1
|
Liu Z, Chen J, Xu M, Ho S, Wei Y, Ho HP, Yong KT. Engineered multi-domain lipid nanoparticles for targeted delivery. Chem Soc Rev 2025. [PMID: 40390667 DOI: 10.1039/d4cs00891j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Engineered lipid nanoparticles (LNPs) represent a breakthrough in targeted drug delivery, enabling precise spatiotemporal control essential to treat complex diseases such as cancer and genetic disorders. However, the complexity of the delivery process-spanning diverse targeting strategies and biological barriers-poses significant challenges to optimizing their design. To address these, this review introduces a multi-domain framework that dissects LNPs into four domains: structure, surface, payload, and environment. Engineering challenges, functional mechanisms, and characterization strategies are analyzed across each domain, along with a discussion of advantages, limitations, and in vivo fate (e.g., biodistribution and clearance). The framework also facilitates comparisons with natural exosomes and exploration of alternative administration routes, such as intranasal and intraocular delivery. We highlight current characterization techniques, such as cryo-TEM and multiscale molecular dynamics simulations, as well as the recently emerging artificial intelligence (AI) applications-ranging from LNP structure screening to the prospective use of generative models for de novo design beyond traditional experimental and simulation paradigms. Finally, we examine how engineered LNPs integrate active, passive, endogenous, and stimuli-responsive targeting mechanisms to achieve programmable delivery, potentially surpassing biological sophistication in therapeutic performance.
Collapse
Affiliation(s)
- Zhaoyu Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Jingxun Chen
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Mingkun Xu
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, 519031, China
| | - Sherwin Ho
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, 90095, USA.
| | - Yuanyuan Wei
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, 90095, USA.
| | - Ho-Pui Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.
- The Biophotonics and Mechano-Bioengineering Lab, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
2
|
Shin J, Won EJ, Xu J, Lee JC, Bang JK, Mitchell MJ, Cha-Molstad H. Transition Temperature-Guided Design of Lipid Nanoparticles for Effective mRNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2025; 17:28012-28024. [PMID: 40325908 PMCID: PMC12086761 DOI: 10.1021/acsami.5c06464] [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] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/21/2025] [Accepted: 04/27/2025] [Indexed: 05/07/2025]
Abstract
Lipid nanoparticles (LNPs) are promising mRNA delivery vehicles due to their biocompatibility and tunable characteristics. While current rational design approaches focus on ionizable lipids' pKa and zeta potential to optimize mRNA encapsulation and endosomal escape, the selection of helper lipids remains largely empirical. We propose that the lipid transition temperature (Tm), marking the shift from the gel to the liquid crystalline phase, can guide rational helper lipid selection. Through screening 54 ionizable lipids, we identified H7T4, which displayed favorable physicochemical properties when combined with its tail variants but exhibited poor transfection efficiency. Using nano differential scanning calorimetry (nDSC) and biological small-angle X-ray scattering (BioSAXS), we found that lowering the system's Tm by combining H7T4 (high transition temperature) with a low-transition-temperature helper lipid such as 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) significantly enhanced mRNA cellular uptake both in vitro and in vivo. These findings establish Tm as a crucial parameter for a rational LNP design.
Collapse
Affiliation(s)
- Jeong
Eun Shin
- Nucleic
Acid Therapeutics Research Center, Korea
Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Republic of Korea
| | - Eun-jeong Won
- Nucleic
Acid Therapeutics Research Center, Korea
Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Republic of Korea
| | - Junchao Xu
- Department
of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jong Cheol Lee
- Dandicure
Inc, Ochang, Chung Buk 28119, Republic of Korea
| | - Jeong Kyu Bang
- Division
of Magnetic Resonance, Korea Basic Science
Institute (KBSI), Ochang 28116, Republic
of Korea
- Dandicure
Inc, Ochang, Chung Buk 28119, Republic of Korea
| | - Michael J. Mitchell
- Department
of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Hyunjoo Cha-Molstad
- Nucleic
Acid Therapeutics Research Center, Korea
Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Republic of Korea
- Advanced
Bioconvergence Department, KRIBB School,
University of Science and Technology, Deajeon 34113, Republic of Korea
| |
Collapse
|
3
|
Moosavi SG, Rahiman N, Jaafari MR, Arabi L. Lipid nanoparticle (LNP) mediated mRNA delivery in neurodegenerative diseases. J Control Release 2025; 381:113641. [PMID: 40120689 DOI: 10.1016/j.jconrel.2025.113641] [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: 01/21/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Neurodegenerative diseases (NDD) are characterized by the progressive loss of neurons and the impairment of cellular functions. Messenger RNA (mRNA) has emerged as a promising therapy for treating NDD, as it can encode missing or dysfunctional proteins and anti-inflammatory cytokines or neuroprotective proteins to halt the progression of these diseases. However, effective mRNA delivery to the central nervous system (CNS) remains a significant challenge due to the limited penetration of the blood-brain barrier (BBB). Lipid nanoparticles (LNPs) offer an efficient solution by encapsulating and protecting mRNA, facilitating transfection and intracellular delivery. This review discusses the pathophysiological mechanisms of neurological disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), Huntington's disease (HD), ischemic stroke, spinal cord injury, and Friedreich's ataxia. Additionally, it explores the potential of LNP-mediated mRNA delivery as a therapeutic strategy for these diseases. Various approaches to overcoming BBB-related challenges and enhancing the delivery and efficacy of mRNA-LNPs are discussed, including non-invasive methods with strong potential for clinical translation. With advancements in artificial intelligence (AI)-guided mRNA and LNP design, targeted delivery, gene editing, and CAR-T cell therapy, mRNA-LNPs could significantly transform the treatment landscape for NDD, paving the way for future clinical applications.
