1
|
Daoud L, Hoang S, Milin-Moguerou A, Eyer J, Breton T, Montembault V, Fontaine L, Passirani C, Saulnier P, Krupka O. One-Step Synthesis of Poly(2-alkyl-2-oxazoline)-Coated Gold Nanospheres: A Greener Approach for Biomedical Uses. Biomacromolecules 2025; 26:1923-1934. [PMID: 40008451 DOI: 10.1021/acs.biomac.4c01738] [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: 02/27/2025]
Abstract
The biological application of gold nanospheres (AuNSs) is often constrained by their stability and cytotoxicity. We present a greener synthetic approach that gives a simple and more environmentally friendly route to synthesizing AuNSs for biomedical applications. In this study, we detail a novel one-step, green synthesis of poly(2-alkyl-2-oxazoline) (POx)-coated AuNSs, which eliminates the need for additional reducing and stabilizing agents. The impact of the polymer structure on the nanoparticle formulation kinetics and nanoparticle characteristics is thoroughly investigated, revealing that the terminal functional group and the alkyl side chain significantly influence the reduction and stabilization process of AuNSs. Finally, POx-coated AuNSs were tested in vitro against F98 glioblastoma cells and proved to be usable without significant toxicity up to 75 μM. Herein, the outlined rapid and efficient method of the preparation of POx-coated AuNSs serves as a foundation for advancing the development of complex AuNSs tailored for biomedical applications.
Collapse
Affiliation(s)
- Léa Daoud
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Stéphane Hoang
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | | | - Joël Eyer
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Tony Breton
- Univ Angers, CNRS, MOLTECH-Anjou, SFR MATRIX, F-49000 Angers, France
| | - Véronique Montembault
- Institut des Molécules et Matériaux du Mans, UMR 6283 CNRS-Le Mans Université, Le Mans 72085, France
| | - Laurent Fontaine
- Institut des Molécules et Matériaux du Mans, UMR 6283 CNRS-Le Mans Université, Le Mans 72085, France
| | | | - Patrick Saulnier
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Oksana Krupka
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| |
Collapse
|
2
|
Vera-López KJ, Aranzamendi-Zenteno M, Davila-Del-Carpio G, Nieto-Montesinos R. Using Immunoliposomes as Carriers to Enhance the Therapeutic Effectiveness of Macamide N-3-Methoxybenzyl-Linoleamide. Neurol Int 2025; 17:38. [PMID: 40137459 PMCID: PMC11945115 DOI: 10.3390/neurolint17030038] [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: 01/28/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Epilepsy is one of the most common chronic neurological disorders, characterized by alterations in neuronal electrical activity that result in recurrent seizures and involuntary body movements. Anticonvulsants are the primary treatment for this condition, helping patients improve their quality of life. However, the development of new drugs with fewer side effects and greater economic accessibility remains a key focus in nanomedicine. Macamides, secondary metabolites derived from Maca (Lepidium meyenii), represent a promising class of novel drugs with diverse therapeutic applications, particularly in the treatment of neurological disorders. METHODS In this study, we optimized the potential of the macamide N-3-methoxybenzyl-linoleamide (3-MBL) as an anticonvulsant agent through its encapsulation in PEGylated liposomes conjugated with OX26 F(ab')2 fragments. RESULTS These immunoliposomes exhibited a size of 120.52 ± 9.46 nm and a zeta potential of -8.57 ± 0.80 mV. Furthermore, in vivo tests using a pilocarpine-induced status epilepticus model revealed that the immunoliposomes provided greater efficacy against epileptic seizures compared to the free form of N-3-methoxybenzyl-linoleamide at the same dose. Notably, the observed anticonvulsant effect was comparable to that of carbamazepine, a traditional FDA-approved antiepileptic drug. CONCLUSIONS This pioneering work employs liposomal nanocarriers to deliver macamides to the brain, aiming to set a new standard for the use of modified liposomes in anticonvulsant epilepsy treatment.
Collapse
Affiliation(s)
| | | | | | - Rita Nieto-Montesinos
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04000, Peru; (K.J.V.-L.); (M.A.-Z.); (G.D.-D.-C.)
| |
Collapse
|
3
|
Roointan A, Xu R, Corrie S, Hagemeyer CE, Alt K. Nanotherapeutics in Kidney Disease: Innovations, Challenges, and Future Directions. J Am Soc Nephrol 2025; 36:500-518. [PMID: 39705082 PMCID: PMC11888965 DOI: 10.1681/asn.0000000608] [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: 08/28/2024] [Accepted: 12/17/2024] [Indexed: 12/22/2024] Open
Abstract
The treatment and management of kidney diseases present a significant global challenge, affecting over 800 million individuals and necessitating innovative therapeutic strategies that transcend symptomatic relief. The application of nanotechnology to therapies for kidney diseases, while still in its early stages, holds transformative potential for improving treatment outcomes. Recent advancements in nanoparticle-based drug delivery leverage the unique physicochemical properties of nanoparticles for targeted and controlled therapeutic delivery to the kidneys. Current research is focused on understanding the functional and phenotypic changes in kidney cells during both acute and chronic conditions, allowing for the identification of optimal target cells. In addition, the development of tailored nanomedicines enhances their retention and binding to key renal membranes and cell populations, ultimately improving localization, tolerability, and efficacy. However, significant barriers remain, including inconsistent nanoparticle synthesis and the complexity of kidney-specific targeting. To overcome these challenges, the field requires advanced synthesis techniques, refined targeting strategies, and the establishment of animal models that accurately reflect human kidney diseases. These efforts are critical for the clinical application of nanotherapeutics, which promise novel solutions for kidney disease management. This review evaluates a substantial body of in vivo research, highlighting the prospects, challenges, and opportunities presented by nanotechnology-mediated therapies and their potential to transform kidney disease treatment.
Collapse
Affiliation(s)
- Amir Roointan
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- NanoTheranostics Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Rong Xu
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Simon Corrie
- Department of Chemical and Biological Engineering, Monash University, Melbourne, Victoria, Australia
| | - Christoph E. Hagemeyer
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Karen Alt
- NanoTheranostics Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Prossnitz AN, Nguyen LT, Eckman N, Borkar S, Tetef S, Autzen AAA, Fuller GG, Appel EA. Defining Structure-Function Relationships of Amphiphilic Excipients Enables Rational Design of Ultra-Stable Biopharmaceuticals. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409604. [PMID: 39764759 PMCID: PMC11848622 DOI: 10.1002/advs.202409604] [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: 08/13/2024] [Revised: 11/16/2024] [Indexed: 02/25/2025]
Abstract
Biopharmaceuticals are the fastest-growing class of drugs in the healthcare industry, but their global reach is severely limited by their propensity for rapid aggregation. Currently, surfactant excipients such as polysorbates and poloxamers are used to prevent protein aggregation, which significantly extends shelf-life. Unfortunately, these excipients are themselves unstable, oxidizing rapidly into 100s of distinct compounds, some of which cause severe adverse events in patients. Here, the highly stable, well-defined, and modular nature of amphiphilic polyacrylamide-derived excipients is leveraged to isolate the key mechanisms responsible for excipient-mediated protein stabilization. With a library of compositionally identical but structurally distinct amphiphilic excipients, a new property is quantified, compositional dispersity, that is key to excipient performance and utilized this property to rationally design new ultra-stable surfactant excipients that increase the stability of a notoriously unstable biopharmaceutical, monomeric insulin, by an order of magnitude. This comprehensive and generalizable understanding of excipient structure-function relationships represents a paradigm shift for the formulation of biopharmaceuticals, moving away from trial-and-error screening approaches toward rational design.
Collapse
Affiliation(s)
| | | | - Noah Eckman
- Department of Chemical EngineeringStanford UniversityStanfordCA94305USA
| | - Suraj Borkar
- Department of Chemical EngineeringStanford UniversityStanfordCA94305USA
| | - Samantha Tetef
- Department of PhysicsUniversity of WashingtonSeattleWA98195USA
| | - Anton A. A. Autzen
- Department of Health Technology Cell and Drug TechnologiesTechnical University of DenmarkLyngby2800Denmark
| | - Gerald G. Fuller
- Department of Chemical EngineeringStanford UniversityStanfordCA94305USA
| | - Eric A. Appel
- Department of Materials Science & EngineeringStanford UniversityStanfordCA94305USA
- Department of Pediatrics – EndocrinologyStanford University School of MedicineStanfordCA94305USA
- Department of BioengineeringStanford UniversityStanfordCA94305USA
- ChEM‐H InstituteStanford UniversityStanfordCA94305USA
- Woods Institute for the EnvironmentStanford UniversityStanfordCA94305USA
| |
Collapse
|
5
|
Hagman C, Chasseigne G, Nelson R, Anlauff F, Kagan M, Goldfine AB, Terszowski G, Jadhav M. Immunogenicity assessment strategy for a chemically modified therapeutic protein in clinical development. Front Immunol 2024; 15:1438251. [PMID: 39588370 PMCID: PMC11586355 DOI: 10.3389/fimmu.2024.1438251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/16/2024] [Indexed: 11/27/2024] Open
Abstract
The clinical immunogenicity assessment for complex multidomain biological drugs is challenging due to multiple factors that must be taken into consideration. Here, we describe a strategy to overcome multiple bioanalytical challenges in order to assess anti-drug antibodies (ADA) for a novel and unique chemically modified protein therapeutic. A risk-centered approach was adopted to evaluate the immunogenic response to a modified version of human growth differentiation factor 15 (GDF15) connected to an albumin-binding fatty acid via a polyethylene glycol (PEG) linker. Key steps include monitoring anti-drug antibodies (ADAs), using a standard tiered approach of screening and confirmation. To deepen our understanding of ADA response, as a third tier of immunogenicity assessment, novel extensive characterization using a set of assays was developed, validated, and used routinely in clinical sample analysis. This characterization step included performance of titration, mapping of ADA response including anti-GDF15 and anti-PEG-fatty-acid antibody characterization, and assessment of the neutralizing anti-drug antibodies (NAbs) using cell-based assays for immunogenicity in parallel. The analytical methods were applied during two clinical trials involving both healthy volunteers and overweight or obese patients. We observed low incident rates for ADA and no ADAs against the PEG linker with fatty acid conjugation. In one of the clinical studies, we identified neutralizing ADAs. The proposed novel strategy of extensive characterization proved effective for monitoring the presence of ADAs and NAbs and can be used to support clinical development of a broad range of chemically modified proteins and multidomain biotherapeutics.
