1
|
Lopalco A, Iacobazzi RM, Lopedota AA, Denora N. Recent Advances in Nanodrug Delivery Systems Production, Efficacy, Safety, and Toxicity. Methods Mol Biol 2025; 2834:303-332. [PMID: 39312172 DOI: 10.1007/978-1-0716-4003-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
In the last three decades, the development of nanoparticles or nano-formulations as drug delivery systems has emerged as a promising tool to overcome the limitations of conventional delivery, potentially to improve the stability and solubility of active molecules, promote their transport across the biological membranes, and prolong circulation times to increase efficacy of a therapy. Despite several nano-formulations having applications in drug delivery, some issues concerning their safety and toxicity are still debated. This chapter describes the recent available information regarding safety, toxicity, and efficacy of nano-formulations for drug delivery. Several key factors can influence the behavior of nanoparticles in a biological environment, and their evaluation is crucial to design non-toxic and effective nano-formulations. Among them, we have focused our attention on materials and methods for their preparation (including the innovative microfluidic technique), mechanisms of interactions with biological systems, purification of nanoparticles, manufacture impurities, and nano-stability. This chapter places emphasis on the utilization of in silico, in vitro, and in vivo models for the assessment and prediction of toxicity associated with these nano-formulations. Furthermore, the chapter includes specific examples of in vitro and in vivo studies conducted on nanoparticles, illustrating their application in this field.
Collapse
Affiliation(s)
- Antonio Lopalco
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Bari, Italy.
| | - Rosa Maria Iacobazzi
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Angela Assunta Lopedota
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Nunzio Denora
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Bari, Italy
| |
Collapse
|
2
|
Singh G, Thakur N, Kumar R. Nanoparticles in drinking water: Assessing health risks and regulatory challenges. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 949:174940. [PMID: 39047836 DOI: 10.1016/j.scitotenv.2024.174940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Nanoparticles (NPs) pose a significant concern in drinking water due to their potential health risks and environmental impact. This review provides a comprehensive analysis of the current understanding of NP sources and contamination in drinking water, focusing on health concerns, mitigation strategies, regulatory frameworks, and future perspectives. This review highlights the importance of nano-specific pathways, fate processes, health risks & toxicity, and the need for realistic toxicity assessments. Different NPs like titanium dioxide, silver, nanoplastics, nanoscale liquid crystal monomers, copper oxide, and others pose potential health risks through ingestion, inhalation, or dermal exposure, impacting organs and potentially leading to oxidative stress, inflammatory responses, DNA damage, cytotoxicity, disrupt intracellular energetic mechanisms, reactive oxygen species generation, respiratory and immune toxicity, and genotoxicity in humans. Utilizing case studies and literature reviews, we investigate the health risks associated with NPs in freshwater environments, emphasizing their relevance to drinking water quality. Various mitigation and treatment strategies, including filtration systems (e.g., reverse osmosis, and ultra/nano-filtration), adsorption processes, coagulation/flocculation, electrocoagulation, advanced oxidation processes, membrane distillation, and ultraviolet treatment, all of which demonstrate high removal efficiencies for NPs from drinking water. Regulatory frameworks and challenges for the production, applications, and disposal of NPs at both national and international levels are discussed, emphasizing the need for tailored regulations to address NP contamination and standardize safety testing and risk assessment practices. Looking ahead, this review underscores the necessity of advancing detection methods and nanomaterial-based treatment technologies while stressing the pivotal role of public awareness and tailored regulatory guidelines in upholding drinking water quality standards. This review emphasizes the urgency of addressing NP contamination in drinking water and provides insights into potential solutions and future research directions. Lastly, this review worth concluded with future recommendations on advanced analytical techniques and sensitive sensors for NP detection for safeguarding public health and policy implementations.
Collapse
Affiliation(s)
- Gagandeep Singh
- Department of Biosciences (UIBT), Chandigarh University, Ludhiana, Punjab 140413, India
| | - Neelam Thakur
- Department of Zoology, Sardar Patel University, Vallabh Government College, Campus, Mandi, Himachal Pradesh 175001, India.
| | - Rakesh Kumar
- Department of Biosystems Engineering, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
3
|
Liu Y, Huang Y, He G, Guo C, Dong J, Wu L. Development of mRNA Lipid Nanoparticles: Targeting and Therapeutic Aspects. Int J Mol Sci 2024; 25:10166. [PMID: 39337651 PMCID: PMC11432440 DOI: 10.3390/ijms251810166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 09/30/2024] Open
Abstract
Lipid nanoparticles (LNPs) have emerged as leading non-viral carriers for messenger RNA (mRNA) delivery in clinical applications. Overcoming challenges in safe and effective mRNA delivery to target tissues and cells, along with controlling release from the delivery vehicle, remains pivotal in mRNA-based therapies. This review elucidates the structure of LNPs, the mechanism for mRNA delivery, and the targeted delivery of LNPs to various cells and tissues, including leukocytes, T-cells, dendritic cells, Kupffer cells, hepatic endothelial cells, and hepatic and extrahepatic tissues. Here, we discuss the applications of mRNA-LNP vaccines for the prevention of infectious diseases and for the treatment of cancer and various genetic diseases. Although challenges remain in terms of delivery efficiency, specific tissue targeting, toxicity, and storage stability, mRNA-LNP technology holds extensive potential for the treatment of diseases.
Collapse
Affiliation(s)
- Yaping Liu
- College of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
- Center for Chemical Biology and Drug Discovery, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yingying Huang
- College of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
- Center for Chemical Biology and Drug Discovery, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Guantao He
- Center for Chemical Biology and Drug Discovery, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chun Guo
- College of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinhua Dong
- College of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linping Wu
- Center for Chemical Biology and Drug Discovery, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| |
Collapse
|
4
|
Mohajeri M, Salehi P, Heidari B, Rafati H, Asghari SM, Behboudi H, Iranpour P. PEGylated Pemetrexed and PolyNIPAM Decorated Gold Nanoparticles: A Biocompatible and Highly Stable CT Contrast Agent for Cancer Imaging. ACS APPLIED BIO MATERIALS 2024; 7:5977-5991. [PMID: 39120942 DOI: 10.1021/acsabm.4c00563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
This study describes a multifunctional nanoparticle platform for targeted CT imaging and therapy of cancers. Pemetrexed (conjugated with polyethylene glycol, MW 2000 Da) and polyNIPAM (PEGylated) were designed for targeted delivery to folate receptors and thermally ablated tumors, respectively. These moieties were coated on gold nanoparticles (7 and 30 nm), and the prepared compounds were characterized using 1H NMR, FT-IR, CHNS, DLS, TEM, TGA, and UV-vis. The resulting agents exhibited 2-4 times higher X-ray attenuation compared to Visipaque and demonstrated specific accumulation in tumor tissue (4T1 xenograft model) 90 min after injection in mice. The nanoparticles displayed anticancer activity against 4T1 and MDA-MB-231 breast cancer cells (IC50: 182.87 and 206.18 μg/mL) and good biocompatibility. Importantly, the platform showed excellent stability over a year and at pH 2-12 and temperature range of -78 to 40 °C, and a water-dichloromethane extraction method was optimized for efficient purification, facilitating large-scale production.
Collapse
Affiliation(s)
- Mohammad Mohajeri
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Evin 1983963113 Tehran, Iran
| | - Peyman Salehi
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Evin 1983963113 Tehran, Iran
| | - Bahareh Heidari
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Evin 1983963113 Tehran, Iran
| | - Hasan Rafati
- Department of Pharmaceutical Engineering, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, 1983963113 Tehran, Iran
| | - S Mohsen Asghari
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 13145-1384, Iran
| | - Hossein Behboudi
- Department of Biology, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, 1983963113 Tehran, Iran
| | - Pooya Iranpour
- Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz 71936-13311, Iran
| |
Collapse
|
5
|
Kachanov A, Kostyusheva A, Brezgin S, Karandashov I, Ponomareva N, Tikhonov A, Lukashev A, Pokrovsky V, Zamyatnin AA, Parodi A, Chulanov V, Kostyushev D. The menace of severe adverse events and deaths associated with viral gene therapy and its potential solution. Med Res Rev 2024; 44:2112-2193. [PMID: 38549260 DOI: 10.1002/med.22036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 08/09/2024]
Abstract
Over the past decade, in vivo gene replacement therapy has significantly advanced, resulting in market approval of numerous therapeutics predominantly relying on adeno-associated viral vectors (AAV). While viral vectors have undeniably addressed several critical healthcare challenges, their clinical application has unveiled a range of limitations and safety concerns. This review highlights the emerging challenges in the field of gene therapy. At first, we discuss both the role of biological barriers in viral gene therapy with a focus on AAVs, and review current landscape of in vivo human gene therapy. We delineate advantages and disadvantages of AAVs as gene delivery vehicles, mostly from the safety perspective (hepatotoxicity, cardiotoxicity, neurotoxicity, inflammatory responses etc.), and outline the mechanisms of adverse events in response to AAV. Contribution of every aspect of AAV vectors (genomic structure, capsid proteins) and host responses to injected AAV is considered and substantiated by basic, translational and clinical studies. The updated evaluation of recent AAV clinical trials and current medical experience clearly shows the risks of AAVs that sometimes overshadow the hopes for curing a hereditary disease. At last, a set of established and new molecular and nanotechnology tools and approaches are provided as potential solutions for mitigating or eliminating side effects. The increasing number of severe adverse reactions and, sadly deaths, demands decisive actions to resolve the issue of immune responses and extremely high doses of viral vectors used for gene therapy. In response to these challenges, various strategies are under development, including approaches aimed at augmenting characteristics of viral vectors and others focused on creating secure and efficacious non-viral vectors. This comprehensive review offers an overarching perspective on the present state of gene therapy utilizing both viral and non-viral vectors.
Collapse
Affiliation(s)
- Artyom Kachanov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Anastasiya Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Sergey Brezgin
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Ivan Karandashov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Natalia Ponomareva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Andrey Tikhonov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Alexander Lukashev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Vadim Pokrovsky
- Laboratory of Biochemical Fundamentals of Pharmacology and Cancer Models, Blokhin Cancer Research Center, Moscow, Russia
- Department of Biochemistry, People's Friendship University, Russia (RUDN University), Moscow, Russia
| | - Andrey A Zamyatnin
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Belozersky Research, Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Vladimir Chulanov
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Infectious Diseases, Sechenov University, Moscow, Russia
| | - Dmitry Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
6
|
Beraza-Millor M, Rodríguez-Castejón J, Del Pozo-Rodríguez A, Rodríguez-Gascón A, Solinís MÁ. Systematic Review of Genetic Substrate Reduction Therapy in Lysosomal Storage Diseases: Opportunities, Challenges and Delivery Systems. BioDrugs 2024; 38:657-680. [PMID: 39177875 PMCID: PMC11358353 DOI: 10.1007/s40259-024-00674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Genetic substrate reduction therapy (gSRT), which involves the use of nucleic acids to downregulate the genes involved in the biosynthesis of storage substances, has been investigated in the treatment of lysosomal storage diseases (LSDs). OBJECTIVE To analyze the application of gSRT to the treatment of LSDs, identifying the silencing tools and delivery systems used, and the main challenges for its development and clinical translation, highlighting the contribution of nanotechnology to overcome them. METHODS A systematic review following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) reporting guidelines was performed. PubMed, Scopus, and Web of Science databases were used for searching terms related to LSDs and gene-silencing strategies and tools. RESULTS Fabry, Gaucher, and Pompe diseases and mucopolysaccharidoses I and III are the only LSDs for which gSRT has been studied, siRNA and lipid nanoparticles being the silencing strategy and the delivery system most frequently employed, respectively. Only in one recently published study was CRISPR/Cas9 applied to treat Fabry disease. Specific tissue targeting, availability of relevant cell and animal LSD models, and the rare disease condition are the main challenges with gSRT for the treatment of these diseases. Out of the 11 studies identified, only two gSRT studies were evaluated in animal models. CONCLUSIONS Nucleic acid therapies are expanding the clinical tools and therapies currently available for LSDs. Recent advances in CRISPR/Cas9 technology and the growing impact of nanotechnology are expected to boost the clinical translation of gSRT in the near future, and not only for LSDs.
Collapse
Affiliation(s)
- Marina Beraza-Millor
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country, UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Spain
| | - Julen Rodríguez-Castejón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country, UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Spain
| | - Ana Del Pozo-Rodríguez
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country, UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Spain
| | - Alicia Rodríguez-Gascón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country, UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Spain
| | - María Ángeles Solinís
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country, UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain.
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Spain.
| |
Collapse
|
7
|
Shah MM, Layhadi JA, Hourcade DE, Fulton WT, Tan TJ, Dunham D, Chang I, Vel MS, Fernandes A, Lee AS, Liu J, Arunachalam PS, Galli SJ, Boyd SD, Pulendran B, Davis MM, O’Hara R, Park H, Mitchell LM, Akk A, Patterson A, Jerath MR, Monroy JM, Ren Z, Kendall PL, Durham SR, Fedina A, Gibbs BF, Agache I, Chinthrajah S, Sindher SB, Heider A, Akdis CA, Shamji MH, Pham CT, Nadeau KC. Elucidating allergic reaction mechanisms in response to SARS-CoV-2 mRNA vaccination in adults. Allergy 2024; 79:2502-2523. [PMID: 39033312 PMCID: PMC11368657 DOI: 10.1111/all.16231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/31/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND During the COVID-19 pandemic, novel nanoparticle-based mRNA vaccines were developed. A small number of individuals developed allergic reactions to these vaccines although the mechanisms remain undefined. METHODS To understand COVID-19 vaccine-mediated allergic reactions, we enrolled 19 participants who developed allergic events within 2 h of vaccination and 13 controls, nonreactors. Using standard hemolysis assays, we demonstrated that sera from allergic participants induced stronger complement activation compared to nonallergic subjects following ex vivo vaccine exposure. RESULTS Vaccine-mediated complement activation correlated with anti-polyethelyne glycol (PEG) IgG (but not IgM) levels while anti-PEG IgE was undetectable in all subjects. Depletion of total IgG suppressed complement activation in select individuals. To investigate the effects of vaccine excipients on basophil function, we employed a validated indirect basophil activation test that stratified the allergic populations into high and low responders. Complement C3a and C5a receptor blockade in this system suppressed basophil response, providing strong evidence for complement involvement in vaccine-mediated basophil activation. Single-cell multiome analysis revealed differential expression of genes encoding the cytokine response and Toll-like receptor (TLR) pathways within the monocyte compartment. Differential chromatin accessibility for IL-13 and IL-1B genes was found in allergic and nonallergic participants, suggesting that in vivo, epigenetic modulation of mononuclear phagocyte immunophenotypes determines their subsequent functional responsiveness, contributing to the overall physiologic manifestation of vaccine reactions. CONCLUSION These findings provide insights into the mechanisms underlying allergic reactions to COVID-19 mRNA vaccines, which may be used for future vaccine strategies in individuals with prior history of allergies or reactions and reduce vaccine hesitancy.