Collapse
Affiliation(s)
- Seyedeh Ghazal Moosavi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Duffrène J, Muzard C, Seguin J, Izabelle C, Vrai T, Ejlalmanesh T, Bombled M, Hamdi S, Lemdani K, Alhareth K, Mignet N. Post-encapsulation methods for the preparation of mRNA-LNPs. Drug Deliv Transl Res 2025:10.1007/s13346-025-01866-0. [PMID: 40329036 DOI: 10.1007/s13346-025-01866-0] [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] [Accepted: 04/17/2025] [Indexed: 05/08/2025]
Abstract
Microfluidics mixing is the current lab-scale method used for producing mRNA-loaded lipid nanoparticles (mRNA-LNPs) thanks to reproducibility and robustness of microfluidic mixing. Despite these advantages, the production of small LNP volumes is associated with significant material waste. Given the high cost of synthetic mRNA, this waste can be a major limitation, particularly for early-stage screening of formulations. This study proposes alternative methods for mRNA-LNP formulation aiming to improve their stability for both formulation and mRNA screening, while reducing material waste on a research scale. Specifically, we investigated post-encapsulation of mRNA into pre-formed vesicles (PFVs) obtained by microfluidic mixing. These PFVs were complexed with mRNA by: (1) a microfluidic or (2) a manual pipetting method. The resulting mRNA-LNPs produced using these two post-encapsulation methods exhibit similar physicochemical properties and morphologies to those obtained by conventional microfluidic protocol. These mRNA-LNPs were assessed on in vitro and in vivo expression. mRNA-LNPs prepared by our alternative methods showed a similar transfection level compared to the conventional formulation taken as a control. The suitability of post-encapsulation methods to other lipids, mRNAs and microfluidic systems was also confirmed. This work offers robust, simple and economic alternative methods for preparing small volumes of mRNA-LNPs. The versatility of post-encapsulation methods allows to screen mRNA formulations in a wide range of laboratories. These methods could be applied to encapsulate tailored doses of mRNA and various mRNA constructs to achieve an optimal and personalized therapy.
Collapse
Affiliation(s)
- Joanna Duffrène
- Université Paris Cité, CNRS, INSERM, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, 75006, France
| | - Chloé Muzard
- Université Paris Cité, CNRS, INSERM, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, 75006, France
- Neovacs SA, Suresnes, France
| | - Johanne Seguin
- Université Paris Cité, CNRS, INSERM, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, 75006, France
| | - Charlotte Izabelle
- Université Paris Cité, INSERM, CNRS, Plateforme d'Imagerie Cellulaire et Moléculaire (PICMO), Paris, 75006, France
| | | | | | - Marianne Bombled
- Université Paris Cité, CNRS, INSERM, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, 75006, France
| | | | | | - Khair Alhareth
- Université Paris Cité, CNRS, INSERM, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, 75006, France.
| | - Nathalie Mignet
- Université Paris Cité, CNRS, INSERM, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, 75006, France.
| |
Collapse
|
5
|
Kart U, Raimbekova A, Yegorov S, Hortelano G. Immune Modulation with Oral DNA/RNA Nanoparticles. Pharmaceutics 2025; 17:609. [PMID: 40430900 PMCID: PMC12115334 DOI: 10.3390/pharmaceutics17050609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 05/29/2025] Open
Abstract
The oral delivery of DNA/RNA nanoparticles represents a transformative approach in immunotherapy and vaccine development. These nanoparticles enable targeted immune modulation by delivering genetic material to specific cells in the gut-associated immune system, triggering both mucosal and systemic immune responses. Unlike parenteral administration, the oral route offers a unique immunological environment that supports both tolerance and activation, depending on the formulation design. This review explores the underlying mechanisms of immune modulation by DNA/RNA nanoparticles, their design and delivery strategies, and recent advances in their application. Emphasis is placed on strategies to overcome physiological barriers such as acidic pH, enzymatic degradation, mucus entrapment, and epithelial tight junctions. Special attention is given to the role of gut-associated lymphoid tissue in mediating immune responses and the therapeutic potential of these systems in oral vaccine platforms, food allergies, autoimmune diseases, and chronic inflammation. Despite challenges, recent advances in nanoparticle formulation support the translation of these technologies into clinical applications for both therapeutic immunomodulation and vaccination.
