1
|
Wu H, Dong L, Jin S, Zhao Y, Zhu L. Innovative gene delivery systems for retinal disease therapy. Neural Regen Res 2026; 21:542-552. [PMID: 39665817 DOI: 10.4103/nrr.nrr-d-24-00797] [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: 08/13/2024] [Accepted: 11/10/2024] [Indexed: 12/13/2024] Open
Abstract
The human retina, a complex and highly specialized structure, includes multiple cell types that work synergistically to generate and transmit visual signals. However, genetic predisposition or age-related degeneration can lead to retinal damage that severely impairs vision or causes blindness. Treatment options for retinal diseases are limited, and there is an urgent need for innovative therapeutic strategies. Cell and gene therapies are promising because of the efficacy of delivery systems that transport therapeutic genes to targeted retinal cells. Gene delivery systems hold great promise for treating retinal diseases by enabling the targeted delivery of therapeutic genes to affected cells or by converting endogenous cells into functional ones to facilitate nerve regeneration, potentially restoring vision. This review focuses on two principal categories of gene delivery vectors used in the treatment of retinal diseases: viral and non-viral systems. Viral vectors, including lentiviruses and adeno-associated viruses, exploit the innate ability of viruses to infiltrate cells, which is followed by the introduction of therapeutic genetic material into target cells for gene correction. Lentiviruses can accommodate exogenous genes up to 8 kb in length, but their mechanism of integration into the host genome presents insertion mutation risks. Conversely, adeno-associated viruses are safer, as they exist as episomes in the nucleus, yet their limited packaging capacity constrains their application to a narrower spectrum of diseases, which necessitates the exploration of alternative delivery methods. In parallel, progress has also occurred in the development of novel non-viral delivery systems, particularly those based on liposomal technology. Manipulation of the ratios of hydrophilic and hydrophobic molecules within liposomes and the development of new lipid formulations have led to the creation of advanced non-viral vectors. These innovative systems include solid lipid nanoparticles, polymer nanoparticles, dendrimers, polymeric micelles, and polymeric nanoparticles. Compared with their viral counterparts, non-viral delivery systems offer markedly enhanced loading capacities that enable the direct delivery of nucleic acids, mRNA, or protein molecules into cells. This bypasses the need for DNA transcription and processing, which significantly enhances therapeutic efficiency. Nevertheless, the immunogenic potential and accumulation toxicity associated with non-viral particulate systems necessitates continued optimization to reduce adverse effects in vivo . This review explores the various delivery systems for retinal therapies and retinal nerve regeneration, and details the characteristics, advantages, limitations, and clinical applications of each vector type. By systematically outlining these factors, our goal is to guide the selection of the optimal delivery tool for a specific retinal disease, which will enhance treatment efficacy and improve patient outcomes while paving the way for more effective and targeted therapeutic interventions.
Collapse
Affiliation(s)
- Hongguang Wu
- Department of Ophthalmology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | |
Collapse
|
2
|
Baghirov H. Mechanisms of receptor-mediated transcytosis at the blood-brain barrier. J Control Release 2025; 381:113595. [PMID: 40056994 DOI: 10.1016/j.jconrel.2025.113595] [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: 12/21/2024] [Revised: 02/09/2025] [Accepted: 02/28/2025] [Indexed: 04/15/2025]
Abstract
In receptor-mediated transcytosis (RMT) of large therapeutics across the blood-brain barrier (BBB), the construct - a macromolecule or a larger carrier with therapeutic payload - binds a protein on brain capillary endothelial cells (BCEC), with internalization and release into the brain parenchyma. The construct's internalization into, trafficking across and release from, but also possible entrapment within BCEC are affected by its engineered properties whose optimization has helped derive insights into transport mechanisms at BCEC. Furthermore, advances in multi-omics, as well as large-scale screening and directed evolution campaigns have helped identify new targets for RMT at BCEC. In this perspective, I raise and reflect on some fundamental questions one can arrive at by comparing the engineered properties of BBB-targeted constructs and the properties of different target proteins. These questions concern the underlying, transcytosis-promoting factors that the optimization of constructs' engineered properties appears to converge on, the precise role of target proteins in RMT, the different mechanisms through which these targets may mediate construct trafficking, and the tentative criteria for target selection on BCEC. Based on these considerations I propose several scenarios and strategies to interfere with the construct's trafficking for more efficient internalization, transport through the endosomal network toward the abluminal membrane, and release from BCEC, both for smaller macromolecules and for larger carriers.