Collapse
Affiliation(s)
- Mihir M. Shah
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
- These authors contributed equally to this work
| | - Janice A. Layhadi
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
- These authors contributed equally to this work
| | - Dennis E. Hourcade
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
- These authors contributed equally to this work
| | - William T. Fulton
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Tiak Ju Tan
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Diane Dunham
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Iris Chang
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Monica S. Vel
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Andrea Fernandes
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Alexandra S. Lee
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - James Liu
- Stanford Health Library; Stanford, CA, USA
| | - Prabhu S. Arunachalam
- Institute for Immunity, Transplantation and Infection,
Stanford University; Stanford, CA, USA
| | - Stephen J. Galli
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
- Department of Pathology, Stanford University School of
Medicine; Stanford, California, USA
- Department of Microbiology and Immunology, Stanford
University School of Medicine; Stanford, California, USA
| | - Scott D. Boyd
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
- Department of Pathology, Stanford University School of
Medicine; Stanford, California, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection,
Stanford University; Stanford, CA, USA
- Department of Pathology, Stanford University School of
Medicine; Stanford, California, USA
- Department of Microbiology and Immunology, Stanford
University School of Medicine; Stanford, California, USA
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection,
Stanford University; Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford
University School of Medicine; Stanford, California, USA
| | - Ruth O’Hara
- Department of Veteran’s Administration and
Dean’s Office, Stanford University; Stanford, CA, USA
| | - Helen Park
- Veterans Affairs Palo Alto Health Care System; Palo Alto,
CA, USA
| | - Lynne M. Mitchell
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
| | - Antonina Akk
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
| | - Alexander Patterson
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Maya R. Jerath
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Jennifer M. Monroy
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Zhen Ren
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Peggy L. Kendall
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Stephen R. Durham
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Aleksandra Fedina
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Bernhard F Gibbs
- Department of Human Medicine, School of Medicine and
Health Sciences, Carl von Ossietzky University of Oldenburg; Oldenburg,
Germany
- Canterbury Christ Church University, UK
| | - Ioana Agache
- Faculty of Medicine, Transilvania University; Brasov,
Romania
| | | | | | - Anja Heider
- Swiss Institute of Allergy and Asthma Research (SIAF),
University of Zurich; Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF),
University of Zurich; Davos, Switzerland
| | - Mohamed H. Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
- These authors contributed equally to this work
| | - Christine T.N. Pham
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
- These authors contributed equally to this work
| | - Kari C. Nadeau
- Harvard T.H. Chan School of Public Health, Harvard
University; Cambridge, Massachusetts
- These authors contributed equally to this work
| |
Collapse
|
8
|
Chen C, Li J, Chen Y, Gao Q, Li N, Le S. The correlation of asparaginase enzyme activity levels after PEG-asparaginase administration with clinical characteristics and adverse effects in Chinese paediatric patients with acute lymphoblastic leukaemia. Br J Haematol 2024; 205:624-633. [PMID: 38934331 DOI: 10.1111/bjh.19605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
Studies on asparaginase enzyme activity (AEA) monitoring in Chinese patients receiving PEG-asparaginase remain limited. We monitored AEA in paediatric patients diagnosed with acute lymphoblastic leukaemia (ALL) and treated according to the Chinese Children's Cancer Group study protocols, CCCG-ALL-2015/CCCG-ALL-2020 protocols. We measured the AEA at days 7 ± 1 and 14 ± 1 and analysed their association with patient characteristics and PEG-asparaginase-related adverse effects (AEs). We measured 2147 samples from 329 patients. Mean AEA levels (interquartile range) were 931 iu/L (654-1174 iu/L) at day 7 ± 1 and 664 iu/L (463-860 iu/L) at day 14 ± 1. The AEA levels were higher in younger children and increased with the cumulative dose numbers. PEG-asparaginase inactivation rate was 19.1%, and the silent inactivation (SI) rate was 12.5%. Nine patients were identified with allergic-like reactions. Hypofibrinogenaemia, hypertriglyceridaemia, pancreatitis and thrombosis were associated with older age, whereas hypoglycaemia was associated with younger age. The risk of hypertriglyceridaemia and hypoglycaemia increased with cumulative dose numbers of PEG-asparaginase. Except for hypofibrinogenaemia, elevated AEA levels did not increase the risk of PEG-asparaginase-related AEs. Drug monitoring can be utilized as guidance for treatment decision-making. Individualizing asparaginase doses do not reduce toxicities. The treatment target of PEG-asparaginase remains to achieve sustained and adequate activity.
Collapse
Affiliation(s)
- Cai Chen
- Department of Pediatric Hematology, Fujian Medical University, Union Hospital, Fuzhou, China
| | - Jian Li
- Department of Pediatric Hematology, Fujian Medical University, Union Hospital, Fuzhou, China
| | - Yiqiao Chen
- Department of Pediatric Hematology, Fujian Medical University, Union Hospital, Fuzhou, China
| | - Qinli Gao
- Department of Pediatric Hematology, Fujian Medical University, Union Hospital, Fuzhou, China
| | - Nainong Li
- Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shaohua Le
- Department of Pediatric Hematology, Fujian Medical University, Union Hospital, Fuzhou, China
| |
Collapse
|
9
|
Humphries J, Fletcher NL, Sonderegger SE, Bell CA, Kempe K, Thurecht KJ. Mitigating the Effects of Persistent Antipolymer Immune Reactions in Nanomedicine: Evaluating Materials-Based Approaches Using Molecular Imaging. ACS NANO 2024. [PMID: 39037055 DOI: 10.1021/acsnano.4c07317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Poly(ethylene glycol) (PEG) is a hydrophilic polymer ubiquitously used in both medical and nonmedical goods. Recent debate surrounding the observed stimulation of immune responses against PEG has spurred the development of materials that may be suitable replacements for this common polymeric component. The underlying view is that these alternative materials with comparable physicochemical properties can overcome the unfavorable and unpredictable effects of antibody-mediated clearance by being chemically, and therefore antigenically, distinct from PEG. However, this hypothesis has not been thoroughly tested in any defined manner, and the immune response observed against PEG has not been rigorously investigated within the context of these emerging materials. Consequently, it remains unclear whether immunity-mediated discrimination between polymeric entities even occurs in vivo and, if this is the case, how it may be exploited. In this study, we utilize positron emission tomography-computed tomography molecular imaging in mice immunized to develop specific antibody responses to PEG and an alternative polymer in order to visualize and quantify the influence of antipolymer antibodies on the biodistribution of synthetic polymers in vivo as a function of immunization status. Under the conditions of this experiment, mice could be primed to exhibit both innate and adaptive immunity to all of the polymer systems to which they were exposed. We demonstrate that alternating between chemically disparate polymers is a viable approach to extend their efficacy when antipolymer humoral immune responses arise.
Collapse
Affiliation(s)
- James Humphries
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Stefan E Sonderegger
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Craig A Bell
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
10
|
Sanchez AJDS, Loughrey D, Echeverri ES, Huayamares SG, Radmand A, Paunovska K, Hatit M, Tiegreen KE, Santangelo PJ, Dahlman JE. Substituting Poly(ethylene glycol) Lipids with Poly(2-ethyl-2-oxazoline) Lipids Improves Lipid Nanoparticle Repeat Dosing. Adv Healthc Mater 2024; 13:e2304033. [PMID: 38318754 DOI: 10.1002/adhm.202304033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/22/2024] [Indexed: 02/07/2024]
Abstract
Poly(ethylene glycol) (PEG)-lipids are used in Food-and-Drug-Administration-approved lipid nanoparticle (LNP)-RNA drugs, which are safe and effective. However, it is reported that PEG-lipids may also contribute to accelerated blood clearance and rare cases of hypersensitivity; this highlights the utility of exploring PEG-lipid alternatives. Here, it is shown that LNPs containing poly(2-ethyl-2-oxazoline) (PEOZ)-lipids can deliver messenger RNA (mRNA) to multiple cell types in mice inside and outside the liver. In addition, it is reported that LNPs formulated with PEOZ-lipids show reduced clearance from the bloodstream and lower levels of antistealth lipid immunoglobulin Ms than LNPs formulated with PEG-lipids. These data justify further exploration of PEOZ-lipids as alternatives to PEG-lipids in LNP-RNA formulations.
Collapse
Affiliation(s)
- Alejandro J Da Silva Sanchez
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Department of Chemical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - David Loughrey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Elisa Schrader Echeverri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Sebastian G Huayamares
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Afsane Radmand
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Department of Chemical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Marine Hatit
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Karen E Tiegreen
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
11
|
Yildiz SN, Entezari M, Paskeh MDA, Mirzaei S, Kalbasi A, Zabolian A, Hashemi F, Hushmandi K, Hashemi M, Raei M, Goharrizi MASB, Aref AR, Zarrabi A, Ren J, Orive G, Rabiee N, Ertas YN. Nanoliposomes as nonviral vectors in cancer gene therapy. MedComm (Beijing) 2024; 5:e583. [PMID: 38919334 PMCID: PMC11199024 DOI: 10.1002/mco2.583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 06/27/2024] Open
Abstract
Nonviral vectors, such as liposomes, offer potential for targeted gene delivery in cancer therapy. Liposomes, composed of phospholipid vesicles, have demonstrated efficacy as nanocarriers for genetic tools, addressing the limitations of off-targeting and degradation commonly associated with traditional gene therapy approaches. Due to their biocompatibility, stability, and tunable physicochemical properties, they offer potential in overcoming the challenges associated with gene therapy, such as low transfection efficiency and poor stability in biological fluids. Despite these advancements, there remains a gap in understanding the optimal utilization of nanoliposomes for enhanced gene delivery in cancer treatment. This review delves into the present state of nanoliposomes as carriers for genetic tools in cancer therapy, sheds light on their potential to safeguard genetic payloads and facilitate cell internalization alongside the evolution of smart nanocarriers for targeted delivery. The challenges linked to their biocompatibility and the factors that restrict their effectiveness in gene delivery are also discussed along with exploring the potential of nanoliposomes in cancer gene therapy strategies by analyzing recent advancements and offering future directions.
Collapse
Affiliation(s)
| | - Maliheh Entezari
- Department of GeneticsFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of Medical Convergence SciencesFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mahshid Deldar Abad Paskeh
- Department of GeneticsFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of Medical Convergence SciencesFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Sepideh Mirzaei
- Department of BiologyFaculty of ScienceIslamic Azad UniversityScience and Research BranchTehranIran
| | - Alireza Kalbasi
- Department of PharmacyBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Amirhossein Zabolian
- Department of OrthopedicsShahid Beheshti University of Medical SciencesTehranIran
| | - Farid Hashemi
- Department of Comparative BiosciencesFaculty of Veterinary MedicineUniversity of TehranTehranIran
| | - Kiavash Hushmandi
- Department of Clinical Sciences InstituteNephrology and Urology Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | - Mehrdad Hashemi
- Department of GeneticsFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of Medical Convergence SciencesFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mehdi Raei
- Department of Epidemiology and BiostatisticsSchool of HealthBaqiyatallah University of Medical SciencesTehranIran
| | | | - Amir Reza Aref
- Belfer Center for Applied Cancer ScienceDana‐Farber Cancer InstituteHarvard Medical SchoolBostonMassachusettsUSA
- Department of Translational SciencesXsphera Biosciences Inc.BostonMassachusettsUSA
| | - Ali Zarrabi
- Department of Biomedical EngineeringFaculty of Engineering and Natural SciencesIstinye UniversityIstanbulTurkey
| | - Jun Ren
- Shanghai Institute of Cardiovascular DiseasesDepartment of CardiologyZhongshan HospitalFudan UniversityShanghaiChina
| | - Gorka Orive
- NanoBioCel Research GroupSchool of PharmacyUniversity of the Basque Country (UPV/EHU)Vitoria‐GasteizSpain
- University Institute for Regenerative Medicine and Oral Implantology ‐ UIRMI (UPV/EHU‐Fundación Eduardo Anitua)Vitoria‐GasteizSpain
- Bioaraba, NanoBioCel Research GroupVitoria‐GasteizSpain
- The AcademiaSingapore Eye Research InstituteSingaporeSingapore
| | - Navid Rabiee
- Centre for Molecular Medicine and Innovative TherapeuticsMurdoch UniversityPerthWestern AustraliaAustralia
| | - Yavuz Nuri Ertas
- Department of Biomedical EngineeringErciyes UniversityKayseriTurkey
- ERNAM—Nanotechnology Research and Application CenterErciyes UniversityKayseriTurkey
- UNAM−National Nanotechnology Research CenterBilkent UniversityAnkaraTurkey
| |
Collapse
|
12
|
Anchordoquy T, Artzi N, Balyasnikova IV, Barenholz Y, La-Beck NM, Brenner JS, Chan WCW, Decuzzi P, Exner AA, Gabizon A, Godin B, Lai SK, Lammers T, Mitchell MJ, Moghimi SM, Muzykantov VR, Peer D, Nguyen J, Popovtzer R, Ricco M, Serkova NJ, Singh R, Schroeder A, Schwendeman AA, Straehla JP, Teesalu T, Tilden S, Simberg D. Mechanisms and Barriers in Nanomedicine: Progress in the Field and Future Directions. ACS NANO 2024; 18:13983-13999. [PMID: 38767983 PMCID: PMC11214758 DOI: 10.1021/acsnano.4c00182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
In recent years, steady progress has been made in synthesizing and characterizing engineered nanoparticles, resulting in several approved drugs and multiple promising candidates in clinical trials. Regulatory agencies such as the Food and Drug Administration and the European Medicines Agency released important guidance documents facilitating nanoparticle-based drug product development, particularly in the context of liposomes and lipid-based carriers. Even with the progress achieved, it is clear that many barriers must still be overcome to accelerate translation into the clinic. At the recent conference workshop "Mechanisms and Barriers in Nanomedicine" in May 2023 in Colorado, U.S.A., leading experts discussed the formulation, physiological, immunological, regulatory, clinical, and educational barriers. This position paper invites open, unrestricted, nonproprietary discussion among senior faculty, young investigators, and students to trigger ideas and concepts to move the field forward.