Collapse
Affiliation(s)
- Charlotte Hagman
- Pharmacokinetic Sciences - Drug Disposition, Biomedical Research, Novartis, Basel, Switzerland
| | - Gaetan Chasseigne
- Pharmacokinetic Sciences - Drug Disposition, Biomedical Research, Novartis, Basel, Switzerland
| | | | | | - Mark Kagan
- Pharmacokinetic Sciences, Biomedical Research, Novartis, East Hanover, NJ, United States
| | - Allison B. Goldfine
- Translational Medicine, Biomedical Research, Novartis, Cambridge, MA, United States
| | - Grzegorz Terszowski
- Pharmacokinetic Sciences - Drug Disposition, Biomedical Research, Novartis, Basel, Switzerland
| | - Maria Jadhav
- Pharmacokinetic Sciences - Drug Disposition, Biomedical Research, Novartis, Cambridge, MA, United States
| |
Collapse
|
6
|
Gonçalves J, Caliceti P. Optimizing Pharmacological and Immunological Properties of Therapeutic Proteins Through PEGylation: Investigating Key Parameters and Their Impact. Drug Des Devel Ther 2024; 18:5041-5062. [PMID: 39529843 PMCID: PMC11552514 DOI: 10.2147/dddt.s481420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/06/2024] [Indexed: 11/16/2024] Open
Abstract
Protein PEGylation represents a significant technological advancement in the development of protein-based therapeutics and is widely used to reduce immunogenicity, enhance pharmacokinetics, and/or improve stability. The improved pharmacokinetic profile of PEGylated proteins compared with the native protein results in sustained versus fluctuating plasma concentrations and carries the potential of less frequent administration. However, attachment of PEG to therapeutic proteins can alter their structural conformation, which exposes new epitopes to the immune system. The design of PEGylated proteins thus needs to balance the intended benefits with the potential risks associated with the immunogenicity of the PEG moiety itself or resulting from alterations in the conformation of the therapeutic protein. In recent years, advancements in protein PEGylation chemistry have offered the capability to target PEG attachment to specific amino acids to create more stable and bioactive therapies. The biophysical and biopharmaceutical features of PEGylated proteins can vary based on polymer size, shape, density, and conjugation site, and the immunogenicity of the conjugate can be further impacted by the properties of the therapeutic protein itself and the characteristics of the patient. It is important to note that not all patients will develop an immune response toward the PEG moiety, and not all immune responses are clinically meaningful. A comprehensive understanding of the factors that influence immunogenic responses to PEGylated proteins is important to optimize their therapeutic benefits. This article reviews the design and optimization of PEGylation strategies to enhance the clinical performance of protein-based therapeutics while minimizing immunogenic responses to the PEG moiety or PEGylated proteins.
Collapse
Affiliation(s)
- João Gonçalves
- Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
7
|
Ismail EA, Omolo CA, Gafar MA, Khan R, Nyandoro VO, Yakubu ES, Mackraj I, Tageldin A, Govender T. Novel peptide and hyaluronic acid coated biomimetic liposomes for targeting bacterial infections and sepsis. Int J Pharm 2024; 662:124493. [PMID: 39048042 DOI: 10.1016/j.ijpharm.2024.124493] [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: 12/07/2023] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Sepsis is a life-threatening syndrome resulting from an imbalanced immune response to severe infections. Despite advances in nanomedicines, effective treatments for sepsis are still lacking. Herein, vancomycin free base (VCM)-loaded dual functionalized biomimetic liposomes based on a novel TLR4-targeting peptide (P3) and hyaluronic acid (HA) (HA-P3-Lipo) were developed to enhance sepsis therapy. The nanocarrier revealed appropriate physicochemical parameters, good stability, and biocompatibility. The release of VCM from HA-P3-Lipo was found to be sustained with 76 % VCM released in 48 h. The biomimicry was elucidated by in silico tools and MST and results confirmed strong binding between the system and TLR4. Furthermore, HA-P3-Lipo revealed 2-fold enhanced antibacterial activity against S. aureus, sustained antibacterial activity against MRSA over 72 h and 5-fold better MRSA biofilm inhibition compared to bare VCM. Bacterial-killing kinetics and flow cytometry confirmed the superiority of HA-P3-Lipo in eliminating MRSA faster than VCM. The in vivo potential of the nanocarrier was elucidated in an MRSA-induced sepsis mice model, and the results confirmed the superiority of HA-P3-Lipo compared to free VCM in eliminating bacteria and down-regulating the proinflammatory markers. Therefore, HA-P3-Lipo exhibits potential as a promising novel multi-functional nanosystem against sepsis and could significantly contribute to the transformation of sepsis therapy.
Collapse
Affiliation(s)
- Eman A Ismail
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa; Department of Pharmaceutics, Faculty of Pharmacy, University of Gezira, Wad Medani, Sudan
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa; United States International University-Africa, School of Pharmacy and Health Sciences, Department of Pharmaceutics, P. O. Box 14634-00800, Nairobi, Kenya.
| | - Mohammed A Gafar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Rene Khan
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Vincent O Nyandoro
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa; School of Pharmacy, Kabarak University, P.O BOX Private bag 20157, Nakuru, Kenya
| | - Elliasu S Yakubu
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Cape Town 7505, South Africa
| | - Irene Mackraj
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Abdelrahman Tageldin
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
| |
Collapse
|
8
|
Saber N, Senti ME, Schiffelers RM. Lipid Nanoparticles for Nucleic Acid Delivery Beyond the Liver. Hum Gene Ther 2024; 35:617-627. [PMID: 39139067 DOI: 10.1089/hum.2024.106] [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: 08/15/2024] Open
Abstract
Lipid nanoparticles (LNPs) are the most clinically advanced drug delivery system for nucleic acid therapeutics, exemplified by the success of the COVID-19 mRNA vaccines. However, their clinical use is currently limited to hepatic diseases and vaccines due to their tendency to accumulate in the liver upon intravenous administration. To fully leverage their potential, it is essential to understand and address their liver tropism, while also developing strategies to enhance delivery to tissues beyond the liver. Ensuring that these therapeutics reach their target cells while avoiding off-target cells is essential for both their efficacy and safety. There are three potential targeting strategies-passive, active, and endogenous-which can be used individually or in combination to target nonhepatic tissues. In this review, we delve into the recent advancements in LNP engineering for delivering nucleic acid beyond the liver.
Collapse
Affiliation(s)
- Nadine Saber
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | |
Collapse
|
9
|
Lisiecka MZ. Polyethylene glycol and immunology: aspects of allergic reactions and their mechanisms, as well as ways to prevent them in clinical practice. Immunol Res 2024; 72:675-682. [PMID: 38502278 DOI: 10.1007/s12026-024-09473-w] [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: 11/01/2023] [Accepted: 03/14/2024] [Indexed: 03/21/2024]
Abstract
In modern medical practice, where polyethylene glycol is widely used as a component of various drugs, such as vaccines, chemotherapy drugs, and antibiotics, including vaccines, the issue of allergic reactions to this substance is becoming increasingly important. The purpose of this study is to review and systematise data on various aspects of allergic reactions to polyethylene glycol with the aim of better understanding their pathogenesis, clinical manifestations, diagnostic methods, and possible treatment approaches. The study analysed literature data in modern databases, such as MEDLINE, PubMed, and Scopus, on allergic reactions to polyethylene glycol, using the keywords: "PEG", "polyethylene glycol", "allergy", "side effect". The main aspects of allergy to this substance were highlighted, including mechanisms of development, diagnostic methods, and possible treatment strategies. The analysis found that allergic reactions to polyethylene glycol can manifest in a variety of ways, including anaphylaxis and systemic reactions. A possible role for the immune response has been identified, including the production of IgE and IgM antibodies, complement activation, and accelerated clearance in response to polyethylene glycol, in blood plasma. Data are also provided on how to diagnose an increased risk of an allergic reaction in patients who have previously received drugs with this type of drug transporter and in patients receiving high molecular weight types of polyethylene glycol. The results of this review contribute to a better understanding of allergic reactions to polyethylene glycol and provide information for the development of more effective diagnostic and treatment methods.
