed the differences in their mode of action, formulation design, and biodistribution.
Collapse
Affiliation(s)
- Yuta Suzuki
- hhc Data Creation Center, Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan.
| | - Hiroshi Ishihara
- hhc Data Creation Center, Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan; Department of Formulation Science and Technology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| |
Collapse
|
302
|
Kuroki A, Tay J, Lee GH, Yang YY. Broad-Spectrum Antiviral Peptides and Polymers. Adv Healthc Mater 2021; 10:e2101113. [PMID: 34599850 DOI: 10.1002/adhm.202101113] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/13/2021] [Indexed: 12/18/2022]
Abstract
As the human cost of the pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still being witnessed worldwide, the development of broad-spectrum antiviral agents against emerging and re-emerging viruses is seen as a necessity to hamper the spread of infections. Various targets during the viral life-cycle can be considered to inhibit viral infection, from viral attachment to viral fusion or replication. Macromolecules represent a particularly attractive class of therapeutics due to their multivalency and versatility. Although several antiviral macromolecules hold great promise in clinical applications, the emergence of resistance after prolonged exposure urges the need for improved solutions. In the present article, the recent advancement in the discovery of antiviral peptides and polymers with diverse structural features and antiviral mechanisms is reviewed. Future perspectives, such as, the development of virucidal peptides/polymers and their coatings against SARS-CoV-2 infection, standardization of antiviral testing protocols, and use of artificial intelligence or machine learning as a tool to accelerate the discovery of antiviral macromolecules, are discussed.
Collapse
Affiliation(s)
- Agnès Kuroki
- Yong Loo Lin School of Medicine National University of Singapore Singapore 117597 Singapore
- Institute of Bioengineering and Bioimaging 31 Biopolis Ways, The Nanos Singapore 138669 Singapore
| | - Joyce Tay
- Institute of Bioengineering and Bioimaging 31 Biopolis Ways, The Nanos Singapore 138669 Singapore
| | - Guan Huei Lee
- Yong Loo Lin School of Medicine National University of Singapore Singapore 117597 Singapore
| | - Yi Yan Yang
- Institute of Bioengineering and Bioimaging 31 Biopolis Ways, The Nanos Singapore 138669 Singapore
| |
Collapse
|
303
|
Tsakiri M, Peraki A, Chountoulesi M, Demetzos C. Chimeric liposomes decorated with P407: an alternative biomaterial for producing stealth nano-therapeutics. J Liposome Res 2021; 32:83-91. [PMID: 34839768 DOI: 10.1080/08982104.2021.1978486] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The aim of the present study is the development and evaluation of the physicochemical properties of chimeric hydrogenated soya phosphatidylcholine (HSPC) and egg phosphatidylcholine (EggPC) liposomes with incorporated triblock copolymer Poloxamer P407 (P407). The physicochemical assay was held in water HPLC-grade and Foetal Bovine Serum (FBS), in order to determine whether these systems can be used as drug or antigen delivery nanosystems. Dynamic and electrophoretic light scattering (DLS/ELS) techniques were used for the measurement of the hydrodynamic diameter, the polydispersity index, and the ζ-potential of the prepared nanosystems. The incorporation of the P407 resulted in a size reduction of all systems. A decrease in the hydrodynamic diameter and polydispersity index were also found as a result of increasing the storage temperature from 4 °C to 25 °C, attributed to P407. The experiments that were carried out in FBS, showed that the addition of P407 improved systems stealth properties. Concluding, we propose P407 as a promising alternative to PEG in the development of lipid nanoparticles with optimized bio- and shelf-stability.
Collapse
Affiliation(s)
- Maria Tsakiri
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Peraki
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Chountoulesi
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
304
|
Tropism of Extracellular Vesicles and Cell-Derived Nanovesicles to Normal and Cancer Cells: New Perspectives in Tumor-Targeted Nucleic Acid Delivery. Pharmaceutics 2021; 13:pharmaceutics13111911. [PMID: 34834326 PMCID: PMC8621453 DOI: 10.3390/pharmaceutics13111911] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
The main advantage of extracellular vesicles (EVs) as a drug carrier system is their low immunogenicity and internalization by mammalian cells. EVs are often considered a cell-specific delivery system, but the production of preparative amounts of EVs for therapeutic applications is challenging due to their laborious isolation and purification procedures. Alternatively, mimetic vesicles prepared from the cellular plasma membrane can be used in the same way as natural EVs. For example, a cytoskeleton-destabilizing agent, such as cytochalasin B, allows the preparation of membrane vesicles by a series of centrifugations. Here, we prepared cytochalasin-B-inducible nanovesicles (CINVs) of various cellular origins and studied their tropism in different mammalian cells. We observed that CINVs derived from human endometrial mesenchymal stem cells exhibited an enhanced affinity to epithelial cancer cells compared to myeloid, lymphoid or neuroblastoma cancer cells. The dendritic cell-derived CINVs were taken up by all studied cell lines with a similar efficiency that differed from the behavior of DC-derived EVs. The ability of cancer cells to internalize CINVs was mainly determined by the properties of recipient cells, and the cellular origin of CINVs was less important. In addition, receptor-mediated interactions were shown to be necessary for the efficient uptake of CINVs. We found that CINVs, derived from late apoptotic/necrotic cells (aCINVs) are internalized by in myelogenous (K562) 10-fold more efficiently than CINVs, and interact much less efficiently with melanocytic (B16) or epithelial (KB-3-1) cancer cells. Finally, we found that CINVs caused a temporal and reversible drop of the rate of cell division, which restored to the level of control cells with a 24 h delay.
Collapse
|
305
|
Niculescu AG, Grumezescu AM. Polymer-Based Nanosystems-A Versatile Delivery Approach. MATERIALS (BASEL, SWITZERLAND) 2021; 14:6812. [PMID: 34832213 PMCID: PMC8619478 DOI: 10.3390/ma14226812] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 01/10/2023]
Abstract
Polymer-based nanoparticles of tailored size, morphology, and surface properties have attracted increasing attention as carriers for drugs, biomolecules, and genes. By protecting the payload from degradation and maintaining sustained and controlled release of the drug, polymeric nanoparticles can reduce drug clearance, increase their cargo's stability and solubility, prolong its half-life, and ensure optimal concentration at the target site. The inherent immunomodulatory properties of specific polymer nanoparticles, coupled with their drug encapsulation ability, have raised particular interest in vaccine delivery. This paper aims to review current and emerging drug delivery applications of both branched and linear, natural, and synthetic polymer nanostructures, focusing on their role in vaccine development.
Collapse
Affiliation(s)
- Adelina-Gabriela Niculescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania;
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania;
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
- Academy of Romanian Scientists, Ilfov no. 3, 50044 Bucharest, Romania
| |
Collapse
|
306
|
Talkington AM, Wessler T, Lai SK, Cao Y, Forest MG. Experimental Data and PBPK Modeling Quantify Antibody Interference in PEGylated Drug Carrier Delivery. Bull Math Biol 2021; 83:123. [PMID: 34751832 PMCID: PMC8576315 DOI: 10.1007/s11538-021-00950-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 09/27/2021] [Indexed: 12/30/2022]
Abstract
Physiologically-based pharmacokinetic (PBPK) modeling is a popular drug development tool that integrates physiology, drug physicochemical properties, preclinical data, and clinical information to predict drug systemic disposition. Since PBPK models seek to capture complex physiology, parameter uncertainty and variability is a prevailing challenge: there are often more compartments (e.g., organs, each with drug flux and retention mechanisms, and associated model parameters) than can be simultaneously measured. To improve the fidelity of PBPK modeling, one approach is to search and optimize within the high-dimensional model parameter space, based on experimental time-series measurements of drug distributions. Here, we employ Latin Hypercube Sampling (LHS) on a PBPK model of PEG-liposomes (PL) that tracks biodistribution in an 8-compartment mouse circulatory system, in the presence (APA+) or absence (naïve) of anti-PEG antibodies (APA). Near-continuous experimental measurements of PL concentration during the first hour post-injection from the liver, spleen, kidney, muscle, lung, and blood plasma, based on PET/CT imaging in live mice, are used as truth sets with LHS to infer optimal parameter ranges for the full PBPK model. The data and model quantify that PL retention in the liver is the primary differentiator of biodistribution patterns in naïve versus APA+ mice, and spleen the secondary differentiator. Retention of PEGylated nanomedicines is substantially amplified in APA+ mice, likely due to PL-bound APA engaging specific receptors in the liver and spleen that bind antibody Fc domains. Our work illustrates how applying LHS to PBPK models can further mechanistic understanding of the biodistribution and antibody-mediated clearance of specific drugs.
Collapse
Affiliation(s)
- Anne M Talkington
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA.
| | - Timothy Wessler
- Department of Mathematics, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Samuel K Lai
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina, Chapel Hill, NC, USA
| | - M Gregory Forest
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA.
- Department of Mathematics, University of North Carolina, Chapel Hill, NC, USA.
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA.
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
307
|
Constantin C, Pisani A, Bardi G, Neagu M. Nano-carriers of COVID-19 vaccines: the main pillars of efficacy. Nanomedicine (Lond) 2021; 16:2377-2387. [PMID: 34632802 PMCID: PMC8544481 DOI: 10.2217/nnm-2021-0250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/09/2021] [Indexed: 12/30/2022] Open
Abstract
As the current COVID-19 pandemic illustrates, vaccination is the most powerful method of disease prevention and public confidence in vaccines depends on their safety and efficacy. The information gathered in the current pandemic is growing at an accelerated pace. Both the key vital protein DNA/RNA messengers and the delivery carriers are the elements of a puzzle including their interactions with the immune system to suppress SARS-CoV-2 infection. A new nano-era is beginning in the vaccine development field and an array of side applications for diagnostic and antiviral tools will likely emerge. This review focuses on the evolution of vaccine carriers up to COVID-19-aimed nanoparticles and the immune-related adverse effects imposed by these nanocarriers.