Collapse
Affiliation(s)
- Thomas Anchordoquy
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Natalie Artzi
- Brigham and Woman's Hospital, Department of Medicine, Division of Engineering in Medicine, Harvard Medical School, Boston, Massachusetts 02215, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| | - Irina V Balyasnikova
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University; Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Yechezkel Barenholz
- Membrane and Liposome Research Lab, IMRIC, Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel
| | - Ninh M La-Beck
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, Texas 79601, United States
| | - Jacob S Brenner
- Departments of Medicine and Pharmacology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Warren C W Chan
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, 16163 Genova, Italy
| | - Agata A Exner
- Departments of Radiology and Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Alberto Gabizon
- The Helmsley Cancer Center, Shaare Zedek Medical Center and The Hebrew University of Jerusalem-Faculty of Medicine, Jerusalem, 9103102, Israel
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Obstetrics and Gynecology, Houston Methodist Hospital, Houston, Texas 77030, United States
- Department of Obstetrics and Gynecology, Weill Cornell Medicine College (WCMC), New York, New York 10065, United States
- Department of Biomedical Engineering, Texas A&M, College Station, Texas 7784,3 United States
| | - Samuel K Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Center for Biohybrid Medical Systems, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - S Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, U.K
- Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, U.K
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Center, Aurora, Colorado 80045, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachela Popovtzer
- Faculty of Engineering and the Institute of Nanotechnology & Advanced Materials, Bar-Ilan University, 5290002 Ramat Gan, Israel
| | - Madison Ricco
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Natalie J Serkova
- Department of Radiology, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Ravi Singh
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina 27101, United States
| | - Avi Schroeder
- Department of Chemical Engineering, Technion, Israel Institute of Technology, Haifa 32000, Israel
| | - Anna A Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48108; Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48108, United States
| | - Joelle P Straehla
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts 02115 United States
- Koch Institute for Integrative Cancer Research at MIT, Cambridge Massachusetts 02139 United States
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Scott Tilden
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Dmitri Simberg
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
13
|
Li Y, Ettah U, Jacques S, Gaikwad H, Monte A, Dylla L, Guntupalli S, Moghimi SM, Simberg D. Optimized Enzyme-Linked Immunosorbent Assay for Anti-PEG Antibody Detection in Healthy Donors and Patients Treated with PEGylated Liposomal Doxorubicin. Mol Pharm 2024; 21:3053-3060. [PMID: 38743264 DOI: 10.1021/acs.molpharmaceut.4c00278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
There is considerable interest in quantifying anti-PEG antibodies, given their potential involvement in accelerated clearance, complement activation, neutralization, and acute reactions associated with drug delivery systems. Published and commercially available anti-PEG enzyme-linked immunosorbent assays (ELISAs) differ significantly in terms of reagents and conditions, which could be confusing to users who want to perform in-house measurements. Here, we optimize the ELISA protocol for specific detection of anti-PEG IgG and IgM in sera from healthy donors and in plasma from cancer patients administered with PEGylated liposomal doxorubicin. The criterion of specificity is the ability of free PEG or PEGylated liposomes to inhibit the ELISA signals. We found that coating high-binding plates with monoamine methoxy-PEG5000, as opposed to bovine serum albumin-PEG20000, and blocking with 1% milk, as opposed to albumin or lysozyme, significantly improve the specificity, with over 95% of the signal being blocked by competition. Despite inherent between-assay variability, setting the cutoff value of the optical density at the 80th percentile consistently identified the same subjects. Using the optimized assay, we longitudinally measured levels of anti-PEG IgG/IgM in cancer patients before and after the PEGylated liposomal doxorubicin chemotherapy cycle (1 month apart, three cycles total). Antibody titers did not show any increase but rather a decrease between treatment cycles, and up to 90% of antibodies was bound to the infused drug. This report is a step toward harmonizing anti-PEG assays in human subjects, emphasizing the cost-effectiveness and optimized specificity.
Collapse
Affiliation(s)
- Yue Li
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
| | - Utibeabasi Ettah
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
| | - Sarah Jacques
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
| | - Hanmant Gaikwad
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
| | - Andrew Monte
- Department of Emergency Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
| | - Layne Dylla
- Department of Emergency Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
| | - Saketh Guntupalli
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Colorado School of Medicine Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
| | - S Moein Moghimi
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU , U.K
- Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, U.K
| | - Dmitri Simberg
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045-2559, United States
| |
Collapse
|
14
|
Wu Y, Luo L, Hao Z, Liu D. DNA-based nanostructures for RNA delivery. MEDICAL REVIEW (2021) 2024; 4:207-224. [PMID: 38919398 PMCID: PMC11195427 DOI: 10.1515/mr-2023-0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/28/2024] [Indexed: 06/27/2024]
Abstract
RNA-based therapeutics have emerged as a promising approach for the treatment of various diseases, including cancer, genetic disorders, and infectious diseases. However, the delivery of RNA molecules into target cells has been a major challenge due to their susceptibility to degradation and inefficient cellular uptake. To overcome these hurdles, DNA-based nano technology offers an unprecedented opportunity as a potential delivery platform for RNA therapeutics. Due to its excellent characteristics such as programmability and biocompatibility, these DNA-based nanostructures, composed of DNA molecules assembled into precise and programmable structures, have garnered significant attention as ideal building materials for protecting and delivering RNA payloads to the desired cellular destinations. In this review, we highlight the current progress in the design and application of three DNA-based nanostructures: DNA origami, lipid-nanoparticle (LNP) technology related to frame guided assembly (FGA), and DNA hydrogel for the delivery of RNA molecules. Their biomedical applications are briefly discussed and the challenges and future perspectives in this field are also highlighted.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Beijing SupraCirc Biotechnology Co., Ltd, Beijing, China
| | - Liangzhi Luo
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Ziyang Hao
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Dongsheng Liu
- Department of Chemistry, Engineering Research Center of Advanced Rare Earth Materials, (Ministry of Education), Tsinghua University, Beijing, China
| |
Collapse
|
15
|
Riedelová Z, de Los Santos Pereira A, Dorado Daza DF, Májek P, Dyčka F, Riedel T. Mass-Spectrometric Identification of Proteins and Pathways Responsible for Fouling on Poly(ethylene glycol) Methacrylate Polymer Brushes. Macromol Biosci 2024; 24:e2300558. [PMID: 38350051 DOI: 10.1002/mabi.202300558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/09/2024] [Indexed: 02/15/2024]
Abstract
Prevention of fouling from proteins in blood plasma attracts significant efforts, and great progress is made in identifying surface coatings that display antifouling properties. In particular, poly(ethylene glycol) (PEG) is widely used and dense PEG-like cylindrical brushes of poly[oligo(ethylene glycol) methacrylate] (poly(OEGMA)) can drastically reduce blood plasma fouling. Herein, a comprehensive study of the variation of blood plasma fouling on this surface, including the analysis of the composition of protein deposits on poly(OEGMA) coatings after contact with blood plasma from many different donors, is reported. Correlation between the plasma fouling behavior and protein deposit composition points to the activation of the complement system as the main culprit of dramatically increased and accelerated deposition of blood plasma proteins on this type of antifouling coating, specifically through the classical pathway. These findings are consistent with observations on PEGylated drug carriers and highlight the importance of understanding the potential interactions between antifouling coatings and their environment.
Collapse
Affiliation(s)
- Zuzana Riedelová
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq. 2, Prague, 162 00, Czech Republic
| | - Andres de Los Santos Pereira
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq. 2, Prague, 162 00, Czech Republic
| | - Diego Fernando Dorado Daza
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq. 2, Prague, 162 00, Czech Republic
| | - Pavel Májek
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, Prague, 128 00, Czech Republic
| | - Filip Dyčka
- Faculty of Science, University of South Bohemia, Branišovská 1760, České Budějovice, 370 05, Czech Republic
| | - Tomáš Riedel
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq. 2, Prague, 162 00, Czech Republic
| |
Collapse
|
16
|
Journeaux T, Bernardes GJL. Homogeneous multi-payload antibody-drug conjugates. Nat Chem 2024; 16:854-870. [PMID: 38760431 DOI: 10.1038/s41557-024-01507-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 03/14/2024] [Indexed: 05/19/2024]
Abstract
Many systemic cancer chemotherapies comprise a combination of drugs, yet all clinically used antibody-drug conjugates (ADCs) contain a single-drug payload. These combination regimens improve treatment outcomes by producing synergistic anticancer effects and slowing the development of drug-resistant cell populations. In an attempt to replicate these regimens and improve the efficacy of targeted therapy, the field of ADCs has moved towards developing techniques that allow for multiple unique payloads to be attached to a single antibody molecule with high homogeneity. However, the methods for generating such constructs-homogeneous multi-payload ADCs-are both numerous and complex owing to the plethora of reactive functional groups that make up the surface of an antibody. Here, by summarizing and comparing the methods of both single- and multi-payload ADC generation and their key preclinical and clinical results, we provide a timely overview of this relatively new area of research. The methods discussed range from branched linker installation to the incorporation of unnatural amino acids, with a generalized comparison tool of the most promising modification strategies also provided. Finally, the successes and challenges of this rapidly growing field are critically evaluated, and from this, future areas of research and development are proposed.
Collapse
Affiliation(s)
- Toby Journeaux
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Gonçalo J L Bernardes
- Department of Chemistry, University of Cambridge, Cambridge, UK.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
17
|
Xie S, Erfani A, Manouchehri S, Ramsey J, Aichele C. Aerosolization of poly(sulfobetaine) microparticles that encapsulate therapeutic antibodies. BIOMATERIALS ADVANCES 2024; 160:213839. [PMID: 38579521 DOI: 10.1016/j.bioadv.2024.213839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/04/2024] [Accepted: 03/23/2024] [Indexed: 04/07/2024]
Abstract
Pulmonary delivery of protein therapeutics poses significant challenges that have not been well addressed in the research literature or practice. In fact, there is currently only one commercial protein therapeutic that is delivered through aerosolization and inhalation. In this study, we propose a drug delivery strategy that enables a high-concentration dosage for the pulmonary delivery of antibodies as an aerosolizable solid powder with desired stability. We utilized zwitterionic polymers for their promising properties as drug delivery vehicles and synthesized swellable, biodegradable poly(sulfo-betaine) (pSB) microparticles. The microparticles were loaded with Immunoglobulin G (IgG) as a model antibody. We quantified the microparticle size and morphology, and the particles were found to have an average diameter of 1.6 μm, falling within the optimal range (~1-5 μm) for pulmonary drug delivery. In addition, we quantified the impact of the crosslinker to monomer ratio on particle morphology and drug loading capacity. The results showed that there is a trade-off between desired morphology and drug loading capacity as the crosslinker density increases. In addition, the particles were aerosolized, and our data indicated that the particles remained intact and retained their initial morphology and size after aerosolization. The combination of morphology, particle size, antibody loading capacity, low cytotoxicity, and ease of aerosolization support the potential use of these particles for pulmonary delivery of protein therapeutics.
Collapse
Affiliation(s)
- Songpei Xie
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, United States of America
| | - Amir Erfani
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States of America
| | - Saeed Manouchehri
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, United States of America
| | - Joshua Ramsey
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, United States of America
| | - Clint Aichele
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, United States of America.
| |
Collapse
|
18
|
Chen R, Sandeman L, Nankivell V, Tan JTM, Rashidi M, Psaltis PJ, Zheng G, Bursill C, McLaughlin RA, Li J. Detection of atherosclerotic plaques with HDL-like porphyrin nanoparticles using an intravascular dual-modality optical coherence tomography and fluorescence system. Sci Rep 2024; 14:12359. [PMID: 38811670 PMCID: PMC11136962 DOI: 10.1038/s41598-024-63132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024] Open
Abstract
Atherosclerosis is the build-up of fatty plaques within blood vessel walls, which can occlude the vessels and cause strokes or heart attacks. It gives rise to both structural and biomolecular changes in the vessel walls. Current single-modality imaging techniques each measure one of these two aspects but fail to provide insight into the combined changes. To address this, our team has developed a dual-modality imaging system which combines optical coherence tomography (OCT) and fluorescence imaging that is optimized for a porphyrin lipid nanoparticle that emits fluorescence and targets atherosclerotic plaques. Atherosclerosis-prone apolipoprotein (Apo)e-/- mice were fed a high cholesterol diet to promote plaque development in descending thoracic aortas. Following infusion of porphyrin lipid nanoparticles in atherosclerotic mice, the fiber-optic probe was inserted into the aorta for imaging, and we were able to robustly detect a porphyrin lipid-specific fluorescence signal that was not present in saline-infused control mice. We observed that the nanoparticle fluorescence colocalized in areas of CD68+ macrophages. These results demonstrate that our system can detect the fluorescence from nanoparticles, providing complementary biological information to the structural information obtained from simultaneously acquired OCT.
Collapse
Affiliation(s)
- Rouyan Chen
- School of Electrical and Mechanical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, SA, 5005, Australia.
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia.