Collapse
Affiliation(s)
- Maria Zofia Lisiecka
- Department of Allergology, National Medical Institute of the Ministry of the Interior and Administration, 137 Woloska Str, 02-507, Warsaw, Poland.
| |
Collapse
|
10
|
Bento C, Katz M, Santos MMM, Afonso CAM. Striving for Uniformity: A Review on Advances and Challenges To Achieve Uniform Polyethylene Glycol. Org Process Res Dev 2024; 28:860-890. [PMID: 38660381 PMCID: PMC11036406 DOI: 10.1021/acs.oprd.3c00428] [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: 11/05/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 04/26/2024]
Abstract
Poly(ethylene glycol) (PEG) is the polymer of choice in drug delivery systems due to its biocompatibility and hydrophilicity. For over 20 years, this polymer has been widely used in the drug delivery of small drugs, proteins, oligonucleotides, and liposomes, improving the stability and pharmacokinetics of many drugs. However, despite the extensive clinical experience with PEG, concerns have emerged related to its use. These include hypersensitivity, purity, and nonbiodegradability. Moreover, conventional PEG is a mixture of polymers that can complicate drug synthesis and purification leading to unwanted immunogenic reactions. Studies have shown that uniform PEGylated drugs may be more effective than conventional PEGylated drugs as they can overcome issues related to molecular heterogeneity and immunogenicity. This has led to significant research efforts to develop synthetic procedures to produce uniform PEGs (monodisperse PEGs). As a result, iterative step-by-step controlled synthesis methods have been created over time and have shown promising results. Nonetheless, these procedures have presented numerous challenges due to their iterative nature and the requirement for multiple purification steps, resulting in increased costs and time consumption. Despite these challenges, the synthetic procedures went through several improvements. This review summarizes and discusses recent advances in the synthesis of uniform PEGs and its derivatives with a focus on overall yields, scalability, and purity of the polymers. Additionally, the available characterization methods for assessing polymer monodispersity are discussed as well as uniform PEG applications, side effects, and possible alternative polymers that can overcome the drawbacks.
Collapse
Affiliation(s)
- Cláudia Bento
- Hovione
Farmaciência S.A., Estrada do Paço do Lumiar, Campus do Lumiar, Edifício
R, 1649-038 Lisboa, Portugal
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Marianna Katz
- Hovione
Farmaciência S.A., Estrada do Paço do Lumiar, Campus do Lumiar, Edifício
R, 1649-038 Lisboa, Portugal
| | - Maria M. M. Santos
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Carlos A. M. Afonso
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
11
|
Gaballa SA, Shimizu T, Takata H, Ando H, Ibrahim M, Emam SE, Amorim Matsuo NC, Kim Y, Naguib YW, Mady FM, Khaled KA, Ishida T. Impact of Anti-PEG IgM Induced via the Topical Application of a Cosmetic Product Containing PEG Derivatives on the Antitumor Effects of PEGylated Liposomal Antitumor Drug Formulations in Mice. Mol Pharm 2024; 21:622-632. [PMID: 38273445 DOI: 10.1021/acs.molpharmaceut.3c00774] [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: 01/27/2024]
Abstract
Poly(ethylene glycol) (PEG) is used in many common products, such as cosmetics. PEG, however, is also used to covalently conjugate drug molecules, proteins, or nanocarriers, which is termed PEGylation, to serve as a shield against the natural immune system of the human body. Repeated administration of some PEGylated products, however, is known to induce anti-PEG antibodies. In addition, preexisting anti-PEG antibodies are now being detected in healthy individuals who have never received PEGylated therapeutics. Both treatment-induced and preexisting anti-PEG antibodies alter the pharmacokinetic properties, which can result in a subsequent reduction in the therapeutic efficacy of administered PEGylated therapeutics through the so-called accelerated blood clearance (ABC) phenomenon. Moreover, these anti-PEG antibodies are widely reported to be related to severe hypersensitivity reactions following the administration of PEGylated therapeutics, including COVID-19 vaccines. We recently reported that the topical application of a cosmetic product containing PEG derivatives induced anti-PEG immunoglobulin M (IgM) in a mouse model. Our finding indicates that the PEG derivatives in cosmetic products could be a major cause of the preexistence of anti-PEG antibodies in healthy individuals. In this study, therefore, the pharmacokinetics and therapeutic effects of Doxil (doxorubicin hydrochloride-loaded PEGylated liposomes) and oxaliplatin-loaded PEGylated liposomes (Liposomal l-OHP) were studied in mice. The anti-PEG IgM antibodies induced by the topical application of cosmetic products obviously accelerated the blood clearance of both PEGylated liposomal formulations. Moreover, in C26 tumor-bearing mice, the tumor growth suppressive effects of both Doxil and Liposomal l-OHP were significantly attenuated in the presence of anti-PEG IgM antibodies induced by the topical application of cosmetic products. These results confirm that the topical application of a cosmetic product containing PEG derivatives could produce preexisting anti-PEG antibodies that then affect the therapeutic efficacy of subsequent doses of PEGylated therapeutics.
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
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
- Institute of Innovative Drug Delivery System, Graduate School 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
- Institute of Innovative Drug Delivery System, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Mohamed Ibrahim
- 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
| | - Sherif E Emam
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Nana Cristina Amorim Matsuo
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Yuri Kim
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - 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
- Institute of Innovative Drug Delivery System, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| |
Collapse
|
12
|
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: 3] [Impact Index Per Article: 3.0] [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
|
13
|
Deuker MFS, Mailänder V, Morsbach S, Landfester K. Anti-PEG antibodies enriched in the protein corona of PEGylated nanocarriers impact the cell uptake. NANOSCALE HORIZONS 2023; 8:1377-1385. [PMID: 37591816 DOI: 10.1039/d3nh00198a] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Poly(ethylene glycol) (PEG) is the gold standard used to reduce unspecific protein adsorption and prolong nanocarrier circulation time. However, this stealth effect could be counteracted by the increasing prevalence of anti-PEG antibodies in the bloodstream. Up to now, the presence of anti-PEG antibodies in the protein corona and their effect on cell uptake has not been investigated yet. Our results showed a high concentration and prevalence of anti-PEG antibodies in the German population. PEGylated nanocarriers exhibited a higher level of anti-PEG antibodies in the protein corona compared to non-PEGylated, which lead to higher uptake in macrophages. Consequently, the anti-PEG antibodies in the protein corona could mitigate the stealth effect of PEG, leading to accelerated blood clearance and unwanted side effects.
Collapse
Affiliation(s)
- Mareike F S Deuker
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Volker Mailänder
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Svenja Morsbach
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| |
Collapse
|
14
|
Guo C, Yuan H, Wang Y, Feng Y, Zhang Y, Yin T, He H, Gou J, Tang X. The interplay between PEGylated nanoparticles and blood immune system. Adv Drug Deliv Rev 2023; 200:115044. [PMID: 37541623 DOI: 10.1016/j.addr.2023.115044] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/11/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
During the last two decades, an increasing number of reports have pointed out that the immunogenicity of polyethylene glycol (PEG) may trigger accelerated blood clearance (ABC) and hypersensitivity reaction (HSR) to PEGylated nanoparticles, which could make PEG modification counterproductive. These phenomena would be detrimental to the efficacy of the load and even life-threatening to patients. Consequently, further elucidation of the interplay between PEGylated nanoparticles and the blood immune system will be beneficial to developing and applying related formulations. Many groups have worked to unveil the relevance of structural factors, dosing schedule, and other factors to the ABC phenomenon and hypersensitivity reaction. Interestingly, the results of some reports seem to be difficult to interpret or contradict with other reports. In this review, we summarize the physiological mechanisms of PEG-specific immune response. Moreover, we speculate on the potential relationship between the induction phase and the effectuation phase to explain the divergent results in published reports. In addition, the role of nanoparticle-associated factors is discussed based on the classification of the action phase. This review may help researchers to develop PEGylated nanoparticles to avoid unfavorable immune responses based on the underlying mechanism.
Collapse
Affiliation(s)
- Chen Guo
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Haoyang Yuan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Yuxiu Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Yupeng Feng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| |
Collapse
|
15
|
Zhang S, Sun Y, Zhang L, Zhang F, Gao W. Thermoresponsive Polypeptide Fused L-Asparaginase with Mitigated Immunogenicity and Enhanced Efficacy in Treating Hematologic Malignancies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300469. [PMID: 37271878 PMCID: PMC10427413 DOI: 10.1002/advs.202300469] [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: 01/20/2023] [Revised: 05/17/2023] [Indexed: 06/06/2023]
Abstract
L-Asparaginase (ASP) is well-known for its excellent efficacy in treating hematological malignancies. Unfortunately, the intrinsic shortcomings of ASP, namely high immunogenicity, severe toxicity, short half-life, and poor stability, restrict its clinical usage. Poly(ethylene glycol) conjugation (PEGylation) of ASP is an effective strategy to address these issues, but it is not ideal in clinical applications due to complex chemical synthesis procedures, reduced ASP activity after conjugation, and pre-existing anti-PEG antibodies in humans. Herein, the authors genetically engineered an elastin-like polypeptide (ELP)-fused ASP (ASP-ELP), a core-shell structured tetramer predicted by AlphaFold2, to overcome the limitations of ASP and PEG-ASP. Notably, the unique thermosensitivity of ASP-ELP enables the in situ formation of a sustained-release depot post-injection with zero-order release kinetics over a long time. The in vitro and in vivo studies reveal that ASP-ELP possesses increased activity retention, improved stability, extended half-life, mitigated immunogenicity, reduced toxicity, and enhanced efficacy compared to ASP and PEG-ASP. Indeed, ASP-ELP treatment in leukemia or lymphoma mouse models of cell line-derived xenograft (CDX) shows potent anti-cancer effects with significantly prolonged survival. The findings also indicate that artificial intelligence (AI)-assisted genetic engineering is instructive in designing protein-polypeptide conjugates and may pave the way to develop next-generation biologics to enhance cancer treatment.