Collapse
Affiliation(s)
- Carolina Constantin
- “Victor Babeş” National Institute of Pathology, 99-101 Spl Independentei, Bucharest, 050096, Romania
- Colentina Clinical Hospital, 19-21, Sos. Stefan cel Mare, Bucharest, Romania
| | - Anissa Pisani
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia, Via Morego 30, Genova, 16163, Italy
- Department of Chemistry & Industrial Chemistry, University of Genova, Via Dodecaneso 31, Genova, 16146, Italy
| | - Giuseppe Bardi
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia, Via Morego 30, Genova, 16163, Italy
| | - Monica Neagu
- “Victor Babeş” National Institute of Pathology, 99-101 Spl Independentei, Bucharest, 050096, Romania
- Colentina Clinical Hospital, 19-21, Sos. Stefan cel Mare, Bucharest, Romania
- University of Bucharest, 93–95 Spl Independentei, Bucharest, Romania
| |
Collapse
|
308
|
Ang MJY, Chan SY, Goh YY, Luo Z, Lau JW, Liu X. Emerging strategies in developing multifunctional nanomaterials for cancer nanotheranostics. Adv Drug Deliv Rev 2021; 178:113907. [PMID: 34371084 DOI: 10.1016/j.addr.2021.113907] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/09/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022]
Abstract
Cancer involves a collection of diseases with a common trait - dysregulation in cell proliferation. At present, traditional therapeutic strategies against cancer have limitations in tackling various tumors in clinical settings. These include chemotherapeutic resistance and the inability to overcome intrinsic physiological barriers to drug delivery. Nanomaterials have presented promising strategies for tumor treatment in recent years. Nanotheranostics combine therapeutic and bioimaging functionalities at the single nanoparticle level and have experienced tremendous growth over the past few years. This review highlights recent developments of advanced nanomaterials and nanotheranostics in three main directions: stimulus-responsive nanomaterials, nanocarriers targeting the tumor microenvironment, and emerging nanomaterials that integrate with phototherapies and immunotherapies. We also discuss the cytotoxicity and outlook of next-generation nanomaterials towards clinical implementation.
Collapse
Affiliation(s)
- Melgious Jin Yan Ang
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Siew Yin Chan
- Institute of Materials Research and Engineering, Agency for Science, Technology, and Research, Singapore 138634, Singapore
| | - Yi-Yiing Goh
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Zichao Luo
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Jun Wei Lau
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore.
| |
Collapse
|
309
|
Chaudhary N, Weissman D, Whitehead KA. mRNA vaccines for infectious diseases: principles, delivery and clinical translation. Nat Rev Drug Discov 2021; 20:817-838. [PMID: 34433919 PMCID: PMC8386155 DOI: 10.1038/s41573-021-00283-5] [Citation(s) in RCA: 730] [Impact Index Per Article: 182.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
Over the past several decades, messenger RNA (mRNA) vaccines have progressed from a scepticism-inducing idea to clinical reality. In 2020, the COVID-19 pandemic catalysed the most rapid vaccine development in history, with mRNA vaccines at the forefront of those efforts. Although it is now clear that mRNA vaccines can rapidly and safely protect patients from infectious disease, additional research is required to optimize mRNA design, intracellular delivery and applications beyond SARS-CoV-2 prophylaxis. In this Review, we describe the technologies that underlie mRNA vaccines, with an emphasis on lipid nanoparticles and other non-viral delivery vehicles. We also overview the pipeline of mRNA vaccines against various infectious disease pathogens and discuss key questions for the future application of this breakthrough vaccine platform.
Collapse
Affiliation(s)
- Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
310
|
Tully M, Hauptstein N, Licha K, Meinel L, Lühmann T, Haag R. Linear Polyglycerol for N-terminal-selective Modification of Interleukin-4. J Pharm Sci 2021; 111:1642-1651. [PMID: 34728175 DOI: 10.1016/j.xphs.2021.10.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 01/13/2023]
Abstract
Polymer conjugation to biologics is of key interest to the pharmaceutical industry for the development of potent and long acting biotherapeutics, with poly(ethylene glycol) (PEG) being the gold standard. Within the last years, unwanted PEG-related side effects (immunological reactions, antibody formation) arose, therefore creating several attempts to establish alternative polymers with similar potential to PEG. In this article, we synthesized N-terminal bioconjugates of the potential therapeutic human interleukin-4 (hIL-4 WT) with linear polyglycerol (LPG) of 10 and 40 kDa and compared it with its PEG analogs of same nominal weights. Polyglycerol is a highly hydrophilic polymer with good biocompatibility and therefore represents an alternative polymer to PEG. Both polymer types resulted in similar conjugation yields, comparable hydrodynamic sizes and an unaltered secondary structure of the protein after modification. LPG- and PEG-bioconjugates remained stable in human plasma, whereas binding to human serum albumin (HSA) decreased after polymer modification. Furthermore, only minor differences in bioactivity were observed between LPG- and PEG-bioconjugates of same nominal weights. The presented findings are promising for future pharmacokinetic evaluation of hIL-4-polymer bioconjugates.
Collapse
Affiliation(s)
- Michael Tully
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin Germany
| | - Niklas Hauptstein
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg Germany
| | - Kai Licha
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin Germany
| | - Lorenz Meinel
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg Germany; Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), 97080 Würzburg, Germany
| | - Tessa Lühmann
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin Germany.
| |
Collapse
|
311
|
Onishchenko N, Tretiakova D, Vodovozova E. Spotlight on the protein corona of liposomes. Acta Biomater 2021; 134:57-78. [PMID: 34364016 DOI: 10.1016/j.actbio.2021.07.074] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/19/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022]
Abstract
Although an established drug delivery platform, liposomes have not fulfilled their true potential. In the body, interactions of liposomes are mediated by the layer of plasma proteins adsorbed on the surface, the protein corona. The review aims to collect the data of the last decade on liposome protein corona, tracing the path from interactions of individual proteins to the effects mediated by the protein corona in vivo. It offers a classification of the approaches to exploitation of the protein corona-rather than elimination thereof-based on the bilayer composition-corona composition-molecular interactions-biological performance framework. The multitude of factors that affect each level of this relationship urge to the widest implementation of bioinformatics tools to predict the most effective liposome compositions relying on the data on protein corona. Supplementing the picture with new pieces of accurately reported experimental data will contribute to the accuracy and efficiency of the predictions. STATEMENT OF SIGNIFICANCE: The review focuses on liposomes as an established nanomedicine platform and analyzes the available data on how the protein corona formed on liposome surface in biological fluids affects performance of the liposomes. The review offers a rigorous account of existing literature and critical analysis of methodology currently applied to the assessment of liposome-plasma protein interactions. It introduces a classification of the approaches to exploitation of the protein corona and tailoring liposome carriers to advance the field of nanoparticulate drug delivery systems for the benefit of patients.
Collapse
|
312
|
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: 34] [Impact Index Per Article: 8.5] [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
|
313
|
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: 251] [Impact Index Per Article: 62.8] [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
|
314
|
Warren CM, Snow TT, Lee AS, Shah MM, Heider A, Blomkalns A, Betts B, Buzzanco AS, Gonzalez J, Chinthrajah RS, Do E, Chang I, Dunham D, Lee G, O’Hara R, Park H, Shamji MH, Schilling L, Sindher SB, Sisodiya D, Smith E, Tsai M, Galli SJ, Akdis C, Nadeau KC. Assessment of Allergic and Anaphylactic Reactions to mRNA COVID-19 Vaccines With Confirmatory Testing in a US Regional Health System. JAMA Netw Open 2021; 4:e2125524. [PMID: 34533570 PMCID: PMC8449279 DOI: 10.1001/jamanetworkopen.2021.25524] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
IMPORTANCE As of May 2021, more than 32 million cases of COVID-19 have been confirmed in the United States, resulting in more than 615 000 deaths. Anaphylactic reactions associated with the Food and Drug Administration (FDA)-authorized mRNA COVID-19 vaccines have been reported. OBJECTIVE To characterize the immunologic mechanisms underlying allergic reactions to these vaccines. DESIGN, SETTING, AND PARTICIPANTS This case series included 22 patients with suspected allergic reactions to mRNA COVID-19 vaccines between December 18, 2020, and January 27, 2021, at a large regional health care network. Participants were individuals who received at least 1 of the following International Statistical Classification of Diseases and Related Health Problems, Tenth Revision anaphylaxis codes: T78.2XXA, T80.52XA, T78.2XXD, or E949.9, with documentation of COVID-19 vaccination. Suspected allergy cases were identified and invited for follow-up allergy testing. EXPOSURES FDA-authorized mRNA COVID-19 vaccines. MAIN OUTCOMES AND MEASURES Allergic reactions were graded using standard definitions, including Brighton criteria. Skin prick testing was conducted to polyethylene glycol (PEG) and polysorbate 80 (P80). Histamine (1 mg/mL) and filtered saline (negative control) were used for internal validation. Basophil activation testing after stimulation for 30 minutes at 37 °C was also conducted. Concentrations of immunoglobulin (Ig) G and IgE antibodies to PEG were obtained to determine possible mechanisms. RESULTS Of 22 patients (20 [91%] women; mean [SD] age, 40.9 [10.3] years; 15 [68%] with clinical allergy history), 17 (77%) met Brighton anaphylaxis criteria. All reactions fully resolved. Of patients who underwent skin prick tests, 0 of 11 tested positive to PEG, 0 of 11 tested positive to P80, and 1 of 10 (10%) tested positive to the same brand of mRNA vaccine used to vaccinate that individual. Among these same participants, 10 of 11 (91%) had positive basophil activation test results to PEG and 11 of 11 (100%) had positive basophil activation test results to their administered mRNA vaccine. No PEG IgE was detected; instead, PEG IgG was found in tested individuals who had an allergy to the vaccine. CONCLUSIONS AND RELEVANCE Based on this case series, women and those with a history of allergic reactions appear at have an elevated risk of mRNA vaccine allergy. Immunological testing suggests non-IgE-mediated immune responses to PEG may be responsible in most individuals.