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, SA, 5005, Australia.
| | - Lauren Sandeman
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Victoria Nankivell
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Mohammad Rashidi
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide, SA, 5000, Australia
| | - Gang Zheng
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, ON, M5G 1L7, Toronto, Canada
| | - Christina Bursill
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Robert A McLaughlin
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, SA, 5005, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Jiawen Li
- School of Electrical and Mechanical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, SA, 5005, Australia.
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, SA, 5005, Australia.
| |
Collapse
|
19
|
Lee Y, Jeong M, Lee G, Park J, Jung H, Im S, Lee H. Development of Lipid Nanoparticle Formulation for the Repeated Administration of mRNA Therapeutics. Biomater Res 2024; 28:0017. [PMID: 38779139 PMCID: PMC11109479 DOI: 10.34133/bmr.0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/13/2024] [Indexed: 05/25/2024] Open
Abstract
During the COVID-19 pandemic, mRNA vaccines emerged as a rapid and effective solution for global immunization. The success of COVID-19 mRNA vaccines has increased interest in the use of lipid nanoparticles (LNPs) for the in vivo delivery of mRNA therapeutics. Although mRNA exhibits robust expression profiles, transient protein expression is often observed, raising uncertainty regarding the frequency of its administration. Additionally, various RNA therapeutics may necessitate repeated dosing to achieve optimal therapeutic outcomes. Nevertheless, the impact of repeated administrations of mRNA/LNP on immune responses and protein expression efficacy remains unclear. In this study, we investigated the influence of the formulation parameters, specifically ionizable lipids and polyethylene glycol (PEG) lipids, on the repeat administration of mRNA/LNP. Our findings revealed that ionizable lipids had no discernible impact on the dose-responsive efficacy of repeat administrations, whereas the lipid structure and molar ratio of PEG lipids were primary factors that affected mRNA/LNP performance. The optimization of the LNP formulation with PEG lipid confirmed the sustained dose-responsive efficacy of mRNA after repeated administrations. This study highlights the critical importance of optimizing LNP formulations for mRNA therapeutics requiring repeated administrations.
Collapse
Affiliation(s)
- Yeji Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Michaela Jeong
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Gyeongseok Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jeongeun Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hyein Jung
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seongeun Im
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hyukjin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
20
|
Zhang Z, Sun H, Giannino J, Wu Y, Cheng C. Biodegradable Zwitterionic Polymers as PEG Alternatives for Drug Delivery. JOURNAL OF POLYMER SCIENCE 2024; 62:2231-2250. [PMID: 39247254 PMCID: PMC11376432 DOI: 10.1002/pol.20230916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/01/2024] [Indexed: 09/10/2024]
Abstract
Poly(ethylene glycol) (PEG) is a highly biocompatible and water-soluble polymer that is widely utilized for biomedical applications. Unfortunately, the immunogenicity and antigenicity of PEG severely restrict the biomedical efficacy of pegylated therapeutics. As emerging PEG alternatives, biodegradable zwitterionic polymers (ZPs) have attracted significant interest in recent years. Biodegradable ZPs generally are not only water-soluble and immunologically inert, but also possess a range of favorable biomedically relevant properties, without causing long-term side effects for in vivo biomedical applications. This review presents a systematic overview of recent studies on biodegradable ZPs. Their structural designs and synthetic strategies by integrating biodegradable base polymers with zwitterions are addressed. Their applications in the delivery of small molecule drugs (as mono-drugs or multi-drugs) and proteins are highlighted.
Collapse
Affiliation(s)
- Ziwen Zhang
- Department of Chemical and Biological Engineering, University at Buffalo, the State University of New York, Buffalo, NY 14260
| | - Haotian Sun
- Department of Chemical and Biological Engineering, University at Buffalo, the State University of New York, Buffalo, NY 14260
| | - Justin Giannino
- Department of Biomedical Engineering, University at Buffalo, the State University of New York, Buffalo, NY 14260
| | - Yun Wu
- Department of Biomedical Engineering, University at Buffalo, the State University of New York, Buffalo, NY 14260
- Cell, Gene and Tissue Engineering Center, University at Buffalo, the State University of New York, Buffalo, NY 14260
| | - Chong Cheng
- Department of Chemical and Biological Engineering, University at Buffalo, the State University of New York, Buffalo, NY 14260
| |
Collapse
|
21
|
Navarro-Becerra JA, Castillo JI, Borden MA. Effect of Poly(ethylene glycol) Configuration on Microbubble Pharmacokinetics. ACS Biomater Sci Eng 2024; 10:3331-3342. [PMID: 38600786 DOI: 10.1021/acsbiomaterials.3c01764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Microbubbles (MBs) hold substantial promise for medical imaging and therapy; nonetheless, knowledge gaps persist between composition, structure, and in vivo performance, especially with respect to pharmacokinetics. Of particular interest is the role of the poly(ethylene glycol) (PEG) layer, which is thought to shield the MB against opsonization and rapid clearance but is also known to cause an antibody response upon multiple injections. The goal of this study was, therefore, to elucidate the role of the PEG layer in circulation persistence of MBs in the naïve animal (prior to an adaptive immune response). Here, we directly observe the number and size of individual MBs obtained from blood samples, unifying size and concentration into the microbubble volume dose (MVD) parameter. This approach enables direct evaluation of the pharmacokinetics of intact MBs, comprising both the lipid shell and gaseous core, rather than separately assessing the lipid or gas components. We examined the in vivo circulation persistence of 3 μm diameter phospholipid-coated MBs with three different mPEG2000 content: 2 mol % (mushroom), 5 mol % (intermediate), and 10 mol % (brush). MB size and concentration in the blood were evaluated by a hemocytometer analysis over 30 min following intravenous injections of 20 and 40 μL/kg MVD in Sprague-Dawley rats. Interestingly, pharmacokinetic analysis demonstrated that increasing PEG concentration on the MB surface resulted in faster clearance. This was evidenced by a 1.6-fold reduction in half-life and area under the curve (AUC) (p < 0.05) in the central compartment. Conversely, the AUC in the peripheral compartment increased with PEG density, suggesting enhanced MB trapping by the mononuclear phagocyte system. This was supported by an in vitro assay, which showed a significant rise in complement C3a activation with a higher PEG content. In conclusion, a minimal PEG concentration on the MB shell (mushroom configuration) was found to prolong circulation and mitigate immunogenicity.
Collapse
Affiliation(s)
- J Angel Navarro-Becerra
- Mechanical Engineering Department, University of Colorado Boulder, Boulder, Colorado 80309, United States
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Jair I Castillo
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Mark A Borden
- Mechanical Engineering Department, University of Colorado Boulder, Boulder, Colorado 80309, United States
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80309, United States
| |
Collapse
|
22
|
Roque JA, Lukesh NR, Hendy DA, Dixon TA, Islam MJ, Ontiveros-Padilla L, Pena ES, Lifshits LM, Simpson SR, Batty CJ, Bachelder EM, Ainslie KM. Enhancement of subunit vaccine delivery with zinc-carnosine coordination polymer through the addition of mannan. Int J Pharm 2024; 656:124076. [PMID: 38569976 PMCID: PMC11062752 DOI: 10.1016/j.ijpharm.2024.124076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/24/2024] [Accepted: 03/31/2024] [Indexed: 04/05/2024]
Abstract
Vaccines represent a pivotal health advancement for preventing infection. However, because carrier systems with repeated administration can invoke carrier-targeted immune responses that diminish subsequent immune responses (e.g., PEG antibodies), there is a continual need to develop novel vaccine platforms. Zinc carnosine microparticles (ZnCar MPs), which are composed of a one-dimensional coordination polymer formed between carnosine and the metal ion zinc, have exhibited efficacy in inducing an immune response against influenza. However, ZnCar MPs' limited suspendability hinders clinical application. In this study, we address this issue by mixing mannan, a polysaccharide derived from yeast, with ZnCar MPs. We show that the addition of mannan increases the suspendability of this promising vaccine formulation. Additionally, since mannan is an adjuvant, we illustrate that the addition of mannan increases the antibody response and T cell response when mixed with ZnCar MPs. Mice vaccinated with mannan + OVA/ZnCar MPs had elevated serum IgG and IgG1 levels in comparison to vaccination without mannan. Moreover, in the mannan + OVA/ZnCar MPs vaccinated group, mucosal washes demonstrated increased IgG, IgG1, and IgG2c titers, and antigen recall assays showed enhanced IFN-γ production in response to MHC-I and MHC-II immunodominant peptide restimulation, compared to the vaccination without mannan. These findings suggest that the use of mannan mixed with ZnCar MPs holds potential for subunit vaccination and its improved suspendability further promotes clinical translation.
Collapse
Affiliation(s)
- John A Roque
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Nicole Rose Lukesh
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Dylan A Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Timothy A Dixon
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Md Jahirul Islam
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Luis Ontiveros-Padilla
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Erik S Pena
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, United States
| | - Liubov M Lifshits
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Sean R Simpson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Cole J Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, United States; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
23
|
Gül D, Önal Acet B, Lu Q, Stauber RH, Odabaşı M, Acet Ö. Revolution in Cancer Treatment: How Are Intelligently Designed Nanostructures Changing the Game? Int J Mol Sci 2024; 25:5171. [PMID: 38791209 PMCID: PMC11120744 DOI: 10.3390/ijms25105171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Nanoparticles (NPs) are extremely important tools to overcome the limitations imposed by therapeutic agents and effectively overcome biological barriers. Smart designed/tuned nanostructures can be extremely effective for cancer treatment. The selection and design of nanostructures and the adjustment of size and surface properties are extremely important, especially for some precision treatments and drug delivery (DD). By designing specific methods, an important era can be opened in the biomedical field for personalized and precise treatment. Here, we focus on advances in the selection and design of nanostructures, as well as on how the structure and shape, size, charge, and surface properties of nanostructures in biological fluids (BFs) can be affected. We discussed the applications of specialized nanostructures in the therapy of head and neck cancer (HNC), which is a difficult and aggressive type of cancer to treat, to give an impetus for novel treatment approaches in this field. We also comprehensively touched on the shortcomings, current trends, and future perspectives when using nanostructures in the treatment of cancer.
Collapse
Affiliation(s)
- Désirée Gül
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (B.Ö.A.); (Q.L.); (R.H.S.)
| | - Burcu Önal Acet
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (B.Ö.A.); (Q.L.); (R.H.S.)
- Chemistry Department, Faculty of Arts and Science, Aksaray University, Aksaray 68100, Turkey;
| | - Qiang Lu
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (B.Ö.A.); (Q.L.); (R.H.S.)
| | - Roland H. Stauber
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (B.Ö.A.); (Q.L.); (R.H.S.)
| | - Mehmet Odabaşı
- Chemistry Department, Faculty of Arts and Science, Aksaray University, Aksaray 68100, Turkey;
| | - Ömür Acet
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (B.Ö.A.); (Q.L.); (R.H.S.)
- Pharmacy Services Program, Vocational School of Health Science, Tarsus University, Tarsus 33100, Turkey
| |
Collapse
|
24
|
Singh A, Lofts A, Krishnan R, Campea M, Chen L, Wan Y, Hoare T. The effect of comb length on the in vitro and in vivo properties of self-assembled poly(oligoethylene glycol methacrylate)-based block copolymer nanoparticles. NANOSCALE ADVANCES 2024; 6:2487-2498. [PMID: 38694467 PMCID: PMC11059560 DOI: 10.1039/d3na01156a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/19/2024] [Indexed: 05/04/2024]
Abstract
Comb copolymer analogues of poly(lactic acid)-polyethylene glycol block copolymers (PLA-b-PEG) offer potential to overcome the inherent chemistry and stability limitations of their linear block copolymer counterparts. Herein, we examine the differences between P(L)LA10K-b-PEG10K and linear-comb copolymer analogues thereof in which the linear PEG block is replaced by poly(oligo(ethylene glycol) methacrylate) (POEGMA) blocks with different side chain (comb) lengths but the same overall molecular weight. P(L)LA10K-b-POEGMA47510K and P(L)LA10K-b-POEGMA200010K block copolymers were synthesized via activators regenerated by electron transfer atom transfer radical polymerization (ARGET ATRP) and fabricated into self-assembled nanoparticles using flash nanoprecipitation via confined impinging jet mixing. Linear-comb copolymer analogues based on PLA-b-POEGMA yielded smaller but still well-controlled nanoparticle sizes (88 ± 2 nm and 114 ± 1 nm respectively compared to 159 ± 2 nm for P(L)LA10K-b-PEG10K nanoparticles) that exhibited improved colloidal stability relative to linear copolymer-based nanoparticles over a 15 day incubation period while maintaining comparably high cytocompatibility, although the comb copolymer analogues had somewhat lower loading capacity for doxorubicin hydrochloride. Cell spheroid studies showed that the linear-comb copolymers promoted enhanced tumor transport and thus cell killing compared to conventional linear block copolymers. In vivo studies showed all NP types could passively accumulate within implanted CT26 tumors but with different accumulation profiles, with P(L)LA10K-b-POEGMA200010K NPs showing continuous accumulation throughout the full 24 h monitoring period whereas tumor accumulation of P(L)LA10K-b-POEGMA47510K NPs was significant only between 8 h and 24 h. Overall, the linear-comb copolymer analogues exhibited superior stability, biodistribution, spheroid penetration, and inherent tunability over linear NP counterparts.