Collapse
Affiliation(s)
- Habib Baghirov
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland.
| |
Collapse
|
3
|
Liang H, Xing Y, Wang K, Zhang Y, Yin F, Li Z. Peptides: potential delivery systems for mRNA. RSC Chem Biol 2025; 6:666-677. [PMID: 40071030 PMCID: PMC11891934 DOI: 10.1039/d4cb00295d] [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: 12/02/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
mRNA-based therapies have broad applications in various disease treatments and have been applied in protein replacement therapy, gene editing, and vaccine development. Numerous research studies have been carried out aiming to increase the stability of mRNA, improve its translational efficiency, and reduce its immunogenicity. However, given mRNA's large molecular size and strong electronegativity, the safety and efficient delivery of mRNA into the target cells remains the critical rate-limiting step in current mRNA drug development. Various nanocarriers, such as liposomes, lipid nanoparticles, polyetherimide, and mesoporous silica nanoparticles, have been employed for mRNA delivery in the past few decades. Among them, peptides have demonstrated great potential as promising carrier candidates for mRNA delivery due to their high cell membrane permeability, good biocompatibility, definite chemical structure, and ease of preparation. Here, peptide-based mRNA delivery systems are systematically analyzed, including their construction strategies, mechanisms of action in mRNA delivery, and the application limitations or challenges. It is hoped that this review will guide the design, optimization, and applications of peptide carriers in mRNA-based drug development.
Collapse
Affiliation(s)
- Huiting Liang
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory Shenzhen 518118 China
| | - Yun Xing
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| | - Kexin Wang
- Department of Chemistry, Southern University of Science and Technology Shenzhen 518055 China
| | - Yaping Zhang
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory Shenzhen 518118 China
| | - Feng Yin
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory Shenzhen 518118 China
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| | - Zigang Li
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory Shenzhen 518118 China
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| |
Collapse
|
4
|
Liu Z, Wu J, Wang N, Lin Y, Song R, Zhang M, Li B. Structure-guided design of endosomolytic chloroquine-like lipid nanoparticles for mRNA delivery and genome editing. Nat Commun 2025; 16:4241. [PMID: 40335474 PMCID: PMC12058976 DOI: 10.1038/s41467-025-59501-y] [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: 12/16/2024] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
Despite remarkable progress in designing RNA delivery systems, endosomal escape remains a recognized challenge for efficient RNA delivery. In this study, we develop a robust mRNA delivery platform termed endosomolytic chloroquine-like optimized lipid nanoparticles (ecoLNPs) for versatile mRNA delivery in vitro and in vivo via integrating the signature scaffold extracted from endosomolytic chloroquine into ionizable lipids. RNase-resistant ecoLNPs are capable of delivering a broad variety of mRNA payloads to diverse cell types, even hard-to-transfect 3D cells, with an efficiency of up to 18.9-fold higher than that of commercial transfection reagents. The pH-responsive endosomolytic activity of ecoLNPs can be largely attributed to the proton sponge effect and saposin B-promoted membrane disruption. In vivo, ecoLNPs enable potent local and systemic mRNA delivery and exhibit comparable potency to the clinically approved mRNA vaccine carrier, but strong tropism for lymph nodes following intramuscular injection. Furthermore, ecoLNPs are able to retain in vivo delivery potency for at least one week under non-frozen conditions and induce efficient genome editing in transgenic mice. Overall, the structure-guided integration strategy provides a pathway for de novo design of endosomolytic mRNA delivery systems.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Infectious Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Jiacai Wu
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Ning Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yongqi Lin
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Ruiteng Song
- Department of Infectious Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Min Zhang
- Department of Ophthalmology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Bin Li
- Department of Infectious Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China.