Collapse
Affiliation(s)
| | | | | | - Gonzalo Hortelano
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, 53 Kabanbay Batyr Ave., Astana 010000, Kazakhstan; (U.K.); (A.R.); (S.Y.)
| |
Collapse
|
6
|
Iwakawa K, Sato R, Konaka M, Yamada Y, Harashima H, Sato Y. Cubic Phase-Inducible Zwitterionic Phospholipids Improve the Functional Delivery of mRNA. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413016. [PMID: 39960324 PMCID: PMC12061338 DOI: 10.1002/advs.202413016] [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] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/07/2025] [Indexed: 05/10/2025]
Abstract
Lipid nanoparticles (LNPs) are clinically advanced delivery systems for RNA. The extensively developed structure of ionizable lipids greatly contributes to the functional delivery of mRNA. However, endosomal escape is one of the severe biological barriers that continue to render this process inefficient (e.g., less than 10%). Although LNPs contain phospholipids, their role is poorly understood, and there have been few attempts to perform the chemical engineering required to improve their functionality. Herein, a cubic phase-inducible fusogenic zwitterionic phospholipid derived from 1,2-dioleoyl-3-sn-glycero-phosphoethanolamine (DOPE), DOPE-Cx is described, that is designed to correct this problem. The orientation of a zwitterionic head group of DOPE is engineered by attaching a series of hydrophobic moieties for zwitterionic intermolecular interaction with the head structure of phosphatidylcholine (PC), and this is followed by a lipid-phase transition into non-lamellar phases to facilitate membrane fusion-mediated endosomal escape. A structure-activity relationship study reveals that DOPE-Cx lipids with small hydrophobic chains induce cubic phases instead of a hexagonal phase when mixed with PC, which enhances the functional delivery of mRNA in the liver as opposed to the action of the typically utilized and naturally occurring phospholipids. Engineered functionalized phospholipids will be of great value for the therapeutic applications of mRNAs.
Collapse
Affiliation(s)
- Kazuki Iwakawa
- Laboratory for Molecular Design of PharmaceuticsFaculty of Pharmaceutical SciencesHokkaido UniversityKita‐12 Nishi‐6, Kita‐kuSapporo060‐0812Japan
| | - Rikako Sato
- Laboratory for Molecular Design of PharmaceuticsFaculty of Pharmaceutical SciencesHokkaido UniversityKita‐12 Nishi‐6, Kita‐kuSapporo060‐0812Japan
| | - Mariko Konaka
- Laboratory for Molecular Design of PharmaceuticsFaculty of Pharmaceutical SciencesHokkaido UniversityKita‐12 Nishi‐6, Kita‐kuSapporo060‐0812Japan
| | - Yuma Yamada
- Laboratory for Molecular Design of PharmaceuticsFaculty of Pharmaceutical SciencesHokkaido UniversityKita‐12 Nishi‐6, Kita‐kuSapporo060‐0812Japan
- Laboratory of Innovative NanomedicineFaculty of Pharmaceutical SciencesHokkaido UniversityKita‐12 Nishi‐6, Kita‐kuSapporo060‐0812Japan
| | - Hideyoshi Harashima
- Laboratory of Innovative NanomedicineFaculty of Pharmaceutical SciencesHokkaido UniversityKita‐12 Nishi‐6, Kita‐kuSapporo060‐0812Japan
| | - Yusuke Sato
- Laboratory for Molecular Design of PharmaceuticsFaculty of Pharmaceutical SciencesHokkaido UniversityKita‐12 Nishi‐6, Kita‐kuSapporo060‐0812Japan
- Laboratory of Innovative NanomedicineFaculty of Pharmaceutical SciencesHokkaido UniversityKita‐12 Nishi‐6, Kita‐kuSapporo060‐0812Japan
| |
Collapse
|
7
|
Sarvepalli S, Pasika SR, Verma V, Thumma A, Bolla S, Nukala PK, Butreddy A, Bolla PK. A Review on the Stability Challenges of Advanced Biologic Therapeutics. Pharmaceutics 2025; 17:550. [PMID: 40430843 PMCID: PMC12114724 DOI: 10.3390/pharmaceutics17050550] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 05/29/2025] Open
Abstract