Collapse
Affiliation(s)
- Sanke Zhang
- Institute of Medical TechnologyPeking University Health Science CenterPeking University School and Hospital of StomatologyBiomedical Engineering DepartmentPeking UniversityPeking University International Cancer InstitutePeking University‐Yunnan Baiyao International Medical Research CenterBeijing100191China
| | - Yuanzi Sun
- Institute of Medical TechnologyPeking University Health Science CenterPeking University School and Hospital of StomatologyBiomedical Engineering DepartmentPeking UniversityPeking University International Cancer InstitutePeking University‐Yunnan Baiyao International Medical Research CenterBeijing100191China
| | - Longshuai Zhang
- Institute of Medical TechnologyPeking University Health Science CenterPeking University School and Hospital of StomatologyBiomedical Engineering DepartmentPeking UniversityPeking University International Cancer InstitutePeking University‐Yunnan Baiyao International Medical Research CenterBeijing100191China
| | - Fan Zhang
- Institute of Medical TechnologyPeking University Health Science CenterPeking University School and Hospital of StomatologyBiomedical Engineering DepartmentPeking UniversityPeking University International Cancer InstitutePeking University‐Yunnan Baiyao International Medical Research CenterBeijing100191China
| | - Weiping Gao
- Institute of Medical TechnologyPeking University Health Science CenterPeking University School and Hospital of StomatologyBiomedical Engineering DepartmentPeking UniversityPeking University International Cancer InstitutePeking University‐Yunnan Baiyao International Medical Research CenterBeijing100191China
| |
Collapse
|
16
|
Okada N, Taro S, Ando H, Nakamura S, Goda M, Abe M, Kitahara T, Ishida T, Ishizawa K. Clinical Impact of Antipolyethylene Glycol (PEG) Antibody in Hematological Patients Administered PEGylated-Granulocyte Colony-Stimulating Factor. Clin Pharmacol Drug Dev 2023; 12:826-831. [PMID: 36708147 DOI: 10.1002/cpdd.1225] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/02/2023] [Indexed: 01/29/2023]
Abstract
Polyethylene glycol (PEG) is a polymer covalently attached to proteins to improve their half-life and efficacy. We previously reported that the PEGylated granulocyte colony-stimulating factor (PEG-G-CSF) is immunogenic, which could adversely impact drug efficacy and safety in animal models. Here, we analyzed the relationship between anti-PEG antibody titers and the clinical impact of PEG-G-CSF in 19 hematological patients. A gradual decrease of anti-PEG antibody titers from baseline was observed after PEG-G-CSF administration. Of the 19 participants, 10 were assessed for noninfectious fever after the first administration of PEG-G-CSF and three experienced this reaction. The receiver operating characteristic curve revealed that the cut-off values of pretreated anti-PEG IgM and IgG titers for noninfectious fever were set at 5.0 and 96.6 U/mL, respectively. All patients who experienced noninfectious fever had anti-PEG antibody titers above this cut-off value (P = .033). An enzyme-linked immunosorbent assay revealed that some anti-PEG antibodies in patients with anti-PEG antibody titers above the cut-off value reacted with the PEGylated liposome. These results indicate the reactivity of the anti-PEG antibodies to PEGylated therapeutics observed in hematologic patients and the possibility of the relationship between high titers of anti-PEG antibodies and the development of adverse events after PEG-G-CSF administration.
Collapse
Affiliation(s)
- Naoto Okada
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
- Pharmacy Department, Yamaguchi University Hospital, Yamaguchi, Japan
| | - Shimizu Taro
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Shingen Nakamura
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Mitsuhiro Goda
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masahiro Abe
- Department of Haematology, Endocrinology, and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takashi Kitahara
- Pharmacy Department, Yamaguchi University Hospital, Yamaguchi, Japan
- Clinical Pharmacology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Keisuke Ishizawa
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
- Clinical Research Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| |
Collapse
|
17
|
Sui D, Wang Y, Sun W, Wei L, Li C, Gui Y, Qi Z, Liu X, Song Y, Deng Y. Cleavable-Branched Polymer-Modified Liposomes Reduce Accelerated Blood Clearance and Enhance Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37384837 DOI: 10.1021/acsami.3c02762] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
In recent years, cationic liposomes have been successfully used as delivery platforms for mRNA vaccines. Poly(ethylene glycol) (PEG)-lipid derivatives are widely used to enhance the stability and reduce the toxicity of cationic liposomes. However, these derivatives are often immunogenic, triggering the rise of anti-PEG antibodies. Understanding the role and impact of PEG-lipid derivatives on PEGylated cationic liposomes is key to solving the PEG dilemma. In this study, we designed linear, branched, and cleavable-branched cationic liposomes modified with PEG-lipid derivatives and investigated the effect of the liposome-induced accelerated blood clearance (ABC) phenomenon on photothermal therapy. Our study indicated that the linear PEG-lipid derivatives mediated the effect of photothermal therapy by stimulating splenic marginal zone (MZ) B cells to secrete anti-PEG antibodies and increasing the level of IgM expression in the follicular region of the spleen. However, the cleavable-branched and branched PEG-lipid derivatives did not activate the complement system and avoided the ABC phenomenon by inducing noticeably lower levels of anti-PEG antibodies. The cleavable-branched PEGylated cationic liposomes improved the effect of photothermal therapy by reversing the charge on the liposome surface. This detailed study of PEG-lipid derivatives contributes to the further development and clinical application of PEGylated cationic liposomes.
Collapse
Affiliation(s)
- Dezhi Sui
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China
| | - Yujie Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China
| | - Wenliang Sun
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China
| | - Lu Wei
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China
| | - Changzhi Li
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China
| | - Yangxu Gui
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China
| | - Zhaowei Qi
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China
| |
Collapse
|
18
|
Münter R, Christensen E, Andresen TL, Larsen JB. Studying how administration route and dose regulates antibody generation against LNPs for mRNA delivery with single-particle resolution. Mol Ther Methods Clin Dev 2023; 29:450-459. [PMID: 37251983 PMCID: PMC10220314 DOI: 10.1016/j.omtm.2023.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 05/08/2023] [Indexed: 05/31/2023]
Abstract
Following the recent approval of both siRNA- and mRNA-based therapeutics, nucleic acid therapies are considered a game changer in medicine. Their envisioned widespread use for many therapeutic applications with an array of cellular target sites means that various administration routes will be employed. Concerns exist regarding adverse reactions against the lipid nanoparticles (LNPs) used for mRNA delivery, as PEG coatings on nanoparticles can induce severe antibody-mediated immune reactions, potentially being boosted by the inherently immunogenic nucleic acid cargo. While exhaustive information is available on how physicochemical features of nanoparticles affects immunogenicity, it remains unexplored how the fundamental choice of administration route regulates anti-particle immunity. Here, we directly compared antibody generation against PEGylated mRNA-carrying LNPs administered by the intravenous, intramuscular, or subcutaneous route, using a novel sophisticated assay capable of measuring antibody binding to authentic LNP surfaces with single-particle resolution. Intramuscular injections in mice were found to generate overall low and dose-independent levels of anti-LNP antibodies, while both intravenous and subcutaneous LNP injections generated substantial and highly dose-dependent levels. These findings demonstrate that before LNP-based mRNA medicines can be safely applied to new therapeutic applications, it will be crucial to carefully consider the choice of administration route.