Collapse
Affiliation(s)
- Andrew Singh
- Department of Chemical Engineering, McMaster University 1280 Main St. W. Hamilton Ontario L8S 4L7 Canada
| | - Andrew Lofts
- Department of Chemical Engineering, McMaster University 1280 Main St. W. Hamilton Ontario L8S 4L7 Canada
| | - Ramya Krishnan
- Department of Pathology and Molecular Medicine, McMaster University 1280 Main St. W. Hamilton Ontario L8S 4L7 Canada
| | - Matthew Campea
- Department of Chemical Engineering, McMaster University 1280 Main St. W. Hamilton Ontario L8S 4L7 Canada
| | - Lan Chen
- Department of Pathology and Molecular Medicine, McMaster University 1280 Main St. W. Hamilton Ontario L8S 4L7 Canada
| | - Yonghong Wan
- Department of Pathology and Molecular Medicine, McMaster University 1280 Main St. W. Hamilton Ontario L8S 4L7 Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University 1280 Main St. W. Hamilton Ontario L8S 4L7 Canada
| |
Collapse
|
25
|
Zou Y, Kamoi K, Zong Y, Zhang J, Yang M, Ohno-Matsui K. Vaccines and the Eye: Current Understanding of the Molecular and Immunological Effects of Vaccination on the Eye. Int J Mol Sci 2024; 25:4755. [PMID: 38731972 PMCID: PMC11084287 DOI: 10.3390/ijms25094755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Vaccination is a public health cornerstone that protects against numerous infectious diseases. Despite its benefits, immunization implications on ocular health warrant thorough investigation, particularly in the context of vaccine-induced ocular inflammation. This review aimed to elucidate the complex interplay between vaccination and the eye, focusing on the molecular and immunological pathways implicated in vaccine-associated ocular adverse effects. Through an in-depth analysis of recent advancements and the existing literature, we explored various mechanisms of vaccine-induced ocular inflammation, such as direct infection by live attenuated vaccines, immune complex formation, adjuvant-induced autoimmunity, molecular mimicry, hypersensitivity reactions, PEG-induced allergic reactions, Type 1 IFN activation, free extracellular RNA, and specific components. We further examined the specific ocular conditions associated with vaccination, such as uveitis, optic neuritis, and retinitis, and discussed the potential impact of novel vaccines, including those against SARS-CoV-2. This review sheds light on the intricate relationships between vaccination, the immune system, and ocular tissues, offering insights into informed discussions and future research directions aimed at optimizing vaccine safety and ophthalmological care. Our analysis underscores the importance of vigilance and further research to understand and mitigate the ocular side effects of vaccines, thereby ensuring the continued success of vaccination programs, while preserving ocular health.
Collapse
Affiliation(s)
| | - Koju Kamoi
- Department of Ophthalmology and Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan; (Y.Z.); (Y.Z.); (J.Z.); (M.Y.); (K.O.-M.)
| | | | | | | | | |
Collapse
|
26
|
Go EB, Lee JH, Cho JH, Kwon NH, Choi JI, Kwon I. Enhanced therapeutic potential of antibody fragment via IEDDA-mediated site-specific albumin conjugation. J Biol Eng 2024; 18:23. [PMID: 38576037 PMCID: PMC10996255 DOI: 10.1186/s13036-024-00418-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND The use of single-chain variable fragments (scFvs) for treating human diseases, such as cancer and immune system disorders, has attracted significant attention. However, a critical drawback of scFv is its extremely short serum half-life, which limits its therapeutic potential. Thus, there is a critical need to prolong the serum half-life of the scFv for clinical applications. One promising serum half-life extender for therapeutic proteins is human serum albumin (HSA), which is the most abundant protein in human serum, known to have an exceptionally long serum half-life. However, conjugating a macromolecular half-life extender to a small protein, such as scFv, often results in a significant loss of its critical properties. RESULTS In this study, we conjugated the HSA to a permissive site of scFv to improve pharmacokinetic profiles. To ensure minimal damage to the antigen-binding capacity of scFv upon HSA conjugation, we employed a site-specific conjugation approach using a heterobifunctional crosslinker that facilitates thiol-maleimide reaction and inverse electron-demand Diels-Alder reaction (IEDDA). As a model protein, we selected 4D5scFv, derived from trastuzumab, a therapeutic antibody used in human epithermal growth factor 2 (HER2)-positive breast cancer treatment. We introduced a phenylalanine analog containing a very reactive tetrazine group (frTet) at conjugation site candidates predicted by computational methods. Using the linker TCO-PEG4-MAL, a single HSA molecule was site-specifically conjugated to the 4D5scFv (4D5scFv-HSA). The 4D5scFv-HSA conjugate exhibited HER2 binding affinity comparable to that of unmodified 4D5scFv. Furthermore, in pharmacokinetic profile in mice, the serum half-life of 4D5scFv-HSA was approximately 12 h, which is 85 times longer than that of 4D5scFv. CONCLUSIONS The antigen binding results and pharmacokinetic profile of 4D5scFv-HSA demonstrate that the site-specifically albumin-conjugated scFv retained its binding affinity with a prolonged serum half-life. In conclusion, we developed an effective strategy to prepare site-specifically albumin-conjugated 4D5scFv, which can have versatile clinical applications with improved efficacy.
Collapse
Affiliation(s)
- Eun Byeol Go
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jae Hun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jeong Haeng Cho
- ProAbTech, Gwangju, 61005, Republic of Korea
- Department of Biotechnology and Bioengineering, Interdisciplinary Program for Bioenergy and Biomaterials, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Na Hyun Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jong-Il Choi
- Department of Biotechnology and Bioengineering, Interdisciplinary Program for Bioenergy and Biomaterials, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Inchan Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
| |
Collapse
|
27
|
Prasad YR, Anakha J, Pande AH. Treating liver cancer through arginine depletion. Drug Discov Today 2024; 29:103940. [PMID: 38452923 DOI: 10.1016/j.drudis.2024.103940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/09/2024]
Abstract
Liver cancer, the sixth most common cancer globally and the second-leading cause of cancer-related deaths, presents a critical public health threat. Diagnosis often occurs in advanced stages of the disease, aligning incidence with fatality rates. Given that established treatments, such as stereotactic body radiation therapy and transarterial radioembolization, face accessibility and affordability challenges, the emerging focus on cancer cell metabolism, particularly arginine (Arg) depletion, offers a promising research avenue. Arg-depleting enzymes show efficacy against Arg-auxotrophic cancers, including hepatocellular carcinoma (HCC). Thus, in this review, we explore the limitations of current therapies and highlight the potential of Arg depletion, emphasizing various Arg-hydrolyzing enzymes in clinical development.
Collapse
Affiliation(s)
- Yenisetti Rajendra Prasad
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - J Anakha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
28
|
Yang C, Lin ZI, Zhang X, Xu Z, Xu G, Wang YM, Tsai TH, Cheng PW, Law WC, Yong KT, Chen CK. Recent Advances in Engineering Carriers for siRNA Delivery. Macromol Biosci 2024; 24:e2300362. [PMID: 38150293 DOI: 10.1002/mabi.202300362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/29/2023] [Indexed: 12/28/2023]
Abstract
RNA interference (RNAi) technology has been a promising treatment strategy for combating intractable diseases. However, the applications of RNAi in clinical are hampered by extracellular and intracellular barriers. To overcome these barriers, various siRNA delivery systems have been developed in the past two decades. The first approved RNAi therapeutic, Patisiran (ONPATTRO) using lipids as the carrier, for the treatment of amyloidosis is one of the most important milestones. This has greatly encouraged researchers to work on creating new functional siRNA carriers. In this review, the recent advances in siRNA carriers consisting of lipids, polymers, and polymer-modified inorganic particles for cancer therapy are summarized. Representative examples are presented to show the structural design of the carriers in order to overcome the delivery hurdles associated with RNAi therapies. Finally, the existing challenges and future perspective for developing RNAi as a clinical modality will be discussed and proposed. It is believed that the addressed contributions in this review will promote the development of siRNA delivery systems for future clinical applications.
Collapse
Affiliation(s)
- Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Zheng-Ian Lin
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Xinmeng Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Zhourui Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Yu-Min Wang
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Tzu-Hsien Tsai
- Division of Cardiology and Department of Internal Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi, 60002, Taiwan
| | - Pei-Wen Cheng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81362, Taiwan
- Department of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Wing-Cheung Law
- Department of Industrial and Systems Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, P. R. China
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Chih-Kuang Chen
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| |
Collapse
|
29
|
Brigitha LJ, Mondelaers V, Liu Y, Albertsen BK, Zalewska-Szewczyk B, Rizzari C, Kotecha RS, Pieters R, Huitema ADR, van der Sluis IM. Pharmacokinetics of PEGasparaginase in Infants with Acute Lymphoblastic Leukemia. Pharm Res 2024; 41:711-720. [PMID: 38538970 DOI: 10.1007/s11095-024-03693-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/14/2024] [Indexed: 04/18/2024]
Abstract
BACKGROUND PEGasparaginase is known to be a critical drug for treating pediatric acute lymphoblastic leukemia (ALL), however, there is insufficient evidence to determine the optimal dose for infants who are less than one year of age at diagnosis. This international study was conducted to identify the pharmacokinetics of PEGasparaginase in infants with newly diagnosed ALL and gather insight into the clearance and dosing of this population. METHODS Infants with ALL who received treatment with PEGasparaginase were included in our population pharmacokinetic assessment employing non-linear mixed effects modelling (NONMEM). RESULTS 68 infants with ALL, with a total of 388 asparaginase activity samples, were included. PEGasparaginase doses ranging from 400 to 3,663 IU/m2 were administered either intravenously or intramuscularly. A one-compartment model with time-dependent clearance, modeled using a transit model, provided the best fit to the data. Body weight was significantly correlated with clearance and volume of distribution. The final model estimated a half-life of 11.7 days just after administration, which decreased to 1.8 days 14 days after administration. Clearance was 19.5% lower during the post-induction treatment phase compared to induction. CONCLUSION The pharmacokinetics of PEGasparaginase in infants diagnosed under one year of age with ALL is comparable to that of older children (1-18 years). We recommend a PEGasparaginase dosing at 1,500 IU/m2 for infants without dose adaptations according to age, and implementing therapeutic drug monitoring as standard practice.
Collapse
Affiliation(s)
- Leiah J Brigitha
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, Netherlands
- Pediatric Oncology and Hematology, Erasmus MC-Sophia Children's Hospital, Dr. Molewaterplein 40, 3015 GD, Rotterdam, Netherlands
| | - Veerle Mondelaers
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Yiwei Liu
- Department of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston, USA
| | - Birgitte K Albertsen
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Palle Juul-Jensens Blvd. 99, 8200, Aarhus, Denmark
| | - Beata Zalewska-Szewczyk
- Department of Pediatrics, Medical University of Lodz, Oncology & Hematology, 91-738, Lodz, Poland
| | - Carmelo Rizzari
- Department of Pediatrics, University of Milano-Bicocca, Piazza Dell'Ateneo Nuovo, 1, Milano, Italy
- Fondazione IRCCS San Gerardo Dei Tintori, Via G.B. Pergolesi 33, Monza, Italy
| | - Rishi S Kotecha
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, Australia
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia
- Curtin Medical School, Curtin University, Perth, Australia
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, Netherlands
| | - Alwin D R Huitema
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Inge M van der Sluis
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, Netherlands.
| |
Collapse
|
30
|
Bitounis D, Jacquinet E, Rogers MA, Amiji MM. Strategies to reduce the risks of mRNA drug and vaccine toxicity. Nat Rev Drug Discov 2024; 23:281-300. [PMID: 38263456 DOI: 10.1038/s41573-023-00859-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/25/2024]
Abstract
mRNA formulated with lipid nanoparticles is a transformative technology that has enabled the rapid development and administration of billions of coronavirus disease 2019 (COVID-19) vaccine doses worldwide. However, avoiding unacceptable toxicity with mRNA drugs and vaccines presents challenges. Lipid nanoparticle structural components, production methods, route of administration and proteins produced from complexed mRNAs all present toxicity concerns. Here, we discuss these concerns, specifically how cell tropism and tissue distribution of mRNA and lipid nanoparticles can lead to toxicity, and their possible reactogenicity. We focus on adverse events from mRNA applications for protein replacement and gene editing therapies as well as vaccines, tracing common biochemical and cellular pathways. The potential and limitations of existing models and tools used to screen for on-target efficacy and de-risk off-target toxicity, including in vivo and next-generation in vitro models, are also discussed.
Collapse
Affiliation(s)
- Dimitrios Bitounis
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
- Moderna, Inc., Cambridge, MA, USA
| | | | | | - Mansoor M Amiji
- Departments of Pharmaceutical Sciences and Chemical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
31
|
Alghamdi Y, Abdulghani F, Huwait HF, Abdulghani M, Samarkandy SJ. An Unusual Presentation of Erythema Multiforme Following the Administration of Pfizer-BioNTech COVID-19 mRNA Vaccine in a Pediatric Patient. Cureus 2024; 16:e58450. [PMID: 38765345 PMCID: PMC11100275 DOI: 10.7759/cureus.58450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2024] [Indexed: 05/22/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused a global calamity that forced emergency use authorization to Pfizer-BioNTech COVID-19 (BNT162b2) vaccine. It is efficacious in preventing symptomatic severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection in seronegative recipients. The safety profile is still unclear; however, commonly reported symptoms post-vaccination are fatigue, headache, muscle pain, chills, and injection-site pain. COVID-19 disease elicits, to some extent, cutaneous side effects like urticaria, morbilliform rash, and chilblain-like eruption. Vaccination against COVID-19 was reported to induce similar dermatologic manifestations, such as urticarial rash, delayed large-local reaction, local injection-site reaction, and morbilliform eruption. Erythema multiforme (EM) is a rare manifestation post-vaccination, and only a few reports implicate it as a culprit in cutaneous eruptions following the BNT162b2 vaccine. This report delineates the presentation of a healthy 14-year-old girl to a dermatology clinic who developed EM post-vaccination with the first dose of BNT162b2. New-onset EM-eruption post-vaccination with BNT162b2 had been reported previously in 14 cases, and one case reported on the flare of preexisting-EM.