- School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
5
|
Shin JE, 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. [PMID: 40325908 DOI: 10.1021/acsami.5c06464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [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
|
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
|
Heiser BJ, Veyssi A, Ghosh D. Recent strategies for enhanced delivery of mRNA to the lungs. Nanomedicine (Lond) 2025; 20:1043-1069. [PMID: 40190037 PMCID: PMC12051540 DOI: 10.1080/17435889.2025.2485669] [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: 12/20/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
mRNA-based therapies have emerged as a transformative tool in modern medicine, gaining significant attention following their successful use in COVID-19 vaccines. Delivery to the lungs offers several compelling advantages for mRNA delivery. The lungs are one of the most vascularized organs in the body, which provides an extensive surface area that can facilitate efficient drug transport. Local delivery to the lungs bypasses gastrointestinal degradation, potentially enhancing therapeutic efficacy. In addition, the extensive capillary network of the lungs provides an ideal target for systemic delivery. However, developing effective mRNA therapies for the lungs presents significant challenges. The complex anatomy of the lungs and the body's immune response to foreign particles create barriers to delivery. This review discusses key approaches for overcoming these challenges and improving mRNA delivery to the lungs. It examines both local and systemic delivery strategies aimed at improving lung delivery while mitigating off-target effects. Although substantial progress has been made in lung-targeted mRNA therapies, challenges remain in optimizing cellular uptake and achieving therapeutic efficacy within pulmonary tissues. The continued refinement of delivery strategies that enhance lung-specific targeting while minimizing degradation is critical for the clinical success of mRNA-based pulmonary therapies.
Collapse
Affiliation(s)
- Brittany J. Heiser
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Arian Veyssi
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Debadyuti Ghosh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
8
|
Karmaker S, Rosales PD, Tirumuruhan B, Viravalli A, Boehnke N. More than a delivery system: the evolving role of lipid-based nanoparticles. NANOSCALE 2025. [PMID: 40293317 DOI: 10.1039/d4nr04508d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Lipid-based nanoparticles, including liposomes and lipid nanoparticles (LNPs), make up an important class of drug delivery systems. Their modularity enables encapsulation of a wide range of therapeutic cargoes, their ease of functionalization allows for incorporation of targeting motifs and anti-fouling coatings, and their scalability facilitates rapid translation to the clinic. While the discovery and early understanding of lipid-based nanoparticles is heavily rooted in biology, formulation development has largely focused on materials properties, such as how liposome and lipid nanoparticle composition can be altered to maximize drug loading, stability and circulation. To achieve targeted delivery and enable improved accumulation of therapeutics at target tissues or disease sites, emphasis is typically placed on the use of external modifications, such as peptide, protein, and polymer motifs. However, these approaches can increase the complexity of the nanocarrier and complicate scale up. In this review, we focus on how our understanding of lipid structure and function in biological contexts can be used to design intrinsically functional and targeted nanocarriers. We highlight formulation-based strategies, such as the incorporation of bioactive lipids, that have been used to modulate liposome and lipid nanoparticle properties and improve their functionality while retaining simple nanocarrier designs. We also highlight classes of naturally occurring lipids, their functions, and how they have been incorporated into lipid-based nanoparticles. We will additionally position these approaches into the historical context of both liposome and LNP development.