Collapse
Affiliation(s)
- Rasmus Münter
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), 2800 Kongens Lyngby, Denmark
| | - Esben Christensen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), 2800 Kongens Lyngby, Denmark
| | - Thomas L. Andresen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), 2800 Kongens Lyngby, Denmark
| | - Jannik B. Larsen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), 2800 Kongens Lyngby, Denmark
| |
Collapse
|
19
|
Barman P, Joshi S, Sharma S, Preet S, Sharma S, Saini A. Strategic Approaches to Improvise Peptide Drugs as Next Generation Therapeutics. Int J Pept Res Ther 2023; 29:61. [PMID: 37251528 PMCID: PMC10206374 DOI: 10.1007/s10989-023-10524-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 05/31/2023]
Abstract
In recent years, the occurrence of a wide variety of drug-resistant diseases has led to an increase in interest in alternate therapies. Peptide-based drugs as an alternate therapy hold researchers' attention in various therapeutic fields such as neurology, dermatology, oncology, metabolic diseases, etc. Previously, they had been overlooked by pharmaceutical companies due to certain limitations such as proteolytic degradation, poor membrane permeability, low oral bioavailability, shorter half-life, and poor target specificity. Over the last two decades, these limitations have been countered by introducing various modification strategies such as backbone and side-chain modifications, amino acid substitution, etc. which improve their functionality. This has led to a substantial interest of researchers and pharmaceutical companies, moving the next generation of these therapeutics from fundamental research to the market. Various chemical and computational approaches are aiding the production of more stable and long-lasting peptides guiding the formulation of novel and advanced therapeutic agents. However, there is not a single article that talks about various peptide design approaches i.e., in-silico and in-vitro along with their applications and strategies to improve their efficacy. In this review, we try to bring different aspects of peptide-based therapeutics under one article with a clear focus to cover the missing links in the literature. This review draws emphasis on various in-silico approaches and modification-based peptide design strategies. It also highlights the recent progress made in peptide delivery methods important for their enhanced clinical efficacy. The article would provide a bird's-eye view to researchers aiming to develop peptides with therapeutic applications. Graphical Abstract
Collapse
Affiliation(s)
- Panchali Barman
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Sector 14, Chandigarh, 160014 India
| | - Shubhi Joshi
- Energy Research Centre, Panjab University, Sector 14, Chandigarh, 160014 India
| | - Sheetal Sharma
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| | - Simran Preet
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| | - Shweta Sharma
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Sector 14, Chandigarh, 160014 India
| | - Avneet Saini
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| |
Collapse
|
20
|
Tenchov R, Sasso JM, Zhou QA. PEGylated Lipid Nanoparticle Formulations: Immunological Safety and Efficiency Perspective. Bioconjug Chem 2023. [PMID: 37162501 DOI: 10.1021/acs.bioconjchem.3c00174] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Lipid nanoparticles (LNPs) have been recognized as efficient vehicles to transport a large variety of therapeutics. Currently in the spotlight as important constituents of the COVID-19 mRNA vaccines, LNPs play a significant role in protecting and transporting mRNA to cells. As one of their key constituents, polyethylene glycol (PEG)-lipid conjugates are important in defining LNP physicochemical characteristics and biological activity. PEGylation has proven particularly efficient in conferring longer systemic circulation of LNPs, thus greatly improving their pharmacokinetics and efficiency. Along with revealing the benefits of PEG conjugates, studies have revealed unexpected immune reactions against PEGylated nanocarriers such as accelerated blood clearance (ABC), involving the production of anti-PEG antibodies at initial injection, which initiates accelerated blood clearance upon subsequent injections, as well as a hypersensitivity reaction referred to as complement activation-related pseudoallergy (CARPA). Further, data have been accumulated indicating consistent yet sometimes controversial correlations between various structural parameters of the PEG-lipids, the properties of the PEGylated LNPs, and the magnitude of the observed adverse effects. Detailed knowledge and comprehension of such correlations are of foremost importance in the efforts to diminish and eliminate the undesirable immune reactions and improve the safety and efficiency of the PEGylated medicines. Here, we present an overview based on analysis of data from the CAS Content Collection regarding the PEGylated LNP immunogenicity and overall safety concerns. A comprehensive summary has been compiled outlining how various structural parameters of the PEG-lipids affect the immune responses and activities of the LNPs, with regards to their efficiency in drug delivery. This Review is thus intended to serve as a helpful resource in understanding the current knowledge in the field, in an effort to further solve the remaining challenges and to achieve full potential.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Janet M Sasso
- CAS, a division of the American Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Qiongqiong Angela Zhou
- CAS, a division of the American Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|
21
|
Bohmer M, Xue Y, Jankovic K, Dong Y. Advances in engineering and delivery strategies for cytokine immunotherapy. Expert Opin Drug Deliv 2023; 20:579-595. [PMID: 37104673 PMCID: PMC10330431 DOI: 10.1080/17425247.2023.2208344] [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: 11/01/2022] [Accepted: 04/25/2023] [Indexed: 04/29/2023]
Abstract
INTRODUCTION Cytokine immunotherapy is a growing field for the treatment of cancer, infectious disease, autoimmunity, and other ailments. Therapeutic cytokines are a class of secreted, small proteins that play a pivotal role in regulating the innate and adaptive immune system by provoking or mitigating immune responses. In the clinic, cytokines are frequently combined with other treatments, such as small molecules and monoclonal antibodies. However, the clinical translation of cytokine therapies is hindered by their short half-life, pleiotropic nature, and off-target effects, which cause diminished efficacy and severe systemic toxicity. Such toxicity limits dosage, thus resulting in suboptimal doses. Accordingly, numerous efforts have been devoted to exploring strategies to promote cytokine therapies by improving their tissue specificity and pharmacokinetics. AREAS COVERED Preclinical and clinical research into bioengineering and delivery strategies for cytokines, consisting of bioconjugation, fusion proteins, nanoparticles, and scaffold-based systems. EXPERT OPINION These approaches pave the way for the development of next-generation cytokine treatments with greater clinical benefit and reduced toxicity, circumventing such issues currently associated with cytokine therapy.
Collapse
Affiliation(s)
- Margaret Bohmer
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Yonger Xue
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Katarina Jankovic
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Pelotonia Institute for Immune-Oncology, The Ohio State University, Columbus, OH, 43210, USA
- Center for Cancer Engineering, The Ohio State University, Columbus, OH, 43210, USA
- Center for Cancer Metabolism, Department of Radiation Oncology, The Ohio State University, Columbus, OH, 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
- The Center for Clinical and Translational Science, The Ohio State University, Columbus, OH, 43210, USA
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
- Icahn Genomics Institute, Precision Immunology Institute, Department of Oncological Sciences, Tisch Cancer Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
22
|
Freire Haddad H, Roe EF, Collier JH. Expanding opportunities to engineer mucosal vaccination with biomaterials. Biomater Sci 2023; 11:1625-1647. [PMID: 36723064 DOI: 10.1039/d2bm01694j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mucosal vaccines are receiving increasing interest both for protecting against infectious diseases and for inducing therapeutic immune responses to treat non-infectious diseases. However, the mucosal barriers of the lungs, gastrointestinal tract, genitourinary tract, nasal, and oral tissues each present unique challenges for constructing efficacious vaccines. Vaccination through each of these mucosae requires transport through the mucus and across specialized epithelia to reach tissue-specific immune cells and lymphoid structures, necessitating finely tuned and multifunctional strategies. Serving as inspiration for mucosal vaccine design, pathogens have evolved elaborate, diverse, and multipronged approaches to penetrate and infect mucosae. This review is focused on biomaterials-based strategies, many inspired by pathogens, for designing mucosal vaccine platforms. Passive and active technologies are discussed, along with the microbial processes that they seek to mimic.
Collapse
Affiliation(s)
- Helena Freire Haddad
- Theodore Kennedy Professor of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA.
| | - Emily F Roe
- Theodore Kennedy Professor of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA.
| | - Joel H Collier
- Theodore Kennedy Professor of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA.
| |
Collapse
|
23
|
Bavli Y, Chen BM, Gross G, Hershko A, Turjeman K, Roffler S, Barenholz Y. Anti-PEG antibodies before and after a first dose of Comirnaty® (mRNA-LNP-based SARS-CoV-2 vaccine). J Control Release 2023; 354:316-322. [PMID: 36549393 PMCID: PMC9838877 DOI: 10.1016/j.jconrel.2022.12.039] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/29/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
The early and massive vaccination campaign in Israel with the mRNA-LNP Comirnaty® (Pfizer-BioNTech) vaccine against the SARS-CoV-2 virus made available large amounts of data regarding the efficacy and safety of this vaccine. Adverse reactions to mRNA-based SARS-CoV-2 vaccines are rare events, but due to large mediatic coverage they became feared and acted as a potential source of delay for the vaccination of the Israeli population. The experience with the reactogenicity of the polyethylene glycol (PEG) moiety of PEGylated liposomes, PEGylated proteins and other PEGylated drugs raised the fear that similar adverse effects can be associated with the PEG lipid which is an essential component of currently used mRNA-LNP vaccines against COVID-19. In this study we quantified the levels of anti-PEG IgG, IgM and IgE present in the blood of 79 volunteers immediately before and 3 weeks after receiving a first dose of Comirnaty® vaccine. Our in vitro results show that different humanized anti-PEG antibodies bind the PEGylated nano-liposomes in a concentration-dependent manner, but they bind with a lower affinity to the Comirnaty vaccine, despite it having a high mole% of neutral PEG2000-lipid on its surface. We found an increase in IgG concentration in the blood 3 weeks after the first vaccine administration, but no increase in IgM or IgE. In addition, no severe signs of adverse reactions to the Comirnaty vaccine were observed in the population studied despite the significant pre-existing high titers of IgG before the first dose of vaccine in 2 donors.