Collapse
Affiliation(s)
- Yara Alghamdi
- Dermatology, King Abdullah Medical Complex, Ministry of Health, Jeddah, SAU
| | | | | | | | - Sahal J Samarkandy
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, SAU
- Dermatology, King Abdulaziz Medical City (KAMC), Jeddah, SAU
| |
Collapse
|
32
|
Berger M, Toussaint F, Ben Djemaa S, Maquoi E, Pendeville H, Evrard B, Jerôme C, Leblond Chain J, Lechanteur A, Mottet D, Debuigne A, Piel G. Poly(N-methyl-N-vinylacetamide): A Strong Alternative to PEG for Lipid-Based Nanocarriers Delivering siRNA. Adv Healthc Mater 2024; 13:e2302712. [PMID: 37994483 DOI: 10.1002/adhm.202302712] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/09/2023] [Indexed: 11/24/2023]
Abstract
Lipid-based nanocarriers have demonstrated high interest in delivering genetic material, exemplified by the success of Onpattro and COVID-19 vaccines. While PEGylation imparts stealth properties, it hampers cellular uptake and endosomal escape, and may trigger adverse reactions like accelerated blood clearance (ABC) and hypersensitivity reactions (HSR). This work highlights the great potential of amphiphilic poly(N-methyl-N-vinylacetamide) (PNMVA) derivatives as alternatives to lipid-PEG for siRNA delivery. PNMVA compounds with different degrees of polymerization and hydrophobic segments, are synthesized. Among them, DSPE (1,2-distearoyl-sn-glycero-3-phosphoethanolamine)-PNMVA efficiently integrates into lipoplexes and LNP membranes and prevents protein corona formation around these lipid carriers, exhibiting stealth properties comparable to DSPE-PEG. However, unlike DSPE-PEG, DSPE-PNMVA24 shows no adverse impact on lipoplexes cell uptake and endosomal escape. In in vivo study with mice, DSPE-PNMVA24 lipoplexes demonstrate no liver accumulation, indicating good stealth properties, extended circulation time after a second dose, reduced immunological reaction, and no systemic pro-inflammatory response. Safety of DSPE-PNMVA24 is confirmed at the cellular level and in animal models of zebrafish and mice. Overall, DSPE-PNMVA is an advantageous substitute to DSPE-PEG for siRNA delivery, offering comparable stealth and toxicity properties while improving efficacy of the lipid-based carriers by minimizing the dilemma effect and reducing immunological reactions, meaning no ABC or HSR effects.
Collapse
Affiliation(s)
- Manon Berger
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liège, Avenue Hippocrate 15, Liège, 4000, Belgium
| | - François Toussaint
- Center for Education and Research on Macromolecules CERM, CESAM Research Unit, University of Liège, Allée du Six Août, 13, Liège, 4000, Belgium
| | - Sanaa Ben Djemaa
- Gene Expression and Cancer Laboratory GEC, GIGA-Molecular Biology of Diseases, University of Liège, Avenue de l'Hôpital 11, Liège, 4000, Belgium
| | - Erik Maquoi
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Avenue Hippocrate, 13, Liège, 4000, Belgium
| | - Hélène Pendeville
- Platform Zebrafish Facility and Transgenics, GIGA, University of Liège, Avenue de l'Hôpital 11, Liège, 4000, Belgium
| | - Brigitte Evrard
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liège, Avenue Hippocrate 15, Liège, 4000, Belgium
| | - Christine Jerôme
- Center for Education and Research on Macromolecules CERM, CESAM Research Unit, University of Liège, Allée du Six Août, 13, Liège, 4000, Belgium
| | - Jeanne Leblond Chain
- University of Bordeaux, CNRS, INSERM, ARNA, UMR 5320, U1212, 146 rue Léo Saignat, Bordeaux, F-33000, France
| | - Anna Lechanteur
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liège, Avenue Hippocrate 15, Liège, 4000, Belgium
| | - Denis Mottet
- Gene Expression and Cancer Laboratory GEC, GIGA-Molecular Biology of Diseases, University of Liège, Avenue de l'Hôpital 11, Liège, 4000, Belgium
| | - Antoine Debuigne
- Center for Education and Research on Macromolecules CERM, CESAM Research Unit, University of Liège, Allée du Six Août, 13, Liège, 4000, Belgium
| | - Géraldine Piel
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liège, Avenue Hippocrate 15, Liège, 4000, Belgium
| |
Collapse
|
33
|
Jeong EJ, Kim C, Lee YC, Rhim T, Lee SK, Lee KY. Tumor-specific cytolysis by peptide-conjugated echogenic polymer micelles. Biomed Pharmacother 2024; 172:116272. [PMID: 38354570 DOI: 10.1016/j.biopha.2024.116272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/16/2024] Open
Abstract
Interest in multifunctional polymer nanoparticles for targeted delivery of anti-cancer drugs has grown significantly in recent years. In this study, tumor-targeting echogenic polymer micelles were prepared from poly(ethylene glycol) methyl ether-alkyl carbonate (mPEG-AC) derivatives, and their potential in cancer therapy was assessed. Various mPEG derivatives with carbonate linkages were synthesized via an alkyl halide reaction between mPEG and alkyl chloroformate. Micelle formation using polymer amphiphiles in aqueous media and the subsequent carbon dioxide (CO2) gas generation from the micelles was confirmed. Their ability to target neuroblastoma was substantially enhanced by incorporating the rabies virus glycoprotein (RVG) peptide. RVG-modified gas-generating micelles significantly inhibited tumor growth in a tumor-bearing mouse model owing to CO2 gas generation within tumor cells and resultant cytolytic effects, showing minimal side effects. The development of multifunctional polymer micelles may offer a promising therapeutic approach for various diseases, including cancer.
Collapse
Affiliation(s)
- Eun Ju Jeong
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea
| | - Choonggu Kim
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea
| | - Yun-Chan Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea
| | - Taiyoun Rhim
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea; Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul 04763, the Republic of Korea.
| | - Sang-Kyung Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea; Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul 04763, the Republic of Korea.
| | - Kuen Yong Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea; Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul 04763, the Republic of Korea.
| |
Collapse
|
34
|
Yu YF, Wu EC, Lin SQ, Chu YX, Yang Y, Pan F, Ding TH, Qian J, Jiang K, Zhan CY. Reexamining the effects of drug loading on the in vivo performance of PEGylated liposomal doxorubicin. Acta Pharmacol Sin 2024; 45:646-659. [PMID: 37845342 PMCID: PMC10834505 DOI: 10.1038/s41401-023-01169-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/13/2023] [Indexed: 10/18/2023] Open
Abstract
Higher drug loading employed in nanoscale delivery platforms is a goal that researchers have long sought after. But such viewpoint remains controversial because the impacts that nanocarriers bring about on bodies have been seriously overlooked. In the present study we investigated the effects of drug loading on the in vivo performance of PEGylated liposomal doxorubicin (PLD). We prepared PLDs with two different drug loading rates: high drug loading rate, H-Dox, 12.9% w/w Dox/HSPC; low drug loading rate, L-Dox, 2.4% w/w Dox/HSPC (L-Dox had about 5 folds drug carriers of H-Dox at the same Dox dose). The pharmaceutical properties and biological effects of H-Dox and L-Dox were compared in mice, rats or 4T1 subcutaneous tumor-bearing mice. We showed that the lowering of doxorubicin loading did not cause substantial shifts to the pharmaceutical properties of PLDs such as in vitro and in vivo stability (stable), anti-tumor effect (equivalent effective), as well as tissue and cellular distribution. Moreover, it was even more beneficial for mitigating the undesired biological effects caused by PLDs, through prolonging blood circulation and alleviating cutaneous accumulation in the presence of pre-existing anti-PEG Abs due to less opsonins (e.g. IgM and C3) deposition on per particle. Our results warn that the effects of drug loading would be much more convoluted than expected due to the complex intermediation between nanocarriers and bodies, urging independent investigation for each individual delivery platform to facilitate clinical translation and application.
Collapse
Affiliation(s)
- Yi-Fei Yu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Er-Can Wu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China
| | - Shi-Qi Lin
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Yu-Xiu Chu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Yang Yang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Feng Pan
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China
| | - Tian-Hao Ding
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Jun Qian
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China.
| | - Kuan Jiang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China.
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China.
| | - Chang-You Zhan
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China.
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China.
| |
Collapse
|
35
|
Gaballa SA, Shimizu T, Ando H, Takata H, Emam SE, Ramadan E, Naguib YW, Mady FM, Khaled KA, Ishida T. Treatment-induced and Pre-existing Anti-peg Antibodies: Prevalence, Clinical Implications, and Future Perspectives. J Pharm Sci 2024; 113:555-578. [PMID: 37931786 DOI: 10.1016/j.xphs.2023.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Polyethylene glycol (PEG) is a versatile polymer that is used in numerous pharmaceutical applications like the food industry, a wide range of disinfectants, cosmetics, and many commonly used household products. PEGylation is the term used to describe the covalent attachment of PEG molecules to nanocarriers, proteins and peptides, and it is used to prolong the circulation half-life of the PEGylated products. Consequently, PEGylation improves the efficacy of PEGylated therapeutics. However, after four decades of research and more than two decades of clinical applications, an unappealing side of PEGylation has emerged. PEG immunogenicity and antigenicity are remarkable challenges that confound the widespread clinical application of PEGylated therapeutics - even those under clinical trials - as anti-PEG antibodies (Abs) are commonly reported following the systemic administration of PEGylated therapeutics. Furthermore, pre-existing anti-PEG Abs have also been reported in healthy individuals who have never been treated with PEGylated therapeutics. The circulating anti-PEG Abs, both treatment-induced and pre-existing, selectively bind to PEG molecules of the administered PEGylated therapeutics inducing activation of the complement system, which results in remarkable clinical implications with varying severity. These include increased blood clearance of the administered PEGylated therapeutics through what is known as the accelerated blood clearance (ABC) phenomenon and initiation of serious adverse effects through complement activation-related pseudoallergic reactions (CARPA). Therefore, the US FDA industry guidelines have recommended the screening of anti-PEG Abs, in addition to Abs against PEGylated proteins, in the clinical trials of PEGylated protein therapeutics. In addition, strategies revoking the immunogenic response against PEGylated therapeutics without compromising their therapeutic efficacy are important for the further development of advanced PEGylated therapeutics and drug-delivery systems.
Collapse
Affiliation(s)
- Sherif A Gaballa
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Sherif E Emam
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig, 44519 Egypt
| | - Eslam Ramadan
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Youssef W Naguib
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Fatma M Mady
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Khaled A Khaled
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan.
| |
Collapse
|
36
|
Kane G, Lusi C, Brassil M, Atukorale P. Engineering approaches for innate immune-mediated tumor microenvironment remodeling. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 21:100406. [PMID: 38213392 PMCID: PMC10777078 DOI: 10.1016/j.iotech.2023.100406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Cancer immunotherapy offers transformative promise particularly for the treatment of lethal cancers, since a correctly trained immune system can comprehensively orchestrate tumor clearance with no need for continued therapeutic intervention. Historically, the majority of immunotherapies have been T cell-focused and have included immune checkpoint inhibitors, chimeric antigen receptor T cells, and T-cell vaccines. Unfortunately T-cell-focused therapies have failed to achieve optimal efficacy in most solid tumors largely because of a highly immunosuppressed 'cold' or immune-excluded tumor microenvironment (TME). Recently, a rapidly growing treatment paradigm has emerged that focuses on activation of tumor-resident innate antigen-presenting cells, such as dendritic cells and macrophages, which can drive a proinflammatory immune response to remodel the TME from 'cold' or immune-excluded to 'hot'. Early strategies for TME remodeling centered on free cytokines and agonists, but these approaches have faced significant hurdles in both delivery and efficacy. Systemic toxicity from off-target inflammation is a paramount concern in these therapies. To address this critical gap, engineering approaches have provided the opportunity to add 'built-in' capabilities to cytokines, agonists, and other therapeutic agents to mediate improved delivery and efficacy. Such capabilities have included protective encapsulation to shield them from degradation, targeting to direct them with high specificity to tumors, and co-delivery strategies to harness synergistic proinflammatory pathways. Here, we review innate immune-mediated TME remodeling engineering approaches that focus on cytokines, innate immune agonists, immunogenic viruses, and cell-based methods, highlighting emerging preclinical approaches and strategies that are either being tested in clinical trials or already Food and Drug Administration approved.
Collapse
Affiliation(s)
- G.I. Kane
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - C.F. Lusi
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - M.L. Brassil
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - P.U. Atukorale
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, USA
| |
Collapse
|
37
|
Eweje F, Walsh ML, Ahmad K, Ibrahim V, Alrefai A, Chen J, Chaikof EL. Protein-based nanoparticles for therapeutic nucleic acid delivery. Biomaterials 2024; 305:122464. [PMID: 38181574 PMCID: PMC10872380 DOI: 10.1016/j.biomaterials.2023.122464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/25/2023] [Accepted: 12/31/2023] [Indexed: 01/07/2024]
Abstract
To realize the full potential of emerging nucleic acid therapies, there is a need for effective delivery agents to transport cargo to cells of interest. Protein materials exhibit several unique properties, including biodegradability, biocompatibility, ease of functionalization via recombinant and chemical modifications, among other features, which establish a promising basis for therapeutic nucleic acid delivery systems. In this review, we highlight progress made in the use of non-viral protein-based nanoparticles for nucleic acid delivery in vitro and in vivo, while elaborating on key physicochemical properties that have enabled the use of these materials for nanoparticle formulation and drug delivery. To conclude, we comment on the prospects and unresolved challenges associated with the translation of protein-based nucleic acid delivery systems for therapeutic applications.
Collapse
Affiliation(s)
- Feyisayo Eweje
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Harvard and MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Harvard/MIT MD-PhD Program, Boston, MA, USA, 02115; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Michelle L Walsh
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Harvard and MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Harvard/MIT MD-PhD Program, Boston, MA, USA, 02115
| | - Kiran Ahmad
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Vanessa Ibrahim
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Assma Alrefai
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Jiaxuan Chen
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| | - Elliot L Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| |
Collapse
|
38
|
Paun RA, Jurchuk S, Tabrizian M. A landscape of recent advances in lipid nanoparticles and their translational potential for the treatment of solid tumors. Bioeng Transl Med 2024; 9:e10601. [PMID: 38435821 PMCID: PMC10905562 DOI: 10.1002/btm2.10601] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 03/05/2024] Open
Abstract
Lipid nanoparticles (LNPs) are biocompatible drug delivery systems that have found numerous applications in medicine. Their versatile nature enables the encapsulation and targeting of various types of medically relevant molecular cargo, including oligonucleotides, proteins, and small molecules for the treatment of diseases, such as cancer. Cancers that form solid tumors are particularly relevant for LNP-based therapeutics due to the enhanced permeation and retention effect that allows nanoparticles to accumulate within the tumor tissue. Additionally, LNPs can be formulated for both locoregional and systemic delivery depending on the tumor type and stage. To date, LNPs have been used extensively in the clinic to reduce systemic toxicity and improve outcomes in cancer patients by encapsulating chemotherapeutic drugs. Next-generation lipid nanoparticles are currently being developed to expand their use in gene therapy and immunotherapy, as well as to enable the co-encapsulation of multiple drugs in a single system. Other developments include the design of targeted LNPs to specific cells and tissues, and triggerable release systems to control cargo delivery at the tumor site. This review paper highlights recent developments in LNP drug delivery formulations and focuses on the treatment of solid tumors, while also discussing some of their current translational limitations and potential opportunities in the field.