Collapse
Affiliation(s)
- Senjuti Karmaker
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Plinio D Rosales
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Barath Tirumuruhan
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Amartya Viravalli
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Natalie Boehnke
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| |
Collapse
|
9
|
Kumar G, Ardekani AM. Machine-Learning Framework to Predict the Performance of Lipid Nanoparticles for Nucleic Acid Delivery. ACS APPLIED BIO MATERIALS 2025. [PMID: 40267508 DOI: 10.1021/acsabm.4c01716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Lipid nanoparticles (LNPs) are highly effective carriers for gene therapies, including mRNA and siRNA delivery, due to their ability to transport nucleic acids across biological membranes, low cytotoxicity, improved pharmacokinetics, and scalability. A typical approach to formulate LNPs is to establish a quantitative structure-activity relationship (QSAR) between their compositions and in vitro/in vivo activities, which allows for the prediction of activity based on molecular structure. However, developing QSAR for LNPs can be challenging due to the complexity of multicomponent formulations, interactions with biological membranes, stability in physiological environments, and diverse physicochemical properties. To address these challenges, we developed a machine-learning (ML) framework to predict the activity and cell viability of LNPs for nucleic acid delivery. We curated data from 6454 LNP formulations reported across 21 independent studies and implemented 11 different molecular featurization techniques, ranging from descriptors and fingerprints to graph-based representations, alongside six ML algorithms for binary and multiclass classification. Using scaffold-based 5-fold cross-validation, our models achieved classification accuracies exceeding 90% for both activity and cell viability prediction tasks. Among all model-feature combinations, descriptor-based features combined with ensemble models such as balanced random forest and extra trees yielded the highest performance. Through SHAP-based feature attribution and interaction analysis, we identified key physicochemical properties and compositional features driving the LNP performance, highlighting the importance of synergistic effects among multiple molecular features. Furthermore, we developed a transfer-learning strategy to bridge in vitro-to-in vivo prediction gaps by incorporating base model predictions along with additional biological attributes, such as the particle size, polydispersity index, and ζ potential. Despite the smaller size and inherent class imbalance of the in vivo data set, the transfer-learning models demonstrated a promising predictive performance, with accuracies exceeding 82%. Our findings underscore the potential of interpretable ML frameworks to guide rational LNP design and provide a scalable approach to QSAR modeling in complex nanomedicine systems.
Collapse
Affiliation(s)
- Gaurav Kumar
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Arezoo M Ardekani
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
10
|
Lin L, Su K, Zhang X, Shi L, Yan X, Fu Q, Yao K, Siegwart DJ, Liu S. A Versatile Strategy to Transform Cationic Polymers for Efficient and Organ-Selective mRNA Delivery. Angew Chem Int Ed Engl 2025; 64:e202500306. [PMID: 39929776 DOI: 10.1002/anie.202500306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Indexed: 02/19/2025]
Abstract
The progress of mRNA therapeutics underscores the imperative demand for the development of targeted delivery systems. While cationic polymers hold promise as genetic vectors, their poor in vivo efficacy and numerous variants highlight the urgent need for a universal functionalization strategy to bolster their delivery capabilities. Here, we present a versatile strategy to transform low-cost commercial cationic polymers into phospholipidated and alkylated polymers (PAPs), enabling efficient and organ-selective mRNA delivery in vivo. This straightforward post-functionalization method can be readily broadened to a diverse array of existing cationic polymers, enhancing their cellular uptake, endosomal escape, and mRNA release functionalities. Consequently, PAPs facilitate up to 30,500-fold higher mRNA expression compared to their unmodified counterparts in vivo. Notably, the one-component PAPs enable spleen-specific mRNA delivery, with their vaccine application validated in a mouse melanoma model following intravenous administration. Better still, PAPs can synergize with different helper lipids to formulate four-component lipid nanoparticles (LNPs), achieving respective lung- and liver-specific mRNA delivery. Noteworthy is that these organ-selective mRNA delivery systems significantly outperform previous polymer and LNP benchmarks. This transformation strategy for cationic polymers represents a generalized methodology to give highly effective mRNA carriers, highlighting substantial potential for clinical translation of mRNA therapies with organ-targeting requirements.