Collapse
Affiliation(s)
- Yaelle Bavli
- Laboratory of Membrane and Liposome Research, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem 9112102, Israel.
| | - Bing-Mae Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan.
| | - Guy Gross
- Bio-Samples Bank (MIDGAM) Hadassah Ein Kerem Hospital, Jerusalem 9112102, Israel.
| | - Alon Hershko
- Department of Medicine C, Hadassah Ein Kerem Hospital, Faculty of Medicine, Jerusalem 9112102, Israel.
| | - Keren Turjeman
- Laboratory of Membrane and Liposome Research, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem 9112102, Israel
| | - Steve Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Yechezkel Barenholz
- Laboratory of Membrane and Liposome Research, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem 9112102, Israel,Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
24
|
Lazewski D, Kucinska M, Potapskiy E, Kuzminska J, Popenda L, Tezyk A, Goslinski T, Wierzchowski M, Murias M. Enhanced Cytotoxic Activity of PEGylated Curcumin Derivatives: Synthesis, Structure-Activity Evaluation, and Biological Activity. Int J Mol Sci 2023; 24:ijms24021467. [PMID: 36674983 PMCID: PMC9867315 DOI: 10.3390/ijms24021467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/25/2022] [Accepted: 01/06/2023] [Indexed: 01/14/2023] Open
Abstract
Curcumin has been modified in various ways to broaden its application in medicine and address its limitations. In this study, we present a series of curcumin-based derivatives obtained by replacing the hydroxy groups in the feruloyl moiety with polyethylene glycol (PEG) chains and the addition of the BF2 moiety to the carbonyl groups. Tested compounds were screened for their cytotoxic activity toward two bladder cancer cell lines, 5637 and SCaBER, and a noncancerous cell line derived from lung fibroblasts (MRC-5). Cell viability was analyzed under normoxic and hypoxic conditions (1% oxygen). Structure-activity relationships (SARs) are discussed, and curcumin derivatives equipped within feruloyl moieties with 3-methoxy and 4-{2-[2-(2-methoxyethoxy)ethoxy]ethoxy} substituents (5) were selected for further analysis. Compound 5 did not affect the viability of MRC-5 cells and exerted a stronger cytotoxic effect under hypoxic conditions. However, the flow cytometry studies showed that PEGylation did not improve cellular uptake. Another observation was that the lack of serum proteins limits the intracellular uptake of curcumin derivative 5. The preliminary mechanism of action studies indicated that compound 5 under hypoxic conditions induced G2/M arrest in a dose-dependent manner and increased the expression of stress-related proteins such as p21/CIP1, phosphorylated HSP27, ADAMTS-1, and phosphorylated JNK. In summary, the results of the studies indicated that PEGylated curcumin is a more potent compound against bladder cancer cell lines than the parent compound, and derivative 5 is worthy of further investigation to clarify its mechanism of anticancer action under hypoxic conditions.
Collapse
Affiliation(s)
- Dawid Lazewski
- Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, Poland
| | - Malgorzata Kucinska
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30 Street, 60-631 Poznan, Poland
| | - Edward Potapskiy
- Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, Poland
| | - Joanna Kuzminska
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, Poland
| | - Lukasz Popenda
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3 Street, 61-614 Poznan, Poland
| | - Artur Tezyk
- Department of Forensic Medicine, Poznan University of Medical Sciences, Rokietnicka 10 Street, 60-806 Poznan, Poland
| | - Tomasz Goslinski
- Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, Poland
| | - Marcin Wierzchowski
- Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, Poland
| | - Marek Murias
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30 Street, 60-631 Poznan, Poland
- Center for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 10 Street, 61-614 Poznan, Poland
| |
Collapse
|
25
|
Reduced FVIII recovery associated with anti-FVIII PEG antibodies after BNT162b2 SARS-CoV-2 vaccination. Blood Adv 2022; 7:174-177. [PMID: 36306338 PMCID: PMC9635221 DOI: 10.1182/bloodadvances.2022008989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 01/07/2023] Open
|
26
|
Ju Y, Lee WS, Pilkington EH, Kelly HG, Li S, Selva KJ, Wragg KM, Subbarao K, Nguyen THO, Rowntree LC, Allen LF, Bond K, Williamson DA, Truong NP, Plebanski M, Kedzierska K, Mahanty S, Chung AW, Caruso F, Wheatley AK, Juno JA, Kent SJ. Anti-PEG Antibodies Boosted in Humans by SARS-CoV-2 Lipid Nanoparticle mRNA Vaccine. ACS NANO 2022; 16:11769-11780. [PMID: 35758934 PMCID: PMC9261834 DOI: 10.1021/acsnano.2c04543] [Citation(s) in RCA: 171] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/21/2022] [Indexed: 05/16/2023]
Abstract
Humans commonly have low level antibodies to poly(ethylene) glycol (PEG) due to environmental exposure. Lipid nanoparticle (LNP) mRNA vaccines for SARS-CoV-2 contain small amounts of PEG, but it is not known whether PEG antibodies are enhanced by vaccination and what their impact is on particle-immune cell interactions in human blood. We studied plasma from 130 adults receiving either the BNT162b2 (Pfizer-BioNTech) or mRNA-1273 (Moderna) mRNA vaccines or no SARS-CoV-2 vaccine for PEG-specific antibodies. Anti-PEG IgG was commonly detected prior to vaccination and was significantly boosted a mean of 13.1-fold (range 1.0-70.9) following mRNA-1273 vaccination and a mean of 1.78-fold (range 0.68-16.6) following BNT162b2 vaccination. Anti-PEG IgM increased 68.5-fold (range 0.9-377.1) and 2.64-fold (0.76-12.84) following mRNA-1273 and BNT162b2 vaccination, respectively. The rise in PEG-specific antibodies following mRNA-1273 vaccination was associated with a significant increase in the association of clinically relevant PEGylated LNPs with blood phagocytes ex vivo. PEG antibodies did not impact the SARS-CoV-2 specific neutralizing antibody response to vaccination. However, the elevated levels of vaccine-induced anti-PEG antibodies correlated with increased systemic reactogenicity following two doses of vaccination. We conclude that PEG-specific antibodies can be boosted by LNP mRNA vaccination and that the rise in PEG-specific antibodies is associated with systemic reactogenicity and an increase of PEG particle-leukocyte association in human blood. The longer-term clinical impact of the increase in PEG-specific antibodies induced by lipid nanoparticle mRNA vaccines should be monitored. It may be useful to identify suitable alternatives to PEG for developing next-generation LNP vaccines to overcome PEG immunogenicity in the future.
Collapse
Affiliation(s)
- Yi Ju
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
- School of Health and Biomedical Sciences,
RMIT University, Bundoora, VIC 3083,
Australia
- Department of Chemical Engineering, The
University of Melbourne, Melbourne, VIC 3000,
Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
| | - Emily H. Pilkington
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
- Department of Drug Delivery, Disposition and Dynamics,
Monash Institute of Pharmaceutical Sciences, Monash University,
Melbourne, VIC 3000, Australia
| | - Hannah G. Kelly
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
| | - Shiyao Li
- Department of Chemical Engineering, The
University of Melbourne, Melbourne, VIC 3000,
Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
| | - Kathleen M. Wragg
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
- WHO Collaborating Centre for Reference and Research on
Influenza, Peter Doherty Institute for Infection and Immunity,
Melbourne, VIC 3000, Australia
| | - Thi H. O. Nguyen
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
| | - Louise C. Rowntree
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
| | - Lilith F. Allen
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
| | - Katherine Bond
- Department of Microbiology, Royal Melbourne
Hospital, Melbourne, VIC 3000, Australia
| | - Deborah A. Williamson
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
- Department of Microbiology, Royal Melbourne
Hospital, Melbourne, VIC 3000, Australia
| | - Nghia P. Truong
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
- Department of Drug Delivery, Disposition and Dynamics,
Monash Institute of Pharmaceutical Sciences, Monash University,
Melbourne, VIC 3000, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences,
RMIT University, Bundoora, VIC 3083,
Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
| | - Siddhartha Mahanty
- Department of Infectious Diseases, Peter Doherty Institute
for Infection and Immunity, The University of Melbourne,
Melbourne, VIC 3000, Australia
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
| | - Frank Caruso
- Department of Chemical Engineering, The
University of Melbourne, Melbourne, VIC 3000,
Australia
| | - Adam K. Wheatley
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
| | - Jennifer A. Juno
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, Peter
Doherty Institute for Infection and Immunity, The University of
Melbourne, Melbourne, VIC 3000, Australia
- Melbourne Sexual Health Centre and Department of Infectious
Diseases, Alfred Hospital and Central Clinical School, Monash
University, Melbourne, VIC 3000, Australia
| |
Collapse
|
27
|
Feenstra LR, Gehring R, van Geijswijk IM, König T, Prinsen HCMT, Vandemeulebroecke K, Lammens T, Krupa A, Teske E. Evaluation of PEG-L-asparaginase in asparagine suppression and anti-drug antibody development in healthy Beagle dogs: A multi-phase preclinical study. Vet J 2022; 286:105854. [PMID: 35781075 DOI: 10.1016/j.tvjl.2022.105854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022]
Abstract
L-asparaginase is a frequently used drug in the treatment of canine malignant lymphoma. Since production and availability of native E. coli-derived L-asparaginase are limited, PEG-L-asparaginase (PEG-ASP) is an alternative. However, recommended doses and dosing intervals are mainly empirically determined. A multi-phase clinical dose-finding study with seven healthy Beagle dogs was conducted to find the minimum effective dose and, potentially, a dosing interval for PEG-ASP in dogs. Plasma concentrations of amino acids and PEG-ASP activity were measured at various time points after administration of different doses of PEG-ASP. Anti-PEG and anti-asparaginase antibody titres were measured. Administration of 10 IU/kg PEG-ASP resulted in asparagine depletion in all dogs, albeit for various durations: for 9 days in all dogs, 15 days in five dogs, 21 days in three dogs and 29 days in one dog. Asparagine suppression occurred at PEG-ASP plasma concentrations < 25 IU/L. Subsequent administrations of a second and third dose of 20 IU/kg and 40 IU/kg PEG-ASP resulted in asparagine suppression at < 9 days in five dogs, accompanied by the development of antibodies against PEG and L-asparaginase. Two dogs with prolonged asparagine suppression after the second and third administration did not develop antibodies. Marked individual variation in the mechanism and duration of response to PEG-ASP was noted. Antibody formation against PEG-ASP was frequently observed and sometimes occurred after one injection. This study suggests that PEG-ASP doses as high as the currently used dose of 40 IU/kg might not be needed in treatment of canine malignant lymphoma.