Advanced biotherapeutic systems such as gene therapy, mRNA lipid nanoparticles, antibody-drug conjugates, fusion proteins, and cell therapy have proven to be promising platforms for delivering targeted biologic therapeutics. Preserving the intrinsic stability of these advanced therapeutics is essential to maintain their innate structure, functionality, and shelf life. Nevertheless, various challenges and obstacles arise during formulation development and throughout the storage period due to their complex nature and sensitivity to various stress factors. Key stability concerns include physical degradation and chemical instability due to various factors such as fluctuations in pH and temperature, which results in conformational and colloidal instabilities of the biologics, adversely affecting their quality and therapeutic efficacy. This review emphasizes key stability issues associated with these advanced biotherapeutic systems and approaches to identify and overcome them. In gene therapy, the brittleness of viral vectors and gene encapsulation limits their stability, requiring the use of stabilizers, excipients, and lyophilization. Keeping cells viable throughout the whole cell therapy process, from culture to final formulation, is still a major difficulty. In mRNA therapeutics, stabilization strategies such as the optimization of mRNA nucleotides and lipid compositions are used to address the instability of both the mRNA and lipid nanoparticles. Monoclonal antibodies are colloidally and conformationally unstable. Hence, buffers and stabilizers are useful to maintain stability. Although fusion proteins and monoclonal antibodies share structural similarities, they show a similar pattern of instability. Antibody-drug conjugates possess issues with conjugation and linker stability. This review outlines the stability issues associated with advanced biotherapeutics and provides insights into the approaches to address these challenges.
Collapse
Affiliation(s)
- Sruthi Sarvepalli
- College of Pharmacy and Health Sciences, St John’s University, Queens, New York, NY 11439, USA; (S.S.); (P.K.N.)
| | - Shashank Reddy Pasika
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research—Raebareli (NIPER-R), Raebareli 226002, India;
| | - Vartika Verma
- Laboratory of Translational Research in Nanomedicines, Lifecare Innovations Private Limited, Lucknow 226021, India;
| | - Anusha Thumma
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Sandeep Bolla
- Department of Statistical Programming, Fortrea, Durham, NC 27709, USA;
| | - Pavan Kumar Nukala
- College of Pharmacy and Health Sciences, St John’s University, Queens, New York, NY 11439, USA; (S.S.); (P.K.N.)
| | - Arun Butreddy
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
| | - Pradeep Kumar Bolla
- Department of Biomedical Engineering, College of Engineering, The University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
8
|
Peña Á, Heredero J, Blandín B, Mata E, De Miguel D, Toro A, Alejo T, Casabona D, López A, Gallego-Lleyda A, Pérez-Herrán E, Martínez-Oliván J, Giménez-Warren J. Multicomponent thiolactone-based ionizable lipid screening platform for efficient and tunable mRNA delivery to the lungs. Commun Chem 2025; 8:116. [PMID: 40234552 PMCID: PMC12000586 DOI: 10.1038/s42004-025-01516-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 04/04/2025] [Indexed: 04/17/2025] Open
Abstract
Ionizable lipids are essential components of lipid nanoparticles (LNPs) for efficient mRNA delivery. However, designing them for high protein expression, endosomal escape, and organ targeting is challenging due to complex structure-activity relationships. Here, we present a high-throughput platform for screening ionizable lipids using a two-step, scalable, one-pot reaction. This enabled the synthesis and vivo screening of 91 new lipids, followed by a structure-activity study, leading to the development of CP-LC-0729, which significantly surpasses the MC3 benchmark in protein expression with preliminary studies showing no in vivo toxicity. Additionally, a one-step strategy helped to yield a permanently cationic lipid which was tested in a fifth-lipid formulation, showing a highly selective lung delivery with a 32-fold increase in protein expression in vivo, outperforming current endogenous targeting strategies. All these findings underscore the potential of lipid CP-LC-0729 and our lipid platform in advancing the efficiency and specificity of mRNA delivery systems.