Collapse
Affiliation(s)
- Christopher Michael Warren
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
- Center for Food Allergy and Asthma Research, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Theo Thomas Snow
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
| | - Alexandra S. Lee
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
| | - Mihir Mukesh Shah
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
| | - Anja Heider
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | - Andra Blomkalns
- Department of Emergency Medicine, Stanford University School of Medicine, Palo Alto, California
| | | | | | - Joseph Gonzalez
- Stanford University School of Medicine, Stanford, California
| | - R. Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Stanford, California
| | - Evan Do
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
| | - Iris Chang
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
| | - Diane Dunham
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
| | - Grace Lee
- Stanford Children’s Health and Stanford University School of Medicine, Department of Pediatrics, Palo Alto, California
| | - Ruth O’Hara
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California
| | - Helen Park
- VA Palo Alto Health Care System, Palo Alto, California
| | - Mohamed H. Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | | | - Sayantani B. Sindher
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Stanford, California
| | | | - Eric Smith
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
| | - Mindy Tsai
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Stephen J. Galli
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
- Department of Pathology, Stanford University School of Medicine, Stanford, California
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California
| | - Cezmi Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | - Kari C. Nadeau
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Stanford, California
| |
Collapse
|
315
|
Yu AM, Tu MJ. Deliver the promise: RNAs as a new class of molecular entities for therapy and vaccination. Pharmacol Ther 2021; 230:107967. [PMID: 34403681 PMCID: PMC9477512 DOI: 10.1016/j.pharmthera.2021.107967] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/06/2021] [Accepted: 07/13/2021] [Indexed: 12/19/2022]
Abstract
The concepts of developing RNAs as new molecular entities for therapies have arisen again and again since the discoveries of antisense RNAs, direct RNA-protein interactions, functional noncoding RNAs, and RNA-directed gene editing. The feasibility was demonstrated with the development and utilization of synthetic RNA agents to selectively control target gene expression, modulate protein functions or alter the genome to manage diseases. Rather, RNAs are labile to degradation and cannot cross cell membrane barriers, making it hard to develop RNA medications. With the development of viable RNA technologies, such as chemistry and pharmaceutics, eight antisense oligonucleotides (ASOs) (fomivirsen, mipomersen, eteplirsen, nusinersen, inotersen, golodirsen, viltolarsen and casimersen), one aptamer (pegaptanib), and three small interfering RNAs (siRNAs) (patisiran, givosiran and lumasiran) have been approved by the United States Food and Drug Administration (FDA) for therapies, and two mRNA vaccines (BNT162b2 and mRNA-1273) under Emergency Use Authorization for the prevention of COVID-19. Therefore, RNAs have become a great addition to small molecules, proteins/antibodies, and cell-based modalities to improve the public health. In this article, we first summarize the general characteristics of therapeutic RNA agents, including chemistry, common delivery strategies, mechanisms of actions, and safety. By overviewing individual RNA medications and vaccines approved by the FDA and some agents under development, we illustrate the unique compositions and pharmacological actions of RNA products. A new era of RNA research and development will likely lead to commercialization of more RNA agents for medical use, expanding the range of therapeutic targets and increasing the diversity of molecular modalities.
Collapse
Affiliation(s)
- Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA.
| | - Mei-Juan Tu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
316
|
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: 20] [Impact Index Per Article: 5.0] [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
|
317
|
Advances in amphiphilic polylactide/vinyl polymer based nano-assemblies for drug delivery. Adv Colloid Interface Sci 2021; 294:102483. [PMID: 34274723 DOI: 10.1016/j.cis.2021.102483] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/28/2021] [Accepted: 07/02/2021] [Indexed: 01/14/2023]
Abstract
Micelles from self-assembled amphiphilic copolymers are highly attractive in drug delivery, due to their small size and hydrophilic stealth corona allowing prolonged lifetimes in the bloodstream and thus improved drug bioavailability. Polylactide (PLA)-based amphiphilic copolymer micelles are key candidates in this field, owing to the well-established biodegradability and biocompatibility of PLA. While PLA-b-poly(ethylene glycol) (PEG) block copolymer micelles can be seen as the "gold standard" in drug delivery research so far, the progresses in controlled radical polymerizations (Atom Transfer Radical Polymerization, Reversible Addition-Fragmentation Transfer and Nitroxide Mediated Polymerization) have offered new opportunities in the design of advanced amphiphilic copolymers for drug delivery due to their flexibility in many regards: (i) they can be easily combined with ring-opening polymerization (ROP) of lactide, with a diversity in types of architectures (e.g., block, graft, star), (ii) they allow (co)polymerization of a wide range of vinyl monomers, possibly circumventing PEG limitations, (iii) functionalization (with biomolecules or stimuli-cleavable moieties) is versatile due to end-group fidelity and copolymerization ability with reactive/functional comonomers. In this review, we report on the advances in the past decade of such amphiphilic PLA/vinyl polymer based nano-carriers, regarding key properties such as stealth character, cell targeting and stimuli-responsiveness.
Collapse
|
318
|
Caballero ML, Krantz MS, Quirce S, Phillips EJ, Stone CA. Hidden Dangers: Recognizing Excipients as Potential Causes of Drug and Vaccine Hypersensitivity Reactions. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2021; 9:2968-2982. [PMID: 33737254 PMCID: PMC8355062 DOI: 10.1016/j.jaip.2021.03.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 12/18/2022]
Abstract
Excipients are necessary as a support to the active ingredients in drugs, vaccines, and other products, and they contribute to their stability, preservation, pharmacokinetics, bioavailability, appearance, and acceptability. For both drugs and vaccines, these are rare reactions; however, for vaccines, they are the primary cause of immediate hypersensitivity. Suspicion for these "hidden dangers" should be high, in particular, when anaphylaxis has occurred in association with multiple chemically distinct drugs. Common excipients implicated include gelatin, carboxymethylcellulose, polyethylene glycols, and products related to polyethylene glycols in immediate hypersensitivity reactions and propylene glycol in delayed hypersensitivity reactions. Complete evaluation of a suspected excipient reaction requires detailed information from the product monograph and package insert to identify all ingredients that are present and to understand the function and structure for these chemicals. This knowledge helps develop a management plan that may include allergy testing to identify the implicated component and to give patients detailed information for future avoidance of relevant foods, drugs, and vaccines. Excipient reactions should be particularly considered for specific classes of drugs where they have been commonly found to be the culprit (eg, corticosteroids, injectable hormones, immunotherapies, monoclonal antibodies, and vaccines). We provide a review of the evidence-based literature outlining epidemiology and mechanisms of excipient reactions and provide strategies for heightened recognition and allergy testing.
Collapse
Affiliation(s)
| | - Matthew S Krantz
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn
| | - Santiago Quirce
- Department of Allergy, La Paz University Hospital, IdiPAZ, Madrid, Spain; Department of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Elizabeth J Phillips
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn; Department of Pharmacology, Vanderbilt School of Medicine, Nashville, Tenn; Institute of Immunology and Infectious Diseases, Murdoch University, Murdoch, Australia
| | - Cosby A Stone
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn.
| |
Collapse
|
319
|
Ndeupen S, Qin Z, Jacobsen S, Estanbouli H, Bouteau A, Igyártó BZ. The mRNA-LNP platform's lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.03.04.430128. [PMID: 33688649 PMCID: PMC7941620 DOI: 10.1101/2021.03.04.430128] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Vaccines based on mRNA-containing lipid nanoparticles (LNPs) are a promising new platform used by two leading vaccines against coronavirus disease in 2019 (COVID-19). Clinical trials and ongoing vaccinations present with very high protection levels and varying degrees of side effects. However, the nature of the reported side effects remains poorly defined. Here we present evidence that LNPs used in many preclinical studies are highly inflammatory in mice. Intradermal injection of these LNPs led to rapid and robust inflammatory responses, characterized by massive neutrophil infiltration, activation of diverse inflammatory pathways, and production of various inflammatory cytokines and chemokines. The same dose of LNP delivered intranasally led to similar inflammatory responses in the lung and resulted in a high mortality rate. In summary, here we show that the LNPs used for many preclinical studies are highly inflammatory. Thus, their potent adjuvant activity and reported superiority comparing to other adjuvants in supporting the induction of adaptive immune responses likely stem from their inflammatory nature. Furthermore, the preclinical LNPs are similar to the ones used for human vaccines, which could also explain the observed side effects in humans using this platform.