Collapse
Affiliation(s)
- Radu A. Paun
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Sarah Jurchuk
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Faculty of Dentistry and Oral Health SciencesMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
39
|
Davis E, Caparco AA, Jones E, Steinmetz NF, Pokorski JK. Study of uricase-polynorbornene conjugates derived from grafting-from ring-opening metathesis polymerization. J Mater Chem B 2024; 12:2197-2206. [PMID: 38323642 DOI: 10.1039/d3tb02726k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
PEGylation has been the 'gold standard' in bioconjugation due to its ability to improve the pharmacokinetics and pharmacodynamics of native proteins. However, growing clinical evidence of hypersensitivity reactions to PEG due to pre-existing anti-PEG antibodies in healthy humans have raised concerns. Advancements in controlled polymerization techniques and conjugation chemistries have paved the way for the development of protein-polymer conjugates that can circumvent these adverse reactions while retaining the benefits of such modifications. Herein, we show the development of polynorbornene based bioconjugates of therapeutically relevant urate oxidase (UO) enzymes used in the treatment of gout synthesized by grafting-from ring-opening metathesis polymerization (ROMP). Notably, these conjugates exhibit comparable levels of bioactivity to PEGylated UO and demonstrate increased stability across varying temperatures and pH conditions. Immune recognition of conjugates by anti-UO antibodies reveal low protein immunogenicity following the conjugation process. Additionally, UO conjugates employing zwitterionic polynorbornene successfully avoid recognition by anti-PEG antibodies, further highlighting a potential replacement for PEG.
Collapse
Affiliation(s)
- Elizabathe Davis
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA.
| | - Adam A Caparco
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Elizabeth Jones
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA.
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA.
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, CA 92093, USA
- Institute for Materials Discovery and Design, University of California San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Jonathan K Pokorski
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA.
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, CA 92093, USA
- Institute for Materials Discovery and Design, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
40
|
Davis E, Caparco AA, Steinmetz NF, Pokorski JK. Poly(Oxanorbornene)-Protein Conjugates Prepared by Grafting-to ROMP as Alternatives for PEG. Macromol Biosci 2024; 24:e2300255. [PMID: 37688508 DOI: 10.1002/mabi.202300255] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/14/2023] [Indexed: 09/11/2023]
Abstract
PEGylation is the gold standard in protein-polymer conjugation, improving circulation half-life of biologics while mitigating the immune response to a foreign substance. However, preexisting anti-PEG antibodies in healthy humans are becoming increasingly prevalent and elicitation of anti-PEG antibodies when patients are administered with PEGylated therapeutics challenges their safety profile. In the current study, two distinct amine-reactive poly(oxanorbornene) (PONB) imide-based water-soluble block co-polymers are synthesized using ring-opening metathesis polymerization (ROMP). The synthesized block-copolymers include PEG-based PONB-PEG and sulfobetaine-based PONB-Zwit. The polymers are then covalently conjugated to amine residues of lysozyme (Lyz) and urate oxidase (UO) using a grafting-to bioconjugation technique. Both Lyz-PONB and UO-PONB conjugates retained significant bioactivities after bioconjugation. Immune recognition studies of UO-PONB conjugates indicated a comparable lowering of protein immunogenicity when compared to PEGylated UO. PEG-specific immune recognition is negligible for UO-PONB-Zwit conjugates, as expected. These polymers provide a new alternative for PEG-based systems that retain high levels of activity for the biologic while showing improved immune recognition profiles.
Collapse
Affiliation(s)
- Elizabathe Davis
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Adam A Caparco
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jonathan K Pokorski
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
41
|
Oliveira Silva R, Counil H, Rabanel JM, Haddad M, Zaouter C, Ben Khedher MR, Patten SA, Ramassamy C. Donepezil-Loaded Nanocarriers for the Treatment of Alzheimer's Disease: Superior Efficacy of Extracellular Vesicles Over Polymeric Nanoparticles. Int J Nanomedicine 2024; 19:1077-1096. [PMID: 38317848 PMCID: PMC10843980 DOI: 10.2147/ijn.s449227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/07/2024] [Indexed: 02/07/2024] Open
Abstract
Introduction Drug delivery across the blood-brain barrier (BBB) is challenging and therefore severely restricts neurodegenerative diseases therapy such as Alzheimer's disease (AD). Donepezil (DNZ) is an acetylcholinesterase (AChE) inhibitor largely prescribed to AD patients, but its use is limited due to peripheral adverse events. Nanodelivery strategies with the polymer Poly (lactic acid)-poly(ethylene glycol)-based nanoparticles (NPs-PLA-PEG) and the extracellular vesicles (EVs) were developed with the aim to improve the ability of DNZ to cross the BBB, its brain targeting and efficacy. Methods EVs were isolated from human plasma and PLA-PEG NPs were synthesized by nanoprecipitation. The toxicity, brain targeting capacity and cholinergic activities of the formulations were evaluated both in vitro and in vivo. Results EVs and NPs-PLA-PEG were designed to be similar in size and charge, efficiently encapsulated DNZ and allowed sustained drug release. In vitro study showed that both formulations EVs-DNZ and NPs-PLA-PEG-DNZ were highly internalized by the endothelial cells bEnd.3. These cells cultured on the Transwell® model were used to analyze the transcytosis of both formulations after validation of the presence of tight junctions, the transendothelial electrical resistance (TEER) values and the permeability of the Dextran-FITC. In vivo study showed that both formulations were not toxic to zebrafish larvae (Danio rerio). However, hyperactivity was evidenced in the NPs-PLA-PEG-DNZ and free DNZ groups but not the EVs-DNZ formulations. Biodistribution analysis in zebrafish larvae showed that EVs were present in the brain parenchyma, while NPs-PLA-PEG remained mainly in the bloodstream. Conclusion The EVs-DNZ formulation was more efficient to inhibit the AChE enzyme activity in the zebrafish larvae head. Thus, the bioinspired delivery system (EVs) is a promising alternative strategy for brain-targeted delivery by substantially improving the activity of DNZ for the treatment of AD.
Collapse
Affiliation(s)
- Rummenigge Oliveira Silva
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Hermine Counil
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | | | - Mohamed Haddad
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Charlotte Zaouter
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Mohamed Raâfet Ben Khedher
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
- Higher Institute of Biotechnology of Beja, University of Jendouba, Beja, Tunisia
| | - Shunmoogum A Patten
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Charles Ramassamy
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| |
Collapse
|
42
|
Li Z, Ma A, Miller I, Starnes R, Talkington A, Stone CA, Phillips EJ, Choudhary SK, Commins SP, Lai SK. Development of anti-PEG IgG/IgM/IgE ELISA assays for profiling anti-PEG immunoglobulin response in PEG-sensitized individuals and patients with alpha-gal allergy. J Control Release 2024; 366:342-348. [PMID: 38182056 PMCID: PMC11182197 DOI: 10.1016/j.jconrel.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024]
Abstract
Polyethylene glycol (PEG) is frequently used in various protein and nanomedicine therapeutics. However, various studies have shown that select PEGylated therapeutics can induce production of anti-PEG antibodies (APA), potentially culminating in rapid clearance from the systemic circulation, loss of efficacy and possibly increased risks of allergic reactions. Although IgE is a frequent cause of immediate hypersensitivity reactions (IHR), the role of IgE APA in PEG-related IHR is not well understood, due in part to a lack of standardized assays for measuring IgE APA. Here, we developed a rigorous competitive ELISA method to measure the concentrations of various APA isotypes, including IgE, with picomolar sensitivities. In a small number of serum samples from patients with known PEG allergy, the assay allowed us to detect a strong correlation between IgG and IgE APA in individuals with history of allergic reactions to PEG or PEGylated drugs, but not between IgM and IgE APA. We detected appreciable levels of IgG and IgM APA in individuals with history of alpha-gal allergy, however, they were not elevated relative to those detected in other healthy controls, and we found no pre-existing IgE APA. While preliminary and should be further investigated, these results suggest that differences in the route and mechanism of PEG exposure may drive variability in APA response.
Collapse
Affiliation(s)
- Zhongbo Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Alice Ma
- Department of Biomedical Engineering, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Ian Miller
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Rachel Starnes
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Anne Talkington
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, USA
| | - Cosby A Stone
- Center for Drug Safety and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth J Phillips
- Center for Drug Safety and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shailesh K Choudhary
- Division of Allergy and Immunology, School of Medicine, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Scott P Commins
- Division of Allergy and Immunology, School of Medicine, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Samuel K Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA; Department of Biomedical Engineering, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA; Department of Immunology and Microbiology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
43
|
Yang M, Zhang Z, Jin P, Jiang K, Xu Y, Pan F, Tian K, Yuan Z, Liu XE, Fu J, Wang B, Yan H, Zhan C, Zhang Z. Effects of PEG antibodies on in vivo performance of LNP-mRNA vaccines. Int J Pharm 2024; 650:123695. [PMID: 38081560 DOI: 10.1016/j.ijpharm.2023.123695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 12/22/2023]
Abstract
Polyethylene glycol (PEG) plays important roles in stabilizing and lengthening circulation time of lipid nanoparticle (LNP) vaccines. Nowadays various levels of PEG antibodies have been detected in human blood, but the impact and mechanism of PEG antibodies on the in vivo performance of LNP vaccines has not been clarified thoroughly. By illustrating the distribution characteristics of PEG antibodies in human, the present study focused on the influence of PEG antibodies on the safety and efficacy of LNP-mRNA vaccine against COVID-19 in animal models. It was found that PEG antibodies led to shortened blood circulation duration, elevated accumulation and mRNA expression in liver and spleen, enhanced expression in macrophage and dendritic cells, while without affecting the production of anti-Spike protein antibodies of COVID-19 LNP vaccine. Noteworthily, PEG antibodies binding on the LNP vaccine increased probability of complement activation in animal as well as in human serum and led to lethal side effect in large dosage via intravenous injection of mice. Our data suggested that PEG antibodies in human was a risky factor of LNP-based vaccines for biosafety concerns but not efficacy.
Collapse
Affiliation(s)
- Min Yang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China
| | - Zengyu Zhang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China
| | - Pengpeng Jin
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China; Department of Chronic Disease Management, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, PR China
| | - Kuan Jiang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China; Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai 200031, PR China
| | - Yifei Xu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 PR China
| | - Feng Pan
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, PR China
| | - Kaisong Tian
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China
| | - Zhou Yuan
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China
| | | | - Jiaru Fu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200032, PR China
| | - Bin Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200032, PR China
| | - Huafang Yan
- Department of Health Management, Pudong Hospital, Fudan University, Shanghai 201399, PR China
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China; State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 PR China; Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai 200032, PR China.
| | - Zui Zhang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China.
| |
Collapse
|
44
|
Arduino I, Di Fonte R, Tiboni M, Porcelli L, Serratì S, Fondaj D, Rafaschieri T, Cutrignelli A, Guida G, Casettari L, Azzariti A, Lopedota AA, Denora N, Iacobazzi RM. Microfluidic development and biological evaluation of targeted therapy-loaded biomimetic nano system to improve the metastatic melanoma treatment. Int J Pharm 2024; 650:123697. [PMID: 38081557 DOI: 10.1016/j.ijpharm.2023.123697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Optimizing current therapies is among next steps in metastatic melanoma (MM) treatment landscape. The innovation of this study is the design of production process by microfluidics of cell membrane (CM)-modified nanoparticles (NPs), as an emerging biomimetic platform that allows for reduced immune clearance, long blood circulation time and improved specific tumor targeting. To achieve melanoma selectivity, direct membrane fusion between synthetic liposomes and CMs extracted from MM cell line was performed by microfluidic sonication approach, then the hybrid liposomes were loaded with cobimetinib (Cob) or lenvatinib (Lenva) targeting agents and challenged against MM cell lines and liver cancer cell line to evaluate homotypic targeting and antitumor efficacy. Characterization studies demonstrated the effective fusion of CM with liposome and the high encapsulation efficiency of both drugs, showing the proficiency of microfluidic-based production. By studying the targeting of melanoma cells by hybrid liposomes versus liposomes, we found that both NPs entered cells through endocytosis, whereas the former showed higher selectivity for MM cells from which CM was extracted, with 8-fold higher cellular uptake than liposomes. Hybrid liposome formulation of Cob and Lenva reduced melanoma cells viability to a greater extent than liposomes and free drug and, notably, showed negligible toxicity as demonstrated by bona fide haemolysis test. The CM-modified NPs presented here have the potential to broaden the choice of therapeutic options in MM treatment.