Collapse
Affiliation(s)
- Álvaro Peña
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Juan Heredero
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Beatriz Blandín
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Elena Mata
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Diego De Miguel
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Alfonso Toro
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Teresa Alejo
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Diego Casabona
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Alexandre López
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Ana Gallego-Lleyda
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Esther Pérez-Herrán
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain
| | - Juan Martínez-Oliván
- Certest Pharma, Certest Biotec S. L., 50840, San Mateo de Gállego, Zaragoza, Spain.
| | | |
Collapse
|
9
|
Wang HL, Kajbaf K, Gau BC, Dawdy AW, Edwards R, Bare B, Raymundo G, Iturrizaga J, Ernsky C, Walker M, Byrne EB, Boslett J, Campbell A, Matthessen R, Goffin B, Cirelli D, Rouse JC, Van Pottelberge R, Friese OV. A novel in-vitro expression assay by LC/MS/MS enables multi-antigen mRNA vaccine characterization. Sci Rep 2025; 15:10336. [PMID: 40133349 PMCID: PMC11937333 DOI: 10.1038/s41598-025-94616-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025] Open
Abstract
The new era of messenger RNA (mRNA) vaccines has led to development of a novel, state-of-the-art characterization method for this class of molecules. Currently, flow cytometry-based assays with antigen-specific antibodies are utilized for monitoring in-vitro expression (IVE) of mRNA. Here we present development, optimization, and application of an in-vitro expression liquid chromatography tandem mass spectrometry (IVE-LC/MS/MS) assay as an orthogonal method to IVE-flow cytometry that can be used for in-depth characterization of the expressed protein antigens and monitoring their relative expression levels in the cell post-mRNA transfection. The IVE-LC/MS/MS assessment accomplished the detection of influenza hemagglutinin (HA) antigens of four distinct strains simultaneously. The workflow is presented here, highlighting the optimization of all necessary steps required for protein purification and mass spectrometry method setup. The IVE-LC/MS/MS assay is a robust and versatile technique that complements the IVE-flow cytometry method and offers several advantages, such as being antibody-free, capable of multiplexing, and highly sensitive and selective. The various studies in this work, including evaluating dose-response relationships, refining transfection protocols, and examining mRNA-LNP stability under various conditions showcase the significant benefits of applying IVE-LC/MS/MS across different experimental settings. IVE-LC/MS/MS is a powerful tool for understanding and improving the performance and quality of mRNA LNPs.
Collapse
Affiliation(s)
- Hanliu Leah Wang
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA
| | - Kimia Kajbaf
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA
| | - Brian C Gau
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA
| | - Andrew W Dawdy
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA
| | - Rachel Edwards
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA
| | - Bradley Bare
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA
| | - Gianna Raymundo
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA
| | - Jose Iturrizaga
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA
| | - Chase Ernsky
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA
| | - Michael Walker
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA
| | - Emilia B Byrne
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA
| | - James Boslett
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, USA
| | - Adam Campbell
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, USA
| | - Roman Matthessen
- Drug Product Center of Excellence, Pfizer Manufacturing Belgium, Puurs‑Sint‑Amands, Belgium
| | - Ben Goffin
- Drug Product Center of Excellence, Pfizer Manufacturing Belgium, Puurs‑Sint‑Amands, Belgium
| | - David Cirelli
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Andover, MA, USA
| | - Jason C Rouse
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Andover, MA, USA
| | - Robbe Van Pottelberge
- Drug Product Center of Excellence, Pfizer Manufacturing Belgium, Puurs‑Sint‑Amands, Belgium.
| | - Olga V Friese
- BioTherapeutics Pharmaceutical Sciences, Pfizer Inc, Chesterfield, MO, USA.
| |
Collapse
|
10
|
Tang S, Huang L, Ge J, Li J, Qiu M, Zhang Y, Long M, Wu G, Zhang R, Ma X, Xia Q, Wan P, Yang T. Influence of salt solution on the physicochemical properties and in vitro/ in vivo expression of mRNA/LNP. J Nanobiotechnology 2025; 23:223. [PMID: 40108620 PMCID: PMC11921543 DOI: 10.1186/s12951-025-03318-w] [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: 01/13/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Lipid nanoparticles (LNPs) have revolutionized nucleic acid delivery, enabling significant advances in mRNA-based therapeutics. While extensive research has focused on lipid composition, the impact of preparation solutions on LNP performance remains underexplored. This study systematically investigated the effects of pH, salt type, and concentration across key preparation solutions-mRNA aqueous, dilution, exchange, and storage solutions-on the physicochemical properties, stability, and expression efficiency of SM102-based mRNA/LNPs. Findings revealed that the pH of the mRNA aqueous solution was critical, with a pH of 4 optimizing encapsulation efficiency (EE) and cellular expression. The exchange solution's pH significantly influenced biodistribution, particularly liver-specific expression following intravenous and intramuscular administration. Sucrose was identified as essential for freeze-thaw stability, with a 300 mM concentration minimizing aggregation and mRNA leakage. Furthermore, preparation solutions were shown to influence the structural integrity of LNPs, impacting their in vivo and in vitro performance. These insights highlight the importance of preparation conditions in optimizing LNP formulations for clinical applications, offering a foundation for enhanced therapeutic design and delivery.