Collapse
Affiliation(s)
- Sonia Ndeupen
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, PA
| | - Zhen Qin
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, PA
| | - Sonya Jacobsen
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, PA
| | - Henri Estanbouli
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, PA
| | - Aurélie Bouteau
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, PA
| | - Botond Z. Igyártó
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, PA
- Address correspondence to: Botond Z. Igyártó,
| |
Collapse
|
320
|
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: 1.5] [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
|
321
|
Gessner I. Optimizing nanoparticle design and surface modification toward clinical translation. MRS BULLETIN 2021; 46:643-649. [PMID: 34305307 PMCID: PMC8279028 DOI: 10.1557/s43577-021-00132-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 05/14/2023]
Abstract
The field of nanomedicine is a rapidly evolving field driven by the need for safer and more efficient therapies as well as ultrasensitive and fast diagnostics. Although the advantages of nanoparticles for diagnostic and therapeutic applications are unambiguous, in vivo requirements, including low toxicity, long blood circulation time, proper clearance, sufficient stability, and reproducible synthesis have, in most cases, bedeviled their clinical translation. Nevertheless, researchers have the opportunity to have a decisive influence on the future of nanomedicine by developing new multifunctional molecules and adapting the material design to the requirements. Ultimately, the goal is to find the right level of functionality without adding unnecessary complexity to the system. This article aims to emphasize the potential and current challenges of nanoparticle-based medical agents and highlights how smart and functional material design considerations can help to overcome many of the current limitations and increase the clinical value of nanoparticles.
Collapse
Affiliation(s)
- Isabel Gessner
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
322
|
Edler C, Klein A, Schröder AS, Sperhake JP, Ondruschka B. Deaths associated with newly launched SARS-CoV-2 vaccination (Comirnaty®). Leg Med (Tokyo) 2021; 51:101895. [PMID: 33895650 PMCID: PMC8052499 DOI: 10.1016/j.legalmed.2021.101895] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/09/2021] [Accepted: 04/11/2021] [Indexed: 12/15/2022]
Abstract
Since 27th December 2020, a mRNA vaccine from BioNTech / Pfizer (Comirnaty®) has been used across Germany. As of 12th March 2021, 286 fatalities of vaccinated German individuals were registered at the Paul-Ehrlich-Institute with time intervals after vaccination between one hour to 40 days. From our catchment area in northern Germany, we have so far become aware of 22 deaths in connection with vaccination in a 5 week period (range: 0-28 days after vaccination). Three death cases after vaccination with Comirnaty®, which were autopsied at the Institute of Legal Medicine Hamburg, are presented in more detail. All three deceased had severe cardiovascular diseases, among other comorbidities, and died in the context of these pre-existing conditions, while one case developed a COVID-19 pneumonia as cause of death. Taking into account the results of the postmortem examination a causal relation between the vaccination and the death was not established in any case. If there are indications of an allergic reaction, histological and postmortem laboratory examinations should be performed subsequent to the autopsy (tryptase, total IgE, CRP, interleukin-6, complement activity C3/C5).
Collapse
Affiliation(s)
- Carolin Edler
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, 22529 Hamburg, Germany.
| | - Anke Klein
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, 22529 Hamburg, Germany
| | - Ann Sophie Schröder
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, 22529 Hamburg, Germany
| | - Jan-Peter Sperhake
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, 22529 Hamburg, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, 22529 Hamburg, Germany
| |
Collapse
|
323
|
Brunzel M, Dirauf M, Sahn M, Czaplewska JA, Fritz N, Weber C, Nischang I, Schubert US. On the identification and quantification of proton-initiated species in the synthesis of poly(2-alkyl-2-oxazoline)s by high resolution liquid chromatography. J Chromatogr A 2021; 1653:462364. [PMID: 34280792 DOI: 10.1016/j.chroma.2021.462364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 12/01/2022]
Abstract
Hydrophilic poly(2-oxazoline)s represent a promising alternative to replace poly(ethylene glycol) in the biomedical field. For that purpose, reliable analytical protocols to confirm identity and quantity of impurities are required. In particular, side products deriving from chain transfer reactions occurring during the cationic ring-opening polymerization and incomplete end-capping processes may be present. The analytical approach must hence be capable of separating polymers according to minor changes regarding their end group. We demonstrate that liquid chromatography, relying on a monolithic C18-modified silica column and isocratic as well as gradient elution using water / acetonitrile mixtures and varying detectors, can accomplish such demanding high resolution separations. Poly(2-ethyl-2-oxazoline)s (PEtOx) with acetyl, hydroxyl, and phthalimide ω-end groups were investigated. Identification of side products was achieved through coupling with electrospray ionization mass spectrometry. UV / Vis detection was applied to quantify chain transfer products in PEtOx comprising biphenyl moieties. In addition, gradient elution enabled the separation of PEtOx into macromolecules according to their specific degrees of polymerization in molar mass ranges around 2,000 g mol-1.
Collapse
Affiliation(s)
- Michaela Brunzel
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstr. 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Michael Dirauf
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstr. 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Martin Sahn
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstr. 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Justyna A Czaplewska
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstr. 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Nicole Fritz
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstr. 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Christine Weber
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstr. 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Ivo Nischang
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstr. 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany.
| | - Ulrich S Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstr. 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany.
| |
Collapse
|
324
|
Martin JD, Miyazaki T, Cabral H. Remodeling tumor microenvironment with nanomedicines. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1730. [PMID: 34124849 DOI: 10.1002/wnan.1730] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/17/2022]
Abstract
The tumor microenvironment (TME) has been recognized as a major contributor to cancer malignancy and therapeutic resistance. Thus, strategies directed to re-engineer the TME are emerging as promising approaches for improving the efficacy of antitumor therapies by enhancing tumor perfusion and drug delivery, as well as alleviating the immunosuppressive TME. In this regard, nanomedicine has shown great potential for developing effective treatments capable of re-modeling the TME by controlling drug action in a spatiotemporal manner and allowing long-lasting modulatory effects on the TME. Herein, we review recent progress on TME re-engineering by using nanomedicine, particularly focusing on formulations controlling TME characteristics through targeted interaction with cellular components of the TME. Importantly, the TME should be re-engineering to a quiescent phenotype rather than be destroyed. Finally, immediate challenges and future perspectives of TME-re-engineering nanomedicines are discussed, anticipating further innovation in this growing field. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
| | - Takuya Miyazaki
- Kanagawa Institute of Industrial Science and Technology, Ebina, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
325
|
AlSawaftah N, Pitt WG, Husseini GA. Dual-Targeting and Stimuli-Triggered Liposomal Drug Delivery in Cancer Treatment. ACS Pharmacol Transl Sci 2021; 4:1028-1049. [PMID: 34151199 PMCID: PMC8205246 DOI: 10.1021/acsptsci.1c00066] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Indexed: 12/31/2022]
Abstract
The delivery of chemotherapeutics to solid tumors using smart drug delivery systems (SDDSs) takes advantage of the unique physiology of tumors (i.e., disordered structure, leaky vasculature, abnormal extracellular matrix (ECM), and limited lymphatic drainage) to deliver anticancer drugs with reduced systemic side effects. Liposomes are the most promising of such SDDSs and have been well investigated for cancer therapy. To improve the specificity, bioavailability, and anticancer efficacy of liposomes at the diseased sites, other strategies such as targeting ligands and stimulus-sensitive liposomes have been developed. This review highlights relevant surface functionalization techniques and stimuli-mediated drug release for enhanced delivery of anticancer agents at tumor sites, with a special focus on dual functionalization and design of multistimuli responsive liposomes.
Collapse
Affiliation(s)
- Nour AlSawaftah
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, UAE
| | - William G. Pitt
- Chemical
Engineering Department, Brigham Young University, Provo, Utah 84602, United States
| | - Ghaleb A. Husseini
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, UAE
| |
Collapse
|
326
|
Miyazawa T, Itaya M, Burdeos GC, Nakagawa K, Miyazawa T. A Critical Review of the Use of Surfactant-Coated Nanoparticles in Nanomedicine and Food Nanotechnology. Int J Nanomedicine 2021; 16:3937-3999. [PMID: 34140768 PMCID: PMC8203100 DOI: 10.2147/ijn.s298606] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Surfactants, whose existence has been recognized as early as 2800 BC, have had a long history with the development of human civilization. With the rapid development of nanotechnology in the latter half of the 20th century, breakthroughs in nanomedicine and food nanotechnology using nanoparticles have been remarkable, and new applications have been developed. The technology of surfactant-coated nanoparticles, which provides new functions to nanoparticles for use in the fields of nanomedicine and food nanotechnology, is attracting a lot of attention in the fields of basic research and industry. This review systematically describes these "surfactant-coated nanoparticles" through various sections in order: 1) surfactants, 2) surfactant-coated nanoparticles, application of surfactant-coated nanoparticles to 3) nanomedicine, and 4) food nanotechnology. Furthermore, current progress and problems of the technology using surfactant-coated nanoparticles through recent research reports have been discussed.
Collapse
Affiliation(s)
- Taiki Miyazawa
- New Industry Creation Hatchery Center (NICHe), Tohoku University, Sendai, Miyagi, Japan
| | - Mayuko Itaya
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Gregor C Burdeos
- Institute for Animal Nutrition and Physiology, Christian Albrechts University Kiel, Kiel, Germany
| | - Kiyotaka Nakagawa
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Teruo Miyazawa
- New Industry Creation Hatchery Center (NICHe), Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
327
|
Crețu BEB, Dodi G, Shavandi A, Gardikiotis I, Șerban IL, Balan V. Imaging Constructs: The Rise of Iron Oxide Nanoparticles. Molecules 2021; 26:3437. [PMID: 34198906 PMCID: PMC8201099 DOI: 10.3390/molecules26113437] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, an important challenge in nanomedicine imaging has been the work to design multifunctional agents that can be detected by single and/or multimodal techniques. Among the broad spectrum of nanoscale materials being investigated for imaging use, iron oxide nanoparticles have gained significant attention due to their intrinsic magnetic properties, low toxicity, large magnetic moments, superparamagnetic behaviour and large surface area-the latter being a particular advantage in its conjunction with specific moieties, dye molecules, and imaging probes. Tracers-based nanoparticles are promising candidates, since they combine synergistic advantages for non-invasive, highly sensitive, high-resolution, and quantitative imaging on different modalities. This study represents an overview of current advancements in magnetic materials with clinical potential that will hopefully provide an effective system for diagnosis in the near future. Further exploration is still needed to reveal their potential as promising candidates from simple functionalization of metal oxide nanomaterials up to medical imaging.