Collapse
Affiliation(s)
- Ilaria Arduino
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy
| | | | - Mattia Tiboni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento 6, 61029 Urbino, Italy
| | | | - Simona Serratì
- IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy
| | - Dafina Fondaj
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy
| | | | - Annalisa Cutrignelli
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy
| | - Gabriella Guida
- Department of Traslational Biomedicine and Neuroscience (DiBraiN), School of Medicine, University of Bari "A. Moro", 70124 Bari, Italy
| | - Luca Casettari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento 6, 61029 Urbino, Italy
| | - Amalia Azzariti
- IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy.
| | | | - Nunzio Denora
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy
| | - Rosa Maria Iacobazzi
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy.
| |
Collapse
|
45
|
Graván P, Peña-Martín J, de Andrés JL, Pedrosa M, Villegas-Montoya M, Galisteo-González F, Marchal JA, Sánchez-Moreno P. Exploring the Impact of Nanoparticle Stealth Coatings in Cancer Models: From PEGylation to Cell Membrane-Coating Nanotechnology. ACS APPLIED MATERIALS & INTERFACES 2024; 16:2058-2074. [PMID: 38159050 PMCID: PMC10797597 DOI: 10.1021/acsami.3c13948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
Nanotechnological platforms offer advantages over conventional therapeutic and diagnostic modalities. However, the efficient biointerfacing of nanomaterials for biomedical applications remains challenging. In recent years, nanoparticles (NPs) with different coatings have been developed to reduce nonspecific interactions, prolong circulation time, and improve therapeutic outcomes. This study aims to compare various NP coatings to enhance surface engineering for more effective nanomedicines. We prepared and characterized polystyrene NPs with different coatings of poly(ethylene glycol), bovine serum albumin, chitosan, and cell membranes from a human breast cancer cell line. The coating was found to affect the colloidal stability, adhesion, and elastic modulus of NPs. Protein corona formation and cellular uptake of NPs were also investigated, and a 3D tumor model was employed to provide a more realistic representation of the tumor microenvironment. The prepared NPs were found to reduce protein adsorption, and cell-membrane-coated NPs showed significantly higher cellular uptake. The secretion of proinflammatory cytokines in human monocytes after incubation with the prepared NPs was evaluated. Overall, the study demonstrates the importance of coatings in affecting the behavior and interaction of nanosystems with biological entities. The findings provide insight into bionano interactions and are important for the effective implementation of stealth surface engineering designs.
Collapse
Affiliation(s)
- Pablo Graván
- Department
of Applied Physics, Faculty of Science, University of Granada, 18071 Granada, Spain
- Department
of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- Instituto
de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Biopathology
and Regenerative Medicine Institute (IBIMER), Centre for Biomedical
Research (CIBM), University of Granada, 18016 Granada, Spain
- Excellence
Research Unit Modelling Nature (MNat), University
of Granada, 18016 Granada, Spain
- BioFab i3D—Biofabrication
and 3D (bio)printing laboratory, University
of Granada, 18100 Granada, Spain
| | - Jesús Peña-Martín
- Department
of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- Instituto
de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Biopathology
and Regenerative Medicine Institute (IBIMER), Centre for Biomedical
Research (CIBM), University of Granada, 18016 Granada, Spain
- Excellence
Research Unit Modelling Nature (MNat), University
of Granada, 18016 Granada, Spain
- BioFab i3D—Biofabrication
and 3D (bio)printing laboratory, University
of Granada, 18100 Granada, Spain
| | - Julia López de Andrés
- Department
of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- Instituto
de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Biopathology
and Regenerative Medicine Institute (IBIMER), Centre for Biomedical
Research (CIBM), University of Granada, 18016 Granada, Spain
- Excellence
Research Unit Modelling Nature (MNat), University
of Granada, 18016 Granada, Spain
- BioFab i3D—Biofabrication
and 3D (bio)printing laboratory, University
of Granada, 18100 Granada, Spain
| | - María Pedrosa
- Department
of Applied Physics, Faculty of Science, University of Granada, 18071 Granada, Spain
- Excellence
Research Unit Modelling Nature (MNat), University
of Granada, 18016 Granada, Spain
| | - Martín Villegas-Montoya
- Department
of Applied Physics, Faculty of Science, University of Granada, 18071 Granada, Spain
- Excellence
Research Unit Modelling Nature (MNat), University
of Granada, 18016 Granada, Spain
- Faculty
of Biology, Calzada de las Américas
and University, Ciudad Universitaria, 80040 Culiacán, Sinaloa, Mexico
| | | | - Juan A. Marchal
- Department
of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- Instituto
de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Biopathology
and Regenerative Medicine Institute (IBIMER), Centre for Biomedical
Research (CIBM), University of Granada, 18016 Granada, Spain
- Excellence
Research Unit Modelling Nature (MNat), University
of Granada, 18016 Granada, Spain
- BioFab i3D—Biofabrication
and 3D (bio)printing laboratory, University
of Granada, 18100 Granada, Spain
| | - Paola Sánchez-Moreno
- Department
of Applied Physics, Faculty of Science, University of Granada, 18071 Granada, Spain
- Excellence
Research Unit Modelling Nature (MNat), University
of Granada, 18016 Granada, Spain
| |
Collapse
|
46
|
Gao Y, Joshi M, Zhao Z, Mitragotri S. PEGylated therapeutics in the clinic. Bioeng Transl Med 2024; 9:e10600. [PMID: 38193121 PMCID: PMC10771556 DOI: 10.1002/btm2.10600] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/27/2023] [Accepted: 09/05/2023] [Indexed: 01/10/2024] Open
Abstract
The covalent attachment of polyethylene glycol (PEG) to therapeutic agents, termed PEGylation, is a well-established and clinically proven drug delivery approach to improve the pharmacokinetics and pharmacodynamics of drugs. Specifically, PEGylation can improve the parent drug's solubility, extend its circulation time, and reduce its immunogenicity, with minimal undesirable properties. PEGylation technology has been applied to various therapeutic modalities including small molecules, aptamers, peptides, and proteins, leading to over 30 PEGylated drugs currently used in the clinic and many investigational PEGylated agents under clinical trials. Here, we summarize the diverse types of PEGylation strategies, the key advantages of PEGylated therapeutics over their parent drugs, and the broad applications and impacts of PEGylation in clinical settings. A particular focus has been given to the size, topology, and functionalities of PEG molecules utilized in clinically used PEGylated drugs, as well as those under clinical trials. An additional section has been dedicated to analyzing some representative PEGylated drugs that were discontinued at different stages of clinical studies. Finally, we critically discuss the current challenges faced in the development and clinical translation of PEGylated agents.
Collapse
Affiliation(s)
- Yongsheng Gao
- John A. Paulson School of Engineering and Applied Sciences, Harvard UniversityAllstonMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMassachusettsUSA
- Present address:
Department of BioengineeringThe University of Texas at DallasRichardsonTXUSA
| | - Maithili Joshi
- John A. Paulson School of Engineering and Applied Sciences, Harvard UniversityAllstonMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMassachusettsUSA
| | - Zongmin Zhao
- Department of Pharmaceutical SciencesCollege of Pharmacy, University of Illinois at ChicagoChicagoIllinoisUSA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard UniversityAllstonMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMassachusettsUSA
| |
Collapse
|
47
|
Xi Z, Jiang Y, Ma Z, Li Q, Xi X, Fan C, Zhu S, Zhang J, Xu L. Using Mesoporous Silica-Based Dual Biomimetic Nano-Erythrocytes for an Improved Antitumor Effect. Pharmaceutics 2023; 15:2785. [PMID: 38140125 PMCID: PMC10747987 DOI: 10.3390/pharmaceutics15122785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/27/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
The nano-delivery system with a dual biomimetic effect can penetrate deeper in tumor microenvironments (TMEs) and release sufficient antitumor drugs, which has attracted much attention. In this study, we synthesized erythrocyte-like mesoporous silica nanoparticles (EMSNs) as the core loaded with doxorubicin (DOX) and coated them with calcium phosphate (CaP) and erythrocyte membrane (EM) to obtain DOX/EsPMs. The transmission electron microscopy (TEM), fluorescent co-localization and protein bands of SDS-PAGE were used to confirm the complete fabrication of EsPMs. The EsPMs with erythrocyte-like shape exhibited superior penetration ability in in vitro diffusion and tumor-sphere penetration experiments. Intracellular Ca2+ and ROS detection experiments showed that the CaP membranes of EsPMs with pH-sensitivity could provide Ca2+ continuously to induce reactive oxide species' (ROS) generation in the TME. The EM as a perfect "camouflaged clothing" which could confuse macrophagocytes into prolonging blood circulation. Hemolysis and non-specific protein adsorption tests proved the desirable biocompatibility of EsPMs. An in vivo pharmacodynamics evaluation showed that the DOX/EsPMs group had a satisfactory tumor-inhibition effect. These advantages of the nano-erythrocytes suggest that by modifying the existing materials to construct a nano-delivery system, nanoparticles will achieve a biomimetic effect from both their structure and function with a facilitated and sufficient drug release profile, which is of great significance for antitumor therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lu Xu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China; (Z.X.); (Y.J.); (Z.M.); (Q.L.); (X.X.); (C.F.); (S.Z.); (J.Z.)
| |
Collapse
|
48
|
Mohammad-Rafiei F, Khojini JY, Ghazvinian F, Alimardan S, Norioun H, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Cell membrane biomimetic nanoparticles in drug delivery. Biotechnol Appl Biochem 2023; 70:1843-1859. [PMID: 37387120 DOI: 10.1002/bab.2487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/05/2023] [Indexed: 07/01/2023]
Abstract
Despite the efficiency of nanoparticle (NP) therapy, in vivo investigations have shown that it does not perform as well as in vitro. In this case, NP confronts many defensive hurdles once they enter the body. The delivery of NP to sick tissue is inhibited by these immune-mediated clearance mechanisms. Hence, using a cell membrane to hide NP for active distribution offers up a new path for focused treatment. These NPs are better able to reach the disease's target location, leading to enhanced therapeutic efficacy. In this emerging class of drug delivery vehicles, the inherent relation between the NPs and the biological components obtained from the human body was utilized, which mimic the properties and activities of native cells. This new technology has shown the viability of using biomimicry to evade immune system-provided biological barriers, with an emphasis on restricting clearance from the body before reaching its intended target. Furthermore, by providing signaling cues and transplanted biological components that favorably change the intrinsic immune response at the disease site, the NPs would be capable interacting with immune cells regarding the biomimetic method. Thus, we aimed to provide a current landscape and future trends of biomimetic NPs in drug delivery.
Collapse
Affiliation(s)
- Fatemeh Mohammad-Rafiei
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Ghazvinian
- Department of Life science and biotechnology, Faculty of Natural Sciences, University of Shahid Beheshti, Tehran, Iran
| | - Sajad Alimardan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Norioun
- Medical Genetics Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Munich, Germany
| |
Collapse
|
49
|
Li M, Guo Q, Zhong C, Zhang Z. Multifunctional cell membranes-based nano-carriers for targeted therapies: a review of recent trends and future perspective. Drug Deliv 2023; 30:2288797. [PMID: 38069500 PMCID: PMC10987056 DOI: 10.1080/10717544.2023.2288797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/05/2023] [Indexed: 12/18/2023] Open
Abstract
Nanotechnology has ignited a transformative revolution in disease detection, prevention, management, and treatment. Central to this paradigm shift is the innovative realm of cell membrane-based nanocarriers, a burgeoning class of biomimetic nanoparticles (NPs) that redefine the boundaries of biomedical applications. These remarkable nanocarriers, designed through a top-down approach, harness the intrinsic properties of cell-derived materials as their fundamental building blocks. Through shrouding themselves in natural cell membranes, these nanocarriers extend their circulation longevity and empower themselves to intricately navigate and modulate the multifaceted microenvironments associated with various diseases. This comprehensive review provides a panoramic view of recent breakthroughs in biomimetic nanomaterials, emphasizing their diverse applications in cancer treatment, cardiovascular therapy, viral infections, COVID-19 management, and autoimmune diseases. In this exposition, we deliver a concise yet illuminating overview of the distinctive properties underpinning biomimetic nanomaterials, elucidating their pivotal role in biomedical innovation. We subsequently delve into the exceptional advantages these nanomaterials offer, shedding light on the unique attributes that position them at the forefront of cutting-edge research. Moreover, we briefly explore the intricate synthesis processes employed in creating these biomimetic nanocarriers, shedding light on the methodologies that drive their development.
Collapse
Affiliation(s)
- Mo Li
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, China
| | - Qiushi Guo
- Pharmacy Department, First Hospital of Jilin University—the Eastern Division, Changchun, China
| | - Chongli Zhong
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, China
| | - Ziyan Zhang
- Department of Orthopedics, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
50
|
Wang H, Wang Y, Yuan C, Xu X, Zhou W, Huang Y, Lu H, Zheng Y, Luo G, Shang J, Sui M. Polyethylene glycol (PEG)-associated immune responses triggered by clinically relevant lipid nanoparticles in rats. NPJ Vaccines 2023; 8:169. [PMID: 37919316 PMCID: PMC10622525 DOI: 10.1038/s41541-023-00766-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 10/13/2023] [Indexed: 11/04/2023] Open
Abstract
With the large-scale vaccination of lipid nanoparticles (LNP)-based COVID-19 mRNA vaccines, elucidating the potential polyethylene glycol (PEG)-associated immune responses triggered by clinically relevant LNP has become imminent. However, inconsistent findings were observed across very limited population-based studies. Herein we initiated a study using LNP carrier of Comirnaty® as a representative, and simulated real-world clinical practice covering a series of time points and various doses correlated with approved LNP-delivered drugs in a rat model. We demonstrated the time- and dose-dependency of LNP-induced anti-PEG antibodies in rats. As a thymus-independent antigen, LNP unexpectedly induced isotype switch and immune memory, leading to rapid enhancement and longer lasting time of anti-PEG IgM and IgG upon re-injection in rats. Importantly, initial LNP injection accelerated the blood clearance of subsequent dosing in rats. These findings refine our understandings on LNP and possibly other PEG derivatives, and may promote optimization of related premarket guidelines and clinical protocols.
Collapse
Affiliation(s)
- Haiyang Wang
- School of Basic Medical Sciences and Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yisha Wang
- School of Basic Medical Sciences and Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Changzheng Yuan
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Xu
- Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Wenbin Zhou
- School of Basic Medical Sciences and Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yuhui Huang
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Huan Lu
- School of Basic Medical Sciences and Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yue Zheng
- School of Basic Medical Sciences and Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Gan Luo
- School of Basic Medical Sciences and Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jia Shang
- Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Meihua Sui
- School of Basic Medical Sciences and Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|