Collapse
Affiliation(s)
- Siyuan Tang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lei Huang
- Micro & Nano Inc, Shanghai, 200127, China
| | - Jiahao Ge
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jie Li
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mingxia Qiu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yiqing Zhang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mei Long
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Gang Wu
- Micro & Nano Inc, Shanghai, 200127, China
| | - Rui Zhang
- XGen Bio Inc, Woburn, MA, 01801, USA
| | - Xueyun Ma
- Laboratory Animal Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
- Shanghai Institute of Organ Transplantation, Shanghai, 200127, China.
- Shanghai Organ Transplantation and Immune Engineering Technology Research Center, Shanghai, 200127, China.
| | - Ping Wan
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Taihua Yang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
11
|
Shi Y, Mao J, Wang S, Ma S, Luo L, You J. Pharmaceutical strategies for optimized mRNA expression. Biomaterials 2025; 314:122853. [PMID: 39342919 DOI: 10.1016/j.biomaterials.2024.122853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Messenger RNA (mRNA)-based immunotherapies and protein in situ production therapies hold great promise for addressing theoretically all the diseases characterized by aberrant protein levels. The safe, stable, and precise delivery of mRNA to target cells via appropriate pharmaceutical strategies is a prerequisite for its optimal efficacy. In this review, we summarize the structural characteristics, mode of action, development prospects, and limitations of existing mRNA delivery systems from a pharmaceutical perspective, with an emphasis on the impacts from formulation adjustments and preparation techniques of non-viral vectors on mRNA stability, target site accumulation and transfection efficiency. In addition, we introduce strategies for synergistical combination of mRNA and small molecules to augment the potency or mitigate the adverse effects of mRNA therapeutics. Lastly, we delve into the challenges impeding the development of mRNA drugs while exploring promising avenues for future advancements.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Jiapeng Mao
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Siyao Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, 166 Qiutaobei Road, Hangzhou, Zhejiang, 310017, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China; The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China; Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, 321299, PR China.
| |
Collapse
|
12
|
Imiołek M, Cojocaru R, Fekete S, Le Huray J, Lauber M. High-Throughput Quantification and Characterization of Dual Payload mRNA/LNP Cargo via Deformulating Size Exclusion and Ion Pairing Reversed Phase Assays. Anal Chem 2025; 97:3091-3098. [PMID: 39883007 PMCID: PMC11822733 DOI: 10.1021/acs.analchem.4c06296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 01/31/2025]
Abstract
Therapeutic drugs and multivalent vaccines based on the delivery of mRNA via lipid nanoparticle (LNP) technologies are expected to dominate the biopharmaceutical industry landscape in the coming years. Many of these innovative therapies include several nucleic acid components (e.g., nuclease mRNA and guide RNA) posing unique analytical challenges when monitoring the quantity and quality of each individual payload substance in the formulated LNP. Current methods were optimized for single payload analysis and often lack resolving power needed to investigate nucleic acid mixtures. Ion pairing reversed phase (IP-RP) and size exclusion chromatography (SEC) are increasingly being used to characterize nucleic acids. Here, we studied their application for payload quantification in formulated LNP drug-like products. Using a detergent to disrupt the LNPs, the liberated payloads can be separated on an octadecyl RP column using a fast gradient. Reproducible results were obtained as lipids, and surfactants were efficiently eluted using a high organic solvent wash protocol. Alternatively, we also established an online SEC disruption analysis of the mRNA/LNPs wherein an alcohol and detergent containing a mobile phase was applied. Such conditions universally deformulated all tested LNP samples, indicating that a 5 min-long SEC separation can be used as a high-throughput platform method. In both approaches, the measurements facilitate a multiattribute analysis. Apart from quantitation, the characterization of specific impurities is achieved: IP-RP reveals mRNA-lipid adducts, while SEC informs on size variants, which in turn reduces a laboratory's analytical workload. These easy-to-adopt LC-based assays are expected to fortify the analytical toolbox for emerging gene therapeutics.