Collapse
Affiliation(s)
- Bianca Elena-Beatrice Crețu
- Advanced Centre for Research-Development in Experimental Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.E.-B.C.); (I.G.)
| | - Gianina Dodi
- Advanced Centre for Research-Development in Experimental Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.E.-B.C.); (I.G.)
| | - Amin Shavandi
- BioMatter-Biomass Transformation Lab, École Polytechnique de Bruxelles, Université Libre de Bruxelles, 1050 Brussels, Belgium;
| | - Ioannis Gardikiotis
- Advanced Centre for Research-Development in Experimental Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.E.-B.C.); (I.G.)
| | - Ionela Lăcrămioara Șerban
- Physiology Department, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania;
| | - Vera Balan
- Faculty of Medical Bioengineering, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania;
| |
Collapse
|
328
|
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: 110] [Impact Index Per Article: 27.5] [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
|
329
|
Cabanillas B, Akdis CA, Novak N. COVID-19 vaccine anaphylaxis: IgE, complement or what else? A reply to: "COVID-19 vaccine anaphylaxis: PEG or not?". Allergy 2021; 76:1938-1940. [PMID: 34128561 PMCID: PMC8441702 DOI: 10.1111/all.14725] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 01/31/2023]
Affiliation(s)
- Beatriz Cabanillas
- Department of Allergy Research Institute Hospital Doce de Octubre Madrid Spain
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
| | - Natalija Novak
- Department of Dermatology and Allergy University Hospital Bonn Bonn Germany
| |
Collapse
|
330
|
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: 33] [Impact Index Per Article: 8.3] [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
|
331
|
Igyártó BZ, Jacobsen S, Ndeupen S. Future considerations for the mRNA-lipid nanoparticle vaccine platform. Curr Opin Virol 2021; 48:65-72. [PMID: 33906124 PMCID: PMC8065267 DOI: 10.1016/j.coviro.2021.03.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 12/16/2022]
Abstract
Vaccines based on mRNA-containing lipid nanoparticles (LNPs) pioneered by Katalin Karikó and Drew Weissman at the University of Pennsylvania are a promising new vaccine platform used by two of the leading vaccines against coronavirus disease in 2019 (COVID-19). However, there are many questions regarding their mechanism of action in humans that remain unanswered. Here we consider the immunological features of LNP components and off-target effects of the mRNA, both of which could increase the risk of side effects. We suggest ways to mitigate these potential risks by harnessing dendritic cell (DC) biology.
Collapse
Affiliation(s)
- Botond Z Igyártó
- Thomas Jefferson University, Department of Microbiology and Immunology, 233 South 10th Street, Philadelphia, PA 19107, United States.
| | - Sonya Jacobsen
- Thomas Jefferson University, Department of Microbiology and Immunology, 233 South 10th Street, Philadelphia, PA 19107, United States
| | - Sonia Ndeupen
- Thomas Jefferson University, Department of Microbiology and Immunology, 233 South 10th Street, Philadelphia, PA 19107, United States
| |
Collapse
|
332
|
Sampath V, Rabinowitz G, Shah M, Jain S, Diamant Z, Jesenak M, Rabin R, Vieths S, Agache I, Akdis M, Barber D, Breiteneder H, Chinthrajah S, Chivato T, Collins W, Eiwegger T, Fast K, Fokkens W, O'Hehir RE, Ollert M, O'Mahony L, Palomares O, Pfaar O, Riggioni C, Shamji MH, Sokolowska M, Jose Torres M, Traidl-Hoffmann C, van Zelm M, Wang DY, Zhang L, Akdis CA, Nadeau KC. Vaccines and allergic reactions: The past, the current COVID-19 pandemic, and future perspectives. Allergy 2021; 76:1640-1660. [PMID: 33811364 PMCID: PMC8251022 DOI: 10.1111/all.14840] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 12/11/2022]
Abstract
Vaccines are essential public health tools with a favorable safety profile and prophylactic effectiveness that have historically played significant roles in reducing infectious disease burden in populations, when the majority of individuals are vaccinated. The COVID-19 vaccines are expected to have similar positive impacts on health across the globe. While serious allergic reactions to vaccines are rare, their underlying mechanisms and implications for clinical management should be considered to provide individuals with the safest care possible. In this review, we provide an overview of different types of allergic adverse reactions that can potentially occur after vaccination and individual vaccine components capable of causing the allergic adverse reactions. We present the incidence of allergic adverse reactions during clinical studies and through post-authorization and post-marketing surveillance and provide plausible causes of these reactions based on potential allergenic components present in several common vaccines. Additionally, we review implications for individual diagnosis and management and vaccine manufacturing overall. Finally, we suggest areas for future research.
Collapse
Affiliation(s)
- Vanitha Sampath
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
| | - Grace Rabinowitz
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
| | - Mihir Shah
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
| | - Surabhi Jain
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
| | - Zuzana Diamant
- Departmentt of Microbiology Immunology & Transplantation, KU Leuven, Catholic University of Leuven, Leuven, Belgium
- Department of Respiratory Medicine & Allergology, Institute for Clinical Science, Skane University Hospital, Lund University, Lund, Sweden
- Department of Clinical Pharmacy &Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Milos Jesenak
- Department of Pediatrics and Department of Clinical Immunology and Allergology, Jessenius Faculty of Medicine in Martin, Center for Vaccination in Special Situations, University Hospital in Martin, Comenius University in Bratislava, Bratislava, Slovakia
| | - Ronald Rabin
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Stefan Vieths
- Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | | | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Zurich, Switzerland
| | - Domingo Barber
- Departamento de CienciasMédicasBásicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, España
- Instituto de Salud Carlos III, RETIC ARADYAL, Madrid, Spain
| | - Heimo Breiteneder
- Division of Medical Biotechnology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Tomas Chivato
- School of Medicine, University CEU San Pablo, Madrid, Spain
| | - William Collins
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Hospital Medicine, Stanford University, Stanford, CA, USA
| | - Thomas Eiwegger
- Division of Immunology and Allergy, Food Allergy and Anaphylaxis Program, The Hospital for Sick Children, Toronto, ON, Canada
- Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Katharine Fast
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
| | - Wytske Fokkens
- Department of Otorhinolaryngology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Robyn E O'Hehir
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Monash University, and Alfred Health, Melbourne, Vic, Australia
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Carmen Riggioni
- Department of Paediatrics, Allergy and Clinical Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Mohamed H Shamji
- Department of National Heart and Lung Institute, Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Imperial College London, London, UK
- Centre in Allergic Mechanisms of Asthma, London, UK
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Zurich, Switzerland
| | - Maria Jose Torres
- Allergy Unit, Malaga Regional University Hospital-UMA-ARADyAL, Málaga, Spain
| | - Claudia Traidl-Hoffmann
- Department of Environmental Medicine, Medical Faculty, University Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany
| | - Menno van Zelm
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
- Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, VIC, Australia
| | - De Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Zurich, Switzerland
| | - Kari C Nadeau
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
333
|
Non-Immunotherapy Application of LNP-mRNA: Maximizing Efficacy and Safety. Biomedicines 2021; 9:biomedicines9050530. [PMID: 34068715 PMCID: PMC8151051 DOI: 10.3390/biomedicines9050530] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
Lipid nanoparticle (LNP) formulated messenger RNA-based (LNP-mRNA) vaccines came into the spotlight as the first vaccines against SARS-CoV-2 virus to be applied worldwide. Long-known benefits of mRNA-based technologies consisting of relatively simple and fast engineering of mRNA encoding for antigens and proteins of interest, no genomic integration, and fast and efficient manufacturing process compared with other biologics have been verified, thus establishing a basis for a broad range of applications. The intrinsic immunogenicity of LNP formulated in vitro transcribed (IVT) mRNA is beneficial to the LNP-mRNA vaccines. However, avoiding immune activation is critical for therapeutic applications of LNP-mRNA for protein replacement where targeted mRNA expression and repetitive administration of high doses for a lifetime are required. This review summarizes our current understanding of immune activation induced by mRNA, IVT byproducts, and LNP. It gives a comprehensive overview of the present status of preclinical and clinical studies in which LNP-mRNA is used for protein replacement and treatment of rare diseases with an emphasis on safety. Moreover, the review outlines innovations and strategies to advance pharmacology and safety of LNP-mRNA for non-immunotherapy applications.