Collapse
Affiliation(s)
- Lixin Lin
- Eye Center of the Second Affiliated Hospital of Zhejiang University School of Medicine College of Pharmaceutical Sciences, Liangzhu Laboratory, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Kexin Su
- Eye Center of the Second Affiliated Hospital of Zhejiang University School of Medicine College of Pharmaceutical Sciences, Liangzhu Laboratory, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Xinyue Zhang
- Eye Center of the Second Affiliated Hospital of Zhejiang University School of Medicine College of Pharmaceutical Sciences, Liangzhu Laboratory, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Lu Shi
- Eye Center of the Second Affiliated Hospital of Zhejiang University School of Medicine College of Pharmaceutical Sciences, Liangzhu Laboratory, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Xinxin Yan
- Eye Center of the Second Affiliated Hospital of Zhejiang University School of Medicine College of Pharmaceutical Sciences, Liangzhu Laboratory, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Qiuli Fu
- Eye Center of the Second Affiliated Hospital of Zhejiang University School of Medicine College of Pharmaceutical Sciences, Liangzhu Laboratory, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Ke Yao
- Eye Center of the Second Affiliated Hospital of Zhejiang University School of Medicine College of Pharmaceutical Sciences, Liangzhu Laboratory, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Daniel J Siegwart
- Department of Biomedical Engineering Department of Biochemistry Simmons Comprehensive Cancer Center Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shuai Liu
- Eye Center of the Second Affiliated Hospital of Zhejiang University School of Medicine College of Pharmaceutical Sciences, Liangzhu Laboratory, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
11
|
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
|
12
|
Tan Z, Zheng L, Bo Y, Kambar N, Wang H, Leal C. Click Lipid Nanoparticles for the Delivery of mRNA to Metabolically Labeled Cancer Cells. Biochemistry 2025; 64:1807-1816. [PMID: 40181500 DOI: 10.1021/acs.biochem.4c00699] [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: 04/05/2025]
Abstract
Lipid nanoparticle (LNP)-based mRNA delivery has a lot of potential in combating a wide range of diseases, but delivering mRNA to specific cell types continues to be challenging. Despite recent advances in organ and cell specificity, the majority of clinical LNP systems cannot fully release their payload to a targeted site. Incorporating active targeting moieties into LNPs is highly desired to expand nanomedicine applications. In this Letter, we developed LNPs that harness the power of bioorthogonal "click" azide-alkyne chemical reactions. We show that the plasma membranes of cancer cells can be labeled with azide groups by metabolic sugar labeling, and these azide groups can react with dibenzocyclooctyne (DBCO) on LNPs to achieve specific binding. To achieve this, we synthesized new and versatile lipids by functionalizing DBCO groups to phospholipids with or without a poly(ethylene glycol) (PEG) linker. The DBCO lipids were successfully formulated into DBCO-LNPs comprising other standard lipid compounds. When using these DBCO-LNPs to deliver mRNA to metabolically labeled cells, DBCO-LNPs showed a remarkable ability to preferentially deliver mRNA to azide-labeled cells. Removing PEG linkers from DBCO lipids enables better integration and retention in the LNP, and the higher the amount of DBCO lipid, the stronger the targeting effect. This work demonstrates that cell-specific targeting can be achieved utilizing azide-alkyne ″click″ chemistry and could inspire the development of the next generation of LNPs for active cyto-tropic nanomedicines.