Collapse
Affiliation(s)
- Mateusz Imiołek
- Waters
Corporation, Rue Michel Servet 1 Geneva, 1211, Switzerland
| | - Razvan Cojocaru
- Acuitas
Therapeutics, 6190 Agronomy
Rd. Suite 405, Vancouver, British Columbia V6T 1Z3, Canada
| | - Szabolcs Fekete
- Waters
Corporation, Rue Michel Servet 1 Geneva, 1211, Switzerland
| | - Jon Le Huray
- Acuitas
Therapeutics, 6190 Agronomy
Rd. Suite 405, Vancouver, British Columbia V6T 1Z3, Canada
| | - Matthew Lauber
- Waters
Corporation, 34 Maple St., Milford, Massachusetts 01757, United States
| |
Collapse
|
13
|
Chentoufi AA, Ulmer JB, BenMohamed L. Antigen Delivery Platforms for Next-Generation Coronavirus Vaccines. Vaccines (Basel) 2024; 13:30. [PMID: 39852809 PMCID: PMC11769099 DOI: 10.3390/vaccines13010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/15/2024] [Accepted: 12/21/2024] [Indexed: 01/26/2025] Open
Abstract
The COVID-19 pandemic, caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), is in its sixth year and is being maintained by the inability of current spike-alone-based COVID-19 vaccines to prevent transmission leading to the continuous emergence of variants and sub-variants of concern (VOCs). This underscores the critical need for next-generation broad-spectrum pan-Coronavirus vaccines (pan-CoV vaccine) to break this cycle and end the pandemic. The development of a pan-CoV vaccine offering protection against a wide array of VOCs requires two key elements: (1) identifying protective antigens that are highly conserved between passed, current, and future VOCs; and (2) developing a safe and efficient antigen delivery system for induction of broad-based and long-lasting B- and T-cell immunity. This review will (1) present the current state of antigen delivery platforms involving a multifaceted approach, including bioinformatics, molecular and structural biology, immunology, and advanced computational methods; (2) discuss the challenges facing the development of safe and effective antigen delivery platforms; and (3) highlight the potential of nucleoside-modified mRNA encapsulated in lipid nanoparticles (LNP) as the platform that is well suited to the needs of a next-generation pan-CoV vaccine, such as the ability to induce broad-based immunity and amenable to large-scale manufacturing to safely provide durable protective immunity against current and future Coronavirus threats.
Collapse
Affiliation(s)
- Aziz A. Chentoufi
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA;
| | - Jeffrey B. Ulmer
- Department of Vaccines and Immunotherapies, TechImmune, LLC, University Lab Partners, Irvine, CA 92660, USA;
| | - Lbachir BenMohamed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA;
- Department of Vaccines and Immunotherapies, TechImmune, LLC, University Lab Partners, Irvine, CA 92660, USA;
- Institute for Immunology, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
14
|
Kafle U, Truong HQ, Nguyen CTG, Meng F. Development of Thermally Stable mRNA-LNP Delivery Systems: Current Progress and Future Prospects. Mol Pharm 2024; 21:5944-5959. [PMID: 39529245 DOI: 10.1021/acs.molpharmaceut.4c00826] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The success of mRNA-LNP-based COVID-19 vaccines opens a new era for mRNA-LNP-based therapy. This breakthrough is expected to catalyze the development of more mRNA-LNP-based medicines, not only for preventive vaccines but also for therapeutic purposes. Despite the promising outlook, there are fundamental challenges impeding the progress and widespread application of mRNA-LNP formulations. One of the significant challenges is their thermal instability, requiring these products to be stored at ultralow temperatures for long-term stability. The specific requirements present significant challenges for the storage, transportation, and distribution of mRNA-LNP formulations. To effectively prepare for future infectious disease outbreaks and broaden the application of mRNA-LNP-based therapies for other illnesses, improving the thermostability of mRNA-LNP formulations is critical. In this review, we discuss the potential factors contributing to the thermal instability of mRNA-LNP formulations and examine the roles of key components such as ionizable lipids, cholesterol, pH, buffers, and stabilizing agents like sugars in maintaining their thermal stability, with the goal of providing insights that can guide the future development of thermally stable mRNA-LNP formulations.
Collapse
Affiliation(s)
- Urmila Kafle
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| | - Hoang Quan Truong
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| | - Cao Thuy Giang Nguyen
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| | - Fanfei Meng
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| |
Collapse
|
15
|
Mrksich K, Padilla MS, Mitchell MJ. Breaking the final barrier: Evolution of cationic and ionizable lipid structure in lipid nanoparticles to escape the endosome. Adv Drug Deliv Rev 2024; 214:115446. [PMID: 39293650 PMCID: PMC11900896 DOI: 10.1016/j.addr.2024.115446] [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: 07/05/2024] [Revised: 08/18/2024] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
In the past decade, nucleic acid therapies have seen a boon in development and clinical translation largely due to advances in nanotechnology that have enabled their safe and targeted delivery. Nanoparticles can protect nucleic acids from degradation by serum enzymes and can facilitate entry into cells. Still, achieving endosomal escape to allow nucleic acids to enter the cytoplasm has remained a significant barrier, where less than 5% of nanoparticles within the endo-lysosomal pathway are able to transfer their cargo to the cytosol. Lipid-based drug delivery vehicles, particularly lipid nanoparticles (LNPs), have been optimized to achieve potent endosomal escape, and thus have been the vector of choice in the clinic as demonstrated by their utilization in the COVID-19 mRNA vaccines. The success of LNPs is in large part due to the rational design of lipids that can specifically overcome endosomal barriers. In this review, we chart the evolution of lipid structure from cationic lipids to ionizable lipids, focusing on structure-function relationships, with a focus on how they relate to endosomal escape. Additionally, we examine recent advancements in ionizable lipid structure as well as discuss the future of lipid design.