Collapse
|
334
|
Cui X, Song K, Lu X, Feng W, Di W. Liposomal Delivery of MicroRNA-7 Targeting EGFR to Inhibit the Growth, Invasion, and Migration of Ovarian Cancer. ACS OMEGA 2021; 6:11669-11678. [PMID: 34056322 PMCID: PMC8153987 DOI: 10.1021/acsomega.1c00992] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/06/2021] [Indexed: 05/03/2023]
Abstract
Ovarian cancer is highly aggressive and has high rates of recurrence and metastasis. Due to the limited effects of current treatments, it is necessary to conduct research and develop new treatment options. The application of gene therapy in tumor therapy is gradually increasing and has exciting prospects. MicroRNA-7 (miR-7) has been reported to inhibit the growth, invasion, and metastasis of a variety of solid tumors. Cationic liposomes are safe and effective gene delivery systems for transfection in vivo and in vitro. To realize the application of miR-7 in the treatment of ovarian cancer, cationic liposomes were prepared with 1,2-dioleoyl-3-trimethylammonium-propane, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine, and cholesterol. The miR-7 liposomes had a suitable particle size, potential, and a high cellular uptake rate. MiR-7 encapsulated by liposomes could be effectively delivered to ovarian cancer cells and successfully targeted to the tumor site in a mouse xenograft model of ovarian cancer. In vitro and in vivo experiments revealed that the miR-7 liposomes had a significant ability to inhibit the growth, invasion, and migration of ovarian cancer, probably by inhibiting the expression of the epidermal growth factor receptor. Our studies of miR-7 liposomes demonstrated a safe and efficient microRNA delivery system for the gene therapy of ovarian cancer.
Collapse
Affiliation(s)
- Xiaojuan Cui
- Department
of Obstetrics and Gynecology, Key Laboratory of Gynecologic Oncology,
and State Key Laboratory of Oncogenes and Related Genes, Shanghai
Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Department
of Obstetrics and Gynecology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Keqi Song
- Department
of Obstetrics and Gynecology, Key Laboratory of Gynecologic Oncology,
and State Key Laboratory of Oncogenes and Related Genes, Shanghai
Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaolan Lu
- Department
of Obstetrics and Gynecology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Weiwei Feng
- Department
of Obstetrics and Gynecology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
- . Phone: +86-21-64370045. Fax: +86-21-64370045
| | - Wen Di
- Department
of Obstetrics and Gynecology, Key Laboratory of Gynecologic Oncology,
and State Key Laboratory of Oncogenes and Related Genes, Shanghai
Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- . Phone: +86-21-68383829. Fax: +86-21-68383829
| |
Collapse
|
335
|
Ashford MB, England RM, Akhtar N. Highway to Success—Developing Advanced Polymer Therapeutics. ADVANCED THERAPEUTICS 2021; 4. [DOI: 10.1002/adtp.202000285] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Indexed: 01/06/2025]
Abstract
AbstractPolymer therapeutics are advancing as an important class of drugs. Polymers have already demonstrated their value in extending the half‐life of proteins. They show great potential as delivery systems for improving the therapeutic index of drugs, via biophysical targeting and more recently with more precision targeting. They are also important for intracellular delivery of nucleic acid based drugs. The same frameworks that have been successfully applied to improve the small molecule drug development can be adopted. This approach together with improved pathophysiological disease knowledge and critical developability considerations, imperative given the size and complexity of polymer therapeutics, provides a structured framework that should improve their clinical translation and exploit their functionality and potential. Progress in understanding the right target, gaining the right tissue and cell exposure, ensuring the right safety, selecting the right patient population is discussed. The right commercial considerations are outlined and the need for a multi‐disciplinary approach is emphasized. Crucial developability factors together with scientific and technical advancements to enable pharmaceutical development of a quality robust product are addressed. It is argued that by applying this structured approach to their design and development, polymer therapeutics will continue to grow and develop as important next generation medicines.
Collapse
Affiliation(s)
- Marianne B. Ashford
- Advanced Drug Delivery Pharmaceutical Sciences, R&D, AstraZeneca Macclesfield SK10 2NA UK
| | - Richard M. England
- Advanced Drug Delivery Pharmaceutical Sciences, R&D, AstraZeneca Macclesfield SK10 2NA UK
| | - Nadim Akhtar
- New Modalities & Parenteral Development Pharmaceutical Technology & Development, Operations, AstraZeneca Macclesfield SK10 2NA UK
| |
Collapse
|
336
|
Kraft M, Renaudin JM, Ensina LF, Kleinheinz A, Bilò MB, Scherer Hofmeier K, Dölle-Bierke S, Worm M. Anaphylaxis to vaccination and polyethylene glycol: a perspective from the European Anaphylaxis Registry. J Eur Acad Dermatol Venereol 2021; 35:e659-e662. [PMID: 33914977 PMCID: PMC8242778 DOI: 10.1111/jdv.17327] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- M Kraft
- Division of Allergy and Immunology, Department of Dermatology, Venerology and Allergology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Emergency Medicine, University Hospital Halle (Saale), Medical Faculty of Martin Luther University Halle-Wittenberg, Halle, Germany
| | - J M Renaudin
- Presidency on behalf of Allergy Vigilance Network, Vandoeuvre les Nancy, France
| | - L F Ensina
- Division of Allergy, Clinical Immunology and Rheumatology, Department of Pediatrics, Federal University of São Paulo, São Paulo, Brazil
| | - A Kleinheinz
- Department of Dermatology, Elbe Medical Centre, Buxtehude, Germany
| | - M B Bilò
- Department of Clinical and Molecular Sciences, Marche Polytechnic University - Allergy Unit, University Hospital Ospedali Riuniti di Ancona, Ancona, Italy
| | - K Scherer Hofmeier
- Division of Allergy, Department of Dermatology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - S Dölle-Bierke
- Division of Allergy and Immunology, Department of Dermatology, Venerology and Allergology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - M Worm
- Division of Allergy and Immunology, Department of Dermatology, Venerology and Allergology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
337
|
Frickenstein AN, Hagood JM, Britten CN, Abbott BS, McNally MW, Vopat CA, Patterson EG, MacCuaig WM, Jain A, Walters KB, McNally LR. Mesoporous Silica Nanoparticles: Properties and Strategies for Enhancing Clinical Effect. Pharmaceutics 2021; 13:570. [PMID: 33920503 PMCID: PMC8072651 DOI: 10.3390/pharmaceutics13040570] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/15/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Due to the theragnostic potential of mesoporous silica nanoparticles (MSNs), these were extensively investigated as a novel approach to improve clinical outcomes. Boasting an impressive array of formulations and modifications, MSNs demonstrate significant in vivo efficacy when used to identify or treat myriad malignant diseases in preclinical models. As MSNs continue transitioning into clinical trials, a thorough understanding of the characteristics of effective MSNs is necessary. This review highlights recent discoveries and advances in MSN understanding and technology. Specific focus is given to cancer theragnostic approaches using MSNs. Characteristics of MSNs such as size, shape, and surface properties are discussed in relation to effective nanomedicine practice and projected clinical efficacy. Additionally, tumor-targeting options used with MSNs are presented with extensive discussion on active-targeting molecules. Methods for decreasing MSN toxicity, improving site-specific delivery, and controlling release of loaded molecules are further explained. Challenges facing the field and translation to clinical environments are presented alongside potential avenues for continuing investigations.
Collapse
Affiliation(s)
- Alex N. Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA; (A.N.F.); (C.A.V.); (W.M.M.)
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.M.H.); (M.W.M.)
| | - Jordan M. Hagood
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.M.H.); (M.W.M.)
| | - Collin N. Britten
- School of Chemical, Biological, and Materials Engineering, University of Oklahoma, Norman, OK 73019, USA; (C.N.B.); (B.S.A.); (K.B.W.)
| | - Brandon S. Abbott
- School of Chemical, Biological, and Materials Engineering, University of Oklahoma, Norman, OK 73019, USA; (C.N.B.); (B.S.A.); (K.B.W.)
| | - Molly W. McNally
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.M.H.); (M.W.M.)
| | - Catherine A. Vopat
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA; (A.N.F.); (C.A.V.); (W.M.M.)
| | - Eian G. Patterson
- Department of Biology, University of Oklahoma, Norman, OK 73019, USA;
| | - William M. MacCuaig
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA; (A.N.F.); (C.A.V.); (W.M.M.)
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.M.H.); (M.W.M.)
| | - Ajay Jain
- Department of Surgery, University of Oklahoma, Oklahoma City, OK 73104, USA;
| | - Keisha B. Walters
- School of Chemical, Biological, and Materials Engineering, University of Oklahoma, Norman, OK 73019, USA; (C.N.B.); (B.S.A.); (K.B.W.)
| | - Lacey R. McNally
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.M.H.); (M.W.M.)
- Department of Surgery, University of Oklahoma, Oklahoma City, OK 73104, USA;
| |
Collapse
|
338
|
Fu D, Wang Z, Tu Y, Peng F. Interactions between Biomedical Micro-/Nano-Motors and the Immune Molecules, Immune Cells, and the Immune System: Challenges and Opportunities. Adv Healthc Mater 2021; 10:e2001788. [PMID: 33506650 DOI: 10.1002/adhm.202001788] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/21/2020] [Indexed: 12/16/2022]
Abstract
Mobile micro- and nano-motors (MNMs) emerge as revolutionary platforms for biomedical applications, including drug delivery, biosensing, non-invasive surgery, and cancer therapy. While for applications in biomedical fields and practical clinical translation, the interactions of these untethered tiny machines with the immune system is an essential issue to be considered. This review highlights the recent approaches of surpassing immune barriers to prevent foreign motors from triggering immune responses. In addition to trials focusing on the function preservation of MNMs, examples of versatile MNMs working with the immune components (immune molecules, immune cells and the whole system) to achieve cancer immunotherapy, immunoassay, and detoxification are outlined. The immune interference part provides researchers an idea about what is the limit presented by the immune components. The coworking part suggests ways to bypass or even utilize the limit. With interdisciplinary cooperation of nanoengineering, materials science, and immunology field, the rationally designed functional MNMs are expected to provide novel opportunities for the biomedical field.