Collapse
Affiliation(s)
- L R Feenstra
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, the Netherlands.
| | - R Gehring
- Institute for Risk Assessment Sciences, Division of Toxicology and Pharmacology, Utrecht University, Utrecht, the Netherlands
| | - I M van Geijswijk
- Institute for Risk Assessment Sciences, Division of Toxicology and Pharmacology, Utrecht University, Utrecht, the Netherlands; Pharmacy Department, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - T König
- Diagnostics Development Department, medac GmbH, Wedel, Germany
| | - H C M T Prinsen
- Department of Genetics, section Metabolic Diagnostics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - K Vandemeulebroecke
- Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - T Lammens
- Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - A Krupa
- AniCura Veterinary Hospital Zeeuws-Vlaanderen, Terneuzen, the Netherlands
| | - E Teske
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, the Netherlands
| |
Collapse
|
28
|
Fay JM, Lim C, Finkelstein A, Batrakova EV, Kabanov AV. PEG-Free Polyion Complex Nanocarriers for Brain-Derived Neurotrophic Factor. Pharmaceutics 2022; 14:pharmaceutics14071391. [PMID: 35890287 PMCID: PMC9317007 DOI: 10.3390/pharmaceutics14071391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 12/10/2022] Open
Abstract
Many therapeutic formulations incorporate poly(ethylene glycol) (PEG) as a stealth component to minimize early clearance. However, PEG is immunogenic and susceptible to accelerated clearance after multiple administrations. Here, we present two novel reformulations of a polyion complex (PIC), originally composed of poly(ethylene glycol)113-b-poly(glutamic acid)50 (PEG-PLE) and brain-derived neurotrophic factor (BDNF), termed Nano-BDNF (Nano-BDNF PEG-PLE). We replace the PEG based block copolymer with two new polymers, poly(sarcosine)127-b-poly(glutamic acid)50 (PSR-PLE) and poly(methyl-2-oxazolines)38-b-poly(oxazolepropanoic acid)27-b-poly(methyl-2-oxazoline)38 (PMeOx-PPaOx-PMeOx), which are driven to association with BDNF via electrostatic interactions and hydrogen bonding to form a PIC. Formulation using a microfluidic mixer yields small and narrowly disperse nanoparticles which associate following similar principles. Additionally, we demonstrate that encapsulation does not inhibit access by the receptor kinase, which affects BDNF’s physiologic benefits. Finally, we investigate the formation of nascent nanoparticles through a series of characterization experiments and isothermal titration experiments which show the effects of pH in the context of particle self-assembly. Our findings indicate that thoughtful reformulation of PEG based, therapeutic PICs with non-PEG alternatives can be accomplished without compromising the self-assembly of the PIC.
Collapse
Affiliation(s)
- James M. Fay
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599-7362, USA; (J.M.F.); (C.L.); (E.V.B.)
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7260, USA
| | - Chaemin Lim
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599-7362, USA; (J.M.F.); (C.L.); (E.V.B.)
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599-7260, USA
| | - Anna Finkelstein
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599-7362, USA; (J.M.F.); (C.L.); (E.V.B.)
| | - Elena V. Batrakova
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599-7362, USA; (J.M.F.); (C.L.); (E.V.B.)
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599-7260, USA
| | - Alexander V. Kabanov
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599-7362, USA; (J.M.F.); (C.L.); (E.V.B.)
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599-7260, USA
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, 119992 Moscow, Russia
- Correspondence:
| |
Collapse
|
29
|
Li S, Ju Y, Zhou J, Faria M, Ang CS, Mitchell AJ, Zhong QZ, Zheng T, Kent SJ, Caruso F. Protein precoating modulates biomolecular coronas and nanocapsule-immune cell interactions in human blood. J Mater Chem B 2022; 10:7607-7621. [PMID: 35713277 DOI: 10.1039/d2tb00672c] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The biomolecular corona that forms on particles upon contact with blood plays a key role in the fate and utility of nanomedicines. Recent studies have shown that precoating nanoparticles with serum proteins can improve the biocompatibility and stealth properties of nanoparticles. However, it is not fully clear how precoating influences biomolecular corona formation and downstream biological responses. Herein, we systematically examine three precoating strategies by coating bovine serum albumin (single protein), fetal bovine serum (FBS, mixed proteins without immunoglobulins), or bovine serum (mixed proteins) on three nanoparticle systems, namely supramolecular template nanoparticles, metal-phenolic network (MPN)-coated template (core-shell) nanoparticles, and MPN nanocapsules (obtained after template removal). The effect of protein precoating on biomolecular corona compositions and particle-immune cell interactions in human blood was characterized. In the absence of a pre-coating, the MPN nanocapsules displayed lower leukocyte association, which correlated to the lower amount (by 2-3 fold) of adsorbed proteins and substantially fewer immunoglobulins (more than 100 times) in the biomolecular corona relative to the template and core-shell nanoparticles. Among the three coating strategies, FBS precoating demonstrated the most significant reduction in leukocyte association (up to 97% of all three nanoparticles). A correlation analysis highlights that immunoglobulins and apolipoproteins may regulate leukocyte recognition. This study demonstrates the impact of different precoating strategies on nanoparticle-immune cell association and the role of immunoglobulins in bio-nano interactions.
Collapse
Affiliation(s)
- Shiyao Li
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Yi Ju
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia. .,School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia.
| | - Jiajing Zhou
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Matthew Faria
- Systems Biology Laboratory, School of Mathematics and Statistics, and the Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andrew J Mitchell
- Department of Chemical Engineering, Materials Characterisation and Fabrication Platform, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Qi-Zhi Zhong
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Tian Zheng
- Department of Chemical Engineering, Materials Characterisation and Fabrication Platform, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
30
|
Kong YW, Dreaden EC. PEG: Will It Come Back to You? Polyethelyne Glycol Immunogenicity, COVID Vaccines, and the Case for New PEG Derivatives and Alternatives. Front Bioeng Biotechnol 2022; 10:879988. [PMID: 35573237 PMCID: PMC9092184 DOI: 10.3389/fbioe.2022.879988] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/11/2022] [Indexed: 11/21/2022] Open
Affiliation(s)
- Yi Wen Kong
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States
- *Correspondence: Yi Wen Kong, ; Erik C Dreaden, ,
| | - Erik C Dreaden
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
- *Correspondence: Yi Wen Kong, ; Erik C Dreaden, ,
| |
Collapse
|
31
|
Cao Y, Dong X, Chen X. Polymer-Modified Liposomes for Drug Delivery: From Fundamentals to Applications. Pharmaceutics 2022; 14:pharmaceutics14040778. [PMID: 35456613 PMCID: PMC9026371 DOI: 10.3390/pharmaceutics14040778] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Liposomes are highly advantageous platforms for drug delivery. To improve the colloidal stability and avoid rapid uptake by the mononuclear phagocytic system of conventional liposomes while controlling the release of encapsulated agents, modification of liposomes with well-designed polymers to modulate the physiological, particularly the interfacial properties of the drug carriers, has been intensively investigated. Briefly, polymers are incorporated into liposomes mainly using “grafting” or “coating”, defined according to the configuration of polymers at the surface. Polymer-modified liposomes preserve the advantages of liposomes as drug-delivery carriers and possess specific functionality from the polymers, such as long circulation, precise targeting, and stimulus-responsiveness, thereby resulting in improved pharmacokinetics, biodistribution, toxicity, and therapeutic efficacy. In this review, we summarize the progress in polymer-modified liposomes for drug delivery, focusing on the change in physiological properties of liposomes and factors influencing the overall therapeutic efficacy.
Collapse
Affiliation(s)
- Yifeng Cao
- Department of Electronic Chemicals, Institute of Zhejiang University-Quzhou, Quzhou 324000, China
- Correspondence: (Y.C.); (X.C.)
| | - Xinyan Dong
- School of Biological and Chemical Engineering, NingboTech University, Ningbo 315100, China;
| | - Xuepeng Chen
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
- Correspondence: (Y.C.); (X.C.)
| |
Collapse
|
32
|
Vincent MP, Navidzadeh JO, Bobbala S, Scott EA. Leveraging self-assembled nanobiomaterials for improved cancer immunotherapy. Cancer Cell 2022; 40:255-276. [PMID: 35148814 PMCID: PMC8930620 DOI: 10.1016/j.ccell.2022.01.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/22/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022]
Abstract
Nanomaterials and targeted drug delivery vehicles improve the therapeutic index of drugs and permit greater control over their pharmacokinetics, biodistribution, and bioavailability. Here, nanotechnologies applied to cancer immunotherapy are discussed with a focus on current and next generation self-assembling drug delivery systems composed of lipids and/or polymers. Topics covered include the fundamental design, suitability, and inherent properties of nanomaterials that induce anti-tumor immune responses and support anti-cancer vaccination. Established active and passive targeting strategies as well as newer "indirect" methods are presented together with insights into how nanocarrier structure and surface chemistry can be leveraged for controlled delivery to the tumor microenvironment while minimizing off-target effects.