Collapse
Affiliation(s)
- Kaitlin Mrksich
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marshall S Padilla
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
16
|
Hashiba K, Taguchi M, Sakamoto S, Otsu A, Maeda Y, Suzuki Y, Ebe H, Okazaki A, Harashima H, Sato Y. Impact of Lipid Tail Length on the Organ Selectivity of mRNA-Lipid Nanoparticles. NANO LETTERS 2024; 24. [PMID: 39373269 PMCID: PMC11487653 DOI: 10.1021/acs.nanolett.4c02566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/08/2024]
Abstract
The delivery of mRNA molecules to organs beyond the liver is valuable for therapeutic applications. Functionalized lipid nanoparticles (LNPs) using exogenous mechanisms can regulate in vivo mRNA expression profiles from hepatocytes to extrahepatic tissues but lead to process complexity and cost escalation. Here, we report that mRNA expression gradually shifts from the liver to the spleen in an ionizable lipid tail length-dependent manner. Remarkably, this simple chemical strategy held true even when different ionizable lipid head structures were employed. As a potential mechanism underlying this discovery, our data suggest that 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) is enriched on the surface of mRNA/LNPs with short-tail lipids. This feature limits their interaction with biological components, avoiding their rapid hepatic clearance. We also show that spleen-targeting LNPs loaded with SARS-CoV-2 receptor-binding domain (RBD) mRNA can efficiently induce immune responses and neutralize activity following intramuscular vaccination priming and boosting.
Collapse
Affiliation(s)
- Kazuki Hashiba
- Nucleic
Acid Medicine Business Division, Nitto Denko
Corporation, 1-1-2, Shimohozumi, Ibaraki, Osaka 567-8680, Japan
| | - Masamitsu Taguchi
- Nucleic
Acid Medicine Business Division, Nitto Denko
Corporation, 1-1-2, Shimohozumi, Ibaraki, Osaka 567-8680, Japan
| | - Sachiko Sakamoto
- Nucleic
Acid Medicine Business Division, Nitto Denko
Corporation, 1-1-2, Shimohozumi, Ibaraki, Osaka 567-8680, Japan
| | - Ayaka Otsu
- Nucleic
Acid Medicine Business Division, Nitto Denko
Corporation, 1-1-2, Shimohozumi, Ibaraki, Osaka 567-8680, Japan
| | - Yoshiki Maeda
- Nucleic
Acid Medicine Business Division, Nitto Denko
Corporation, 1-1-2, Shimohozumi, Ibaraki, Osaka 567-8680, Japan
| | - Yuichi Suzuki
- Laboratory
for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-Ku, Sapporo 060-0812, Japan
| | - Hirofumi Ebe
- Nucleic
Acid Medicine Business Division, Nitto Denko
Corporation, 1-1-2, Shimohozumi, Ibaraki, Osaka 567-8680, Japan
| | - Arimichi Okazaki
- Nucleic
Acid Medicine Business Division, Nitto Denko
Corporation, 1-1-2, Shimohozumi, Ibaraki, Osaka 567-8680, Japan
| | - Hideyoshi Harashima
- Laboratory
for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-Ku, Sapporo 060-0812, Japan
| | - Yusuke Sato
- Laboratory
for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-Ku, Sapporo 060-0812, Japan
| |
Collapse
|
17
|
Niazi SK. The United States Food and Drug Administration's Platform Technology Designation to Expedite the Development of Drugs. Pharmaceutics 2024; 16:918. [PMID: 39065616 PMCID: PMC11279857 DOI: 10.3390/pharmaceutics16070918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Drug development costs can be significantly reduced if proven "platform" technologies are allowed to be used without having to validate their use. The most recent US Food and Drug Administration (FDA) guideline brings more clarity, as well as a greater focus on the most complex technologies that can now be used for faster drug development. The FDA has highlights the use of lipid nanoparticles (LNPs) to package and deliver mRNA vaccines, gene therapy, and short (2-20 length) synthetic nucleotides (siRNA). Additionally, monoclonal antibody cell development is targeted. The FDA provides a systematic process of requesting platform status to benefit from its advantages. It brings advanced science and rationality into regulatory steps for the FDA's approval of drugs and biologicals.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|