Collapse
Affiliation(s)
- Dongmei Fu
- School of Materials Science and Engineering Sun‐Yat‐sen University Guangzhou 510275 China
| | - Zhen Wang
- School of Materials Science and Engineering Sun‐Yat‐sen University Guangzhou 510275 China
| | - Yingfeng Tu
- School of Pharmaceutical Science Southern Medical University Guangzhou 510515 China
| | - Fei Peng
- School of Materials Science and Engineering Sun‐Yat‐sen University Guangzhou 510275 China
| |
Collapse
|
339
|
Tully M, Dimde M, Weise C, Pouyan P, Licha K, Schirner M, Haag R. Polyglycerol for Half-Life Extension of Proteins-Alternative to PEGylation? Biomacromolecules 2021; 22:1406-1416. [PMID: 33792290 DOI: 10.1021/acs.biomac.0c01627] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since several decades, PEGylation is known to be the clinical standard to enhance pharmacokinetics of biotherapeutics. In this study, we introduce polyglycerol (PG) of different lengths and architectures (linear and hyperbranched) as an alternative polymer platform to poly(ethylene glycol) (PEG) for half-life extension (HLE). We designed site-selective N-terminally modified PG-protein conjugates of the therapeutic protein anakinra (IL-1ra, Kineret) and compared them systematically with PEG analogues of similar molecular weights. Linear PG and PEG conjugates showed comparable hydrodynamic sizes and retained their secondary structure, whereas binding affinity to IL-1 receptor 1 decreased with increasing polymer length, yet remained in the low nanomolar range for all conjugates. The terminal half-life of a 40 kDa linear PG-modified anakinra was extended 4-fold compared to the unmodified protein, close to its PEG analogue. Our results demonstrate similar performances of PEG- and PG-anakinra conjugates and therefore highlight the outstanding potential of polyglycerol as a PEG alternative for half-life extension of biotherapeutics.
Collapse
Affiliation(s)
- Michael Tully
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Mathias Dimde
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Christoph Weise
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Paria Pouyan
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Kai Licha
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Michael Schirner
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Rainer Haag
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| |
Collapse
|
340
|
Taguchi K, Okamoto Y, Matsumoto K, Otagiri M, Chuang VTG. When Albumin Meets Liposomes: A Feasible Drug Carrier for Biomedical Applications. Pharmaceuticals (Basel) 2021; 14:ph14040296. [PMID: 33810483 PMCID: PMC8065628 DOI: 10.3390/ph14040296] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022] Open
Abstract
Albumin, the most abundant protein in plasma, possesses some inherent beneficial structural and physiological characteristics that make it suitable for use as a drug delivery agent, such as an extraordinary drug-binding capacity and long blood retention, with a high biocompatibility. The use of these characteristics as a nanoparticle drug delivery system (DDS) offers several advantages, including a longer circulation time, lower toxicity, and more significant drug loading. To date, many innovative liposome preparations have been developed in which albumin is involved as a DDS. These novel albumin-containing liposome preparations show superior deliverability for genes, hydrophilic/hydrophobic substances and proteins/peptides to the targeting area compared to original liposomes by virtue of their high biocompatibility, stability, effective loading content, and the capacity for targeting. This review summarizes the current status of albumin applications in liposome-based DDS, focusing on albumin-coated liposomes and albumin-encapsulated liposomes as a DDS carrier for potential medical applications.
Collapse
Affiliation(s)
- Kazuaki Taguchi
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; (K.T.); (K.M.)
| | - Yuko Okamoto
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan; (Y.O.); (M.O.)
| | - Kazuaki Matsumoto
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; (K.T.); (K.M.)
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan; (Y.O.); (M.O.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan
| | - Victor Tuan Giam Chuang
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA 6102, Australia
- Correspondence:
| |
Collapse
|
341
|
Briolay T, Petithomme T, Fouet M, Nguyen-Pham N, Blanquart C, Boisgerault N. Delivery of cancer therapies by synthetic and bio-inspired nanovectors. Mol Cancer 2021; 20:55. [PMID: 33761944 PMCID: PMC7987750 DOI: 10.1186/s12943-021-01346-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND As a complement to the clinical development of new anticancer molecules, innovations in therapeutic vectorization aim at solving issues related to tumor specificity and associated toxicities. Nanomedicine is a rapidly evolving field that offers various solutions to increase clinical efficacy and safety. MAIN: Here are presented the recent advances for different types of nanovectors of chemical and biological nature, to identify the best suited for translational research projects. These nanovectors include different types of chemically engineered nanoparticles that now come in many different flavors of 'smart' drug delivery systems. Alternatives with enhanced biocompatibility and a better adaptability to new types of therapeutic molecules are the cell-derived extracellular vesicles and micro-organism-derived oncolytic viruses, virus-like particles and bacterial minicells. In the first part of the review, we describe their main physical, chemical and biological properties and their potential for personalized modifications. The second part focuses on presenting the recent literature on the use of the different families of nanovectors to deliver anticancer molecules for chemotherapy, radiotherapy, nucleic acid-based therapy, modulation of the tumor microenvironment and immunotherapy. CONCLUSION This review will help the readers to better appreciate the complexity of available nanovectors and to identify the most fitting "type" for efficient and specific delivery of diverse anticancer therapies.
Collapse
Affiliation(s)
- Tina Briolay
- Université de Nantes, Inserm, CRCINA, F-44000, Nantes, France
| | | | - Morgane Fouet
- Université de Nantes, Inserm, CRCINA, F-44000, Nantes, France
| | | | | | | |
Collapse
|
342
|
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: 34] [Impact Index Per Article: 8.5] [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
|
343
|
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
|
344
|
Bayerl C, Herbst M. Polyethylenglycol – Freund oder Feind? AKTUELLE DERMATOLOGIE 2021. [DOI: 10.1055/a-1352-6241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
ZusammenfassungAnaphylaktische Reaktionen nach Impfungen sind selten. Zwei Fälle von Reaktionen nach der Pfizer-BioNTech-Covid-19-Vakzine, BNT162b2, wurden aus dem UK berichtet, weitere 19 aus den USA. Als möglicher Auslöser wurde Polyethylenglycol (PEG) angeschuldigt. Pegylierung ist ein üblicher Vorgang in der Impfstoffherstellung, um die Substanzen vor schnellem immunologischem Abbau zu schützen. PEG ist ein Polyether (Synonyme z. B. Laureth-9, Polidocanol, Thesit, Macrogol) und findet sich in Kosmetika, Körperpflegeprodukten, Nahrungsmitteln, Liposomen und Nanopartikeln als Drug-Delivery-Systeme für Medikamente und als osmotisches Laxans vor Gastro-/Koloskopien. IgM-Antikörper auf PEG werden bei 70 % der Bevölkerung nachgewiesen. Bei 2 Fällen von Soforttyp-Reaktionen auf PEG fanden sich IgE-Antiköper, bei anderen Medikamentengaben mit PEG geht man von pseudoallergischen Reaktionen über die Komplement-Aktivierungs-related Pseudoallergie (CARPA) aus. In Relation zum weit verbreiteten Einsatz von PEG sind die Impfreaktionen sehr selten und aktuell kein Grund, Allergiker oder Menschen mit Anaphylaxien bei entsprechenden Vorsichtsmaßnahmen auf bekannte Auslöser auszuschließen. Aufgrund der Meldepflicht (Gesundheitsamt und Arzneimittelkommission der deutschen Ärzteschaft) ist bald mit weiteren Erkenntnissen zu rechnen.
Collapse
Affiliation(s)
- C. Bayerl
- Klinik für Dermatologie und Allergologie, Hauttumorzentrum Wiesbaden, Helios Dr. Horst Schmidt Kliniken, Wiesbaden
| | | |
Collapse
|
345
|
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: 2.0] [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
|
346
|
Clauss ZS, Kramer JR. Design, synthesis and biological applications of glycopolypeptides. Adv Drug Deliv Rev 2021; 169:152-167. [PMID: 33352223 DOI: 10.1016/j.addr.2020.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/12/2020] [Accepted: 12/12/2020] [Indexed: 12/15/2022]
Abstract
Carbohydrates play essential structural and biochemical roles in all living organisms. Glycopolymers are attractive as well-defined biomimetic analogs to study carbohydrate-dependent processes, and are widely applicable biocompatible materials in their own right. Glycopolypeptides have shown great promise in this area since they are closer structural mimics of natural glycoproteins than other synthetic glycopolymers and can serve as carriers for biologically active carbohydrates. This review highlights advances in the area of design and synthesis of such materials, and their biomedical applications in therapeutic delivery, tissue engineering, and beyond.
Collapse
|
347
|
Slor G, Olea AR, Pujals S, Tigrine A, De La Rosa VR, Hoogenboom R, Albertazzi L, Amir RJ. Judging Enzyme-Responsive Micelles by Their Covers: Direct Comparison of Dendritic Amphiphiles with Different Hydrophilic Blocks. Biomacromolecules 2021; 22:1197-1210. [PMID: 33512161 PMCID: PMC7944483 DOI: 10.1021/acs.biomac.0c01708] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Enzymatically
degradable polymeric micelles have great potential
as drug delivery systems, allowing the selective release of their
active cargo at the site of disease. Furthermore, enzymatic degradation
of the polymeric nanocarriers facilitates clearance of the delivery
system after it has completed its task. While extensive research is
dedicated toward the design and study of the enzymatically degradable
hydrophobic block, there is limited understanding on how the hydrophilic
shell of the micelle can affect the properties of such enzymatically
degradable micelles. In this work, we report a systematic head-to-head
comparison of well-defined polymeric micelles with different polymeric
shells and two types of enzymatically degradable hydrophobic cores.