Collapse
Affiliation(s)
- Michael P Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Justin O Navidzadeh
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA; Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
33
|
Angelini G, Mura G, Messina G. Therapeutic approaches to preserve the musculature in Duchenne Muscular Dystrophy: The importance of the secondary therapies. Exp Cell Res 2022; 410:112968. [PMID: 34883113 DOI: 10.1016/j.yexcr.2021.112968] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/15/2021] [Accepted: 12/04/2021] [Indexed: 02/07/2023]
Abstract
Muscular dystrophies (MDs) are heterogeneous diseases, characterized by primary wasting of skeletal muscle, which in severe cases, such as Duchenne Muscular Dystrophy (DMD), leads to wheelchair dependency, respiratory failure, and premature death. Research is ongoing to develop efficacious therapies, particularly for DMD. Most of the efforts, currently focusing on correcting or restoring the primary defect of MDs, are based on gene-addition, exon-skipping, stop codon read-through, and genome-editing. Although promising, most of them revealed several practical limitations. Shared knowledge in the field is that, in order to be really successful, any therapeutic approach has to rely on spared functional muscle tissue, restricting the number of patients eligible for clinical trials to the youngest and less compromised individuals. In line with this, many therapeutic strategies aim to preserve muscle tissue and function. This Review outlines the most interesting and recent studies addressing the secondary outcomes of DMD and how to better deliver the therapeutic agents. In the future, the effective treatment of DMD will likely require combinations of therapies addressing both the primary genetic defect and its consequences.
Collapse
Affiliation(s)
- Giuseppe Angelini
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Giada Mura
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Graziella Messina
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
34
|
Fay JM, Kabanov AV. Interpolyelectrolyte Complexes as an Emerging Technology for Pharmaceutical Delivery of Polypeptides. REVIEWS AND ADVANCES IN CHEMISTRY 2022; 12. [PMCID: PMC9987408 DOI: 10.1134/s2634827622600177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Polyelectrolyte complexes and the derivatives thereof comprise some of the most promising vehicles for the encapsulation and delivery of macromolecular therapeutics. In particular, protein therapeutics, which present a host of special considerations, can often be effectively packaged and delivered using interpolyelectrolyte complexes. While the technologies are still in the developmental phase, there are numerous examples of complexes where control is exerted over spacial and temporal delivery of a model protein cargo or candidate protein therapeutic agent. Here we provide a historical and practical background to promote a deeper understanding of interpolyelectrolyte complexes and the derivative technologies. Additionally, we review the physical principles underlying the association of polyelectrolyte complexes and the application of those principles to novel strategies and technologies driving interpolyelectrolyte complexation. Then, the application of polyelectrolyte complex technology to protein therapeutics is discussed in detail including discussions of several types of protein cargo with a special emphasis on Brain-Derived Neurotrophic Factor. Finally, we focus on the use of stealth polymers in block ionomer complexes, specifically PEG; its benefits, flaws, and possible alternatives. Comprehensive understanding of the field may promote the continued development of derivative technologies for the delivery of particularly intransigent protein therapeutics, much as has been accomplished for small molecule drugs. We also aim to link current advances to the historical developments which inaugurated the field. With consideration to the field, industrial and academic researchers can utilize the discussed technologies and continue to elucidate novel modalities for a myriad of therapeutic and commercial applications.
Collapse
Affiliation(s)
- James M. Fay
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina, NC 27599-7362 Chapel Hill, USA ,Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina, NC 27599-7260 Chapel Hill, USA
| | - Alexander V. Kabanov
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina, NC 27599-7362 Chapel Hill, USA ,Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, NC 27599-7260 Chapel Hill, USA ,Faculty of Chemistry, Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
35
|
In vivo biocompatibility evaluation of in situ-forming polyethylene glycol-collagen hydrogels in corneal defects. Sci Rep 2021; 11:23913. [PMID: 34903788 PMCID: PMC8668970 DOI: 10.1038/s41598-021-03270-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 12/01/2021] [Indexed: 12/26/2022] Open
Abstract
The available treatment options include corneal transplantation for significant corneal defects and opacity. However, shortage of donor corneas and safety issues in performing corneal transplantation are the main limitations. Accordingly, we adopted the injectable in situ-forming hydrogels of collagen type I crosslinked via multifunctional polyethylene glycol (PEG)-N-hydroxysuccinimide (NHS) for treatment and evaluated in vivo biocompatibility. The New Zealand White rabbits (N = 20) were randomly grouped into the keratectomy-only and keratectomy with PEG-collagen hydrogel-treated groups. Samples were processed for immunohistochemical evaluation. In both clinical and histologic observations, epithelial cells were able to migrate and form multilayers over the PEG-collagen hydrogels at the site of the corneal stromal defect. There was no evidence of inflammatory or immunological reactions or increased IOP for PEG-collagen hydrogel-treated corneas during the four weeks of observation. Immunohistochemistry revealed the presence of α-smooth muscle actin (α-SMA) in the superior corneal stroma of the keratectomy-only group (indicative of fibrotic healing), whereas low stromal α-SMA expression was detected in the keratectomy with PEG-collagen hydrogel-treated group. Taken together, we suggest that PEG-collagen may be used as a safe and effective alternative in treating corneal defect in clinical setting.
Collapse
|
36
|
Flögel U, Temme S, Jacoby C, Oerther T, Keul P, Flocke V, Wang X, Bönner F, Nienhaus F, Peter K, Schrader J, Grandoch M, Kelm M, Levkau B. Multi-targeted 1H/ 19F MRI unmasks specific danger patterns for emerging cardiovascular disorders. Nat Commun 2021; 12:5847. [PMID: 34615876 PMCID: PMC8494909 DOI: 10.1038/s41467-021-26146-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/20/2021] [Indexed: 12/28/2022] Open
Abstract
Prediction of the transition from stable to acute coronary syndromes driven by vascular inflammation, thrombosis with subsequent microembolization, and vessel occlusion leading to irreversible myocardial damage is still an unsolved problem. Here, we introduce a multi-targeted and multi-color nanotracer platform technology that simultaneously visualizes evolving danger patterns in the development of progressive coronary inflammation and atherothrombosis prior to spontaneous myocardial infarction in mice. Individual ligand-equipped perfluorocarbon nanoemulsions are used as targeting agents and are differentiated by their specific spectral signatures via implementation of multi chemical shift selective 19F MRI. Thereby, we are able to identify areas at high risk of and predictive for consecutive development of myocardial infarction, at a time when no conventional parameter indicates any imminent danger. The principle of this multi-targeted approach can easily be adapted to monitor also a variety of other disease entities and constitutes a technology with disease-predictive potential.
Collapse
Affiliation(s)
- Ulrich Flögel
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany.
- Cardiovascular Research Institute Düsseldorf (CARID), Heinrich Heine University, Düsseldorf, Germany.
- Department of Cardiology, Pneumology and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany.
| | - Sebastian Temme
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Heinrich Heine University, Düsseldorf, Germany
- Department of Cardiology, Pneumology and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Anesthesiology, Heinrich Heine University, Düsseldorf, Germany
| | - Christoph Jacoby
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
- Department of Cardiology, Pneumology and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Petra Keul
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| | - Vera Flocke
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Xiaowei Wang
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Florian Bönner
- Department of Cardiology, Pneumology and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Fabian Nienhaus
- Department of Cardiology, Pneumology and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Jürgen Schrader
- Cardiovascular Research Institute Düsseldorf (CARID), Heinrich Heine University, Düsseldorf, Germany
- Department of Cardiology, Pneumology and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Maria Grandoch
- Department of Pharmacology and Clinical Pharmacology, Heinrich Heine University, Düsseldorf, Germany
| | - Malte Kelm
- Cardiovascular Research Institute Düsseldorf (CARID), Heinrich Heine University, Düsseldorf, Germany
- Department of Cardiology, Pneumology and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
37
|
Wielgat P, Niemirowicz-Laskowska K, Wilczewska AZ, Car H. Sialic Acid-Modified Nanoparticles-New Approaches in the Glioma Management-Perspective Review. Int J Mol Sci 2021; 22:ijms22147494. [PMID: 34299113 PMCID: PMC8304714 DOI: 10.3390/ijms22147494] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/10/2021] [Indexed: 12/23/2022] Open
Abstract
The cell surface is covered by a dense and complex network of glycans attached to the membrane proteins and lipids. In gliomas, the aberrant sialylation, as the final stage of glycosylation, is an important regulatory mechanism of malignant cell behavior and correlates with worse prognosis. Better understanding of the role of sialylation in cellular and molecular processes opens a new way in the development of therapeutic tools for human brain tumors. According to the recent clinical observation, the cellular heterogeneity, activity of brain cancer stem cells (BCSCs), immune evasion, and function of the blood–brain barrier (BBB) are attractive targets for new therapeutic strategies. In this review, we summarize the importance of sialic acid-modified nanoparticles in brain tumor progression.
Collapse
Affiliation(s)
- Przemyslaw Wielgat
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland;
- Correspondence: (P.W.); (K.N.-L.); Tel.: +48-85-7450647 (P.W.); +48-85-7485554 (K.N.-L.)
| | - Katarzyna Niemirowicz-Laskowska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-265 Bialystok, Poland
- Correspondence: (P.W.); (K.N.-L.); Tel.: +48-85-7450647 (P.W.); +48-85-7485554 (K.N.-L.)
| | | | - Halina Car
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland;
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-265 Bialystok, Poland
| |
Collapse
|