To carry out this direct comparison, we developed a highly modular
approach for preparing clickable, spectrally active enzyme-responsive
dendrons with adjustable degree of hydrophobicity. The dendrons were
linked with three different widely used hydrophilic polymers—poly(ethylene
glycol), poly(2-ethyl-2-oxazoline), and poly(acrylic acid) using the
CuAAC click reaction. The high modularity and molecular precision
of the synthetic methodology enabled us to easily prepare well-defined
amphiphiles that differ either in their hydrophilic block composition
or in their hydrophobic dendron. The micelles of the different amphiphiles
were thoroughly characterized and their sizes, critical micelle concentrations,
drug loading, stability, and cell internalization were compared. We
found that the micelle diameter was almost solely dependent on the
hydrophobicity of the dendritic hydrophobic block, whereas the enzymatic
degradation rate was strongly dependent on the composition of both
blocks. Drug encapsulation capacity was very sensitive to the type
of the hydrophilic block, indicating that, in addition to the hydrophobic
core, the micellar shell also has a significant role in drug encapsulation.
Incubation of the spectrally active micelles in the presence of cells
showed that the hydrophilic shell significantly affects the micellar
stability, localization, cell internalization kinetics, and the cargo
release mechanism. Overall, the high molecular precision and the ability
of these amphiphiles to report their disassembly, even in complex
biological media, allowed us to directly compare the different types
of micelles, providing striking insights into how the composition
of the micelle shells and cores can affect their properties and potential
to serve as nanocarriers.
Collapse
Affiliation(s)
- Gadi Slor
- Department of Organic Chemistry, School of Chemistry, Faculty of Exact Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel.,Tel Aviv University Center for Nanoscience and Nanotechnology, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Alis R Olea
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028 Barcelona, Spain
| | - Sílvia Pujals
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028 Barcelona, Spain.,Department of Electronic and Biomedical Engineering, Faculty of Physics, University of Barcelona, Carrer Martí I Franquès 1, 08028 Barcelona, Spain
| | - Ali Tigrine
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, B-9000 Ghent, Belgium
| | - Victor R De La Rosa
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, B-9000 Ghent, Belgium
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, B-9000 Ghent, Belgium
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028 Barcelona, Spain.,Department of Biomedical Engineering, Institute of Complex Molecular Systems (ICMS), Eindhoven University of Technology (TUE), Eindhoven 5612 AZ, The Netherlands
| | - Roey J Amir
- Department of Organic Chemistry, School of Chemistry, Faculty of Exact Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel.,Tel Aviv University Center for Nanoscience and Nanotechnology, Tel-Aviv University, Tel-Aviv 6997801, Israel.,BLAVATNIK Center for Drug Discovery, Tel-Aviv University, Tel-Aviv 6997801, Israel.,ADAMA Center for Novel Delivery Systems in Crop Protection, Tel-Aviv University, Tel-Aviv 6997801, Israel.,The Center for Physics and Chemistry of Living Systems, Tel-Aviv University, Tel-Aviv 6997801, Israel
| |
Collapse
|
348
|
Worm M, Bauer A, Wedi B, Treudler R, Pfuetzner W, Brockow K, Buhl T, Zuberbier T, Fluhr J, Wurpts G, Klimek L, Jakob T, Merk HF, Mülleneisen N, Roeseler S, Dickel H, Raap U, Kleine-Tebbe J. Practical recommendations for the allergological risk assessment of the COVID-19 vaccination - a harmonized statement of allergy centers in Germany. Allergol Select 2021; 5:72-76. [PMID: 33521511 PMCID: PMC7841415 DOI: 10.5414/alx02225e] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023] Open
Abstract
Severe allergic reactions to vaccines are very rare. Single severe reactions have occurred worldwide after vaccination with the new mRNA-based COVID-19 vaccines. PEG2000 is discussed as a possible trigger. We provide guidance on risk assessment regarding COVID-19 vaccination in patients with allergic diseases and suggest a standardized, resource-oriented diagnostic and therapeutic procedure. Reports of severe allergic reactions in the context of COVID-19 vaccination can be made via www.anaphylaxie.net using an online questionnaire.
Collapse
Affiliation(s)
- Margitta Worm
- Allergology and Immunology, Department of Dermatology, Venereology and Allergology, Campus Charité Mitte, University Medicine Berlin
| | - Andrea Bauer
- Clinic and Polyclinic for Dermatology, University Hospital Carl Gustav Carus at the Technical University Dresden
| | - Bettina Wedi
- Department of Dermatology, Allergology and Venereology Comprehensive Allergy Center (CAC) Treatment Center for Hereditary Angioedema in the ZSE, Hannover Medical School
| | - Regina Treudler
- Clinic of Dermatology, Venereology and Allergology, Leipzig University Medical Center
| | - Wolfgang Pfuetzner
- Hesse Allergy Center, Clinic for Dermatology and Allergology, Marburg University Hospital
| | - Knut Brockow
- Dermatology Clinic Campus Biederstein, Klinikum rechts der Isar, Technical University of Munich
| | - Timo Buhl
- Dermatology Venereology and Allergology Clinic, University Medical Center Göttingen Georg-August-University
| | - Torsten Zuberbier
- Department of Dermatology and Allergy, Comprehensive Allergy Center Charité - Universitätsmedizin Berlin, Germany, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health
| | - Joachim Fluhr
- Department of Dermatology and Allergy, Comprehensive Allergy Center Charité - Universitätsmedizin Berlin, Germany, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health
| | - Gerda Wurpts
- Department of Dermatology and Allergology, University Hospital Aachen
| | - Ludger Klimek
- Center for Rhinology and Allergology of the ENT University Clinic Mannheim, Wiesbaden
| | - Thilo Jakob
- Department of Dermatology, Venerology and Allergology, University Hospital Giessen
| | | | | | - Stefani Roeseler
- Allergy, Asthma and Anaphylaxis Center, Clinic of Pneumology, Allergology, Sleep and Respiratory Medicine, Augustinians Hospital, Cologne
| | - Heinrich Dickel
- Department of Allergology, Occupational and Environmental Dermatology, Clinic for Dermatology, Venereology and Allergology, St. Josef Hospital, Ruhr University Bochum
| | - Ulrike Raap
- Department of Experimental Allergology and Immunodermatology, Department of Human Medicine, University of Oldenburg, and
| | | |
Collapse
|
349
|
Fumoto S, Yamamoto T, Okami K, Maemura Y, Terada C, Yamayoshi A, Nishida K. Understanding In Vivo Fate of Nucleic Acid and Gene Medicines for the Rational Design of Drugs. Pharmaceutics 2021; 13:159. [PMID: 33530309 PMCID: PMC7911509 DOI: 10.3390/pharmaceutics13020159] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Nucleic acid and genetic medicines are increasingly being developed, owing to their potential to treat a variety of intractable diseases. A comprehensive understanding of the in vivo fate of these agents is vital for the rational design, discovery, and fast and straightforward development of the drugs. In case of intravascular administration of nucleic acids and genetic medicines, interaction with blood components, especially plasma proteins, is unavoidable. However, on the flip side, such interaction can be utilized wisely to manipulate the pharmacokinetics of the agents. In other words, plasma protein binding can help in suppressing the elimination of nucleic acids from the blood stream and deliver naked oligonucleotides and gene carriers into target cells. To control the distribution of these agents in the body, the ligand conjugation method is widely applied. It is also important to understand intracellular localization. In this context, endocytosis pathway, endosomal escape, and nuclear transport should be considered and discussed. Encapsulated nucleic acids and genes must be dissociated from the carriers to exert their activity. In this review, we summarize the in vivo fate of nucleic acid and gene medicines and provide guidelines for the rational design of drugs.
Collapse
Affiliation(s)
- Shintaro Fumoto
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan; (T.Y.); (K.O.); (Y.M.); (C.T.); (A.Y.); (K.N.)
| | | | | | | | | | | | | |
Collapse
|
350
|
Najahi-Missaoui W, Arnold RD, Cummings BS. Safe Nanoparticles: Are We There Yet? Int J Mol Sci 2020; 22:ijms22010385. [PMID: 33396561 PMCID: PMC7794803 DOI: 10.3390/ijms22010385] [Citation(s) in RCA: 235] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/24/2020] [Accepted: 12/27/2020] [Indexed: 12/14/2022] Open
Abstract
The field of nanotechnology has grown over the last two decades and made the transition from the benchtop to applied technologies. Nanoscale-sized particles, or nanoparticles, have emerged as promising tools with broad applications in drug delivery, diagnostics, cosmetics and several other biological and non-biological areas. These advances lead to questions about nanoparticle safety. Despite considerable efforts to understand the toxicity and safety of these nanoparticles, many of these questions are not yet fully answered. Nevertheless, these efforts have identified several approaches to minimize and prevent nanoparticle toxicity to promote safer nanotechnology. This review summarizes our current knowledge on nanoparticles, their toxic effects, their interactions with mammalian cells and finally current approaches to minimizing their toxicity.
Collapse
Affiliation(s)
- Wided Najahi-Missaoui
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA;
- Correspondence: ; Tel.: +1-706-542-6552; Fax: +70-6542-5358
| | - Robert D. Arnold
- Department of Drug Discovery & Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA;
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA 30602, USA
| | - Brian S. Cummings
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA;
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|