Collapse
Affiliation(s)
- Zhengzhong Tan
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Lining Zheng
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yang Bo
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Nurila Kambar
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hua Wang
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Cecilia Leal
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
13
|
Sung Y, Choi Y, Kim ES, Ryu JH, Kwon IC. Receptor-ligand interactions for optimized endocytosis in targeted therapies. J Control Release 2025; 380:524-538. [PMID: 39875075 DOI: 10.1016/j.jconrel.2025.01.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
Receptor-mediated endocytosis plays a crucial role in the success of numerous therapies and remains central to advancing drug development. This process begins with ligand binding to specific receptors, triggering the internalization and intracellular trafficking of receptor-ligand complexes. These complexes are subsequently directed into distinct routes, either toward lysosomal degradation or recycling to the cell surface, with implications for therapeutic outcomes. This review examines receptor-ligand interactions as key modulators of endocytosis, emphasizing their role in shaping therapeutic design and efficacy. Advances in selecting receptor-ligand pairs and engineering ligands with optimized properties have enabled precise control over internalization, endosomal sorting, and trafficking, providing tailored solutions for diverse therapeutic applications. Leveraging these insights, strategies such as RNA-based therapies, antibody-drug conjugates (ADCs), and targeted protein degradation (TPD) platforms have been refined to selectively avoid or promote lysosomal degradation, thereby enhancing therapeutic efficacy. By bridging fundamental mechanisms of receptor-mediated endocytosis with innovative therapeutic approaches, this review offers a framework for advancing precision medicine.
Collapse
Affiliation(s)
- Yejin Sung
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Youngjin Choi
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Eun Sun Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 20841, Republic of Korea
| | - Ju Hee Ryu
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| | - Ick Chan Kwon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
14
|
Heredero J, Peña Á, Broset E, Blandín B, de Miguel D, Alejo T, Toro A, Mata E, López-Gavín A, Gallego-Lleyda A, Casabona D, Lampaya V, Larraga A, Pérez-Herrán E, Luna D, Orera I, Romanos E, García A, Martínez-Oliván J, Giménez-Warren J. Predictive Lung- and Spleen-Targeted mRNA Delivery with Biodegradable Ionizable Lipids in Four-Component LNPs. Pharmaceutics 2025; 17:459. [PMID: 40284454 PMCID: PMC12030499 DOI: 10.3390/pharmaceutics17040459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Lipid nanoparticles (LNPs) are leading mRNA delivery vehicles, with ionizable lipids (ILs) as their key component. However, the relationship between the IL structure and LNP endogenous organ-targeting is not well understood. In this study, we developed a novel library of biodegradable ILs featuring beta-propionate linkers, which, when incorporated into a four-component LNP formulation, show excellent extrahepatic selectivity and high protein expression. Methods: We explored the impact of structural modifications in the hydrophobic chains and polar-head groups in the ILs while keeping the linkers unchanged. In vivo results were evaluated to examine how structural changes influence the biodistribution to spleen or lungs. LNP formulations were assessed for their protein expression levels and organ-specific targeting. Additionally, protein corona formation by the best-performing LNPs was examined to provide further mechanistic insights. Results: Organ targeting was significantly influenced by structural changes in the ILs, allowing for precise control of the biodistribution between the spleen and lungs. Branched hydrophobic chains demonstrated a higher propensity for spleen targeting, while modifications in the polar-head group could drastically shift biodistribution from the lung to the spleen. This led to the identification of LNPs' zeta potential as a key determinant of their extrahepatic targeting properties. Notably, ionizable lipid A3T2C7, also known as CP-LC-1495, displayed strong lung selectivity (97%) and high protein expression in lung tissue (1.21 × 108 p/s). Similarly, several promising candidates for spleen-targeting LNPs displayed protein expression levels exceeding 1 × 107 p/s (selectivity >80%). Conclusions: This study elucidates the structure-function relationships of ILs in passive organ-specific mRNA delivery, highlighting how the fine-tuning of hydrophobic chains, polar-head groups, and surface charge (zeta potential) allows for the precise control of LNP endogenous biodistribution, a mechanism influenced by protein corona formation. These findings enable the rational design of targeted LNP systems, enhancing their therapeutic potential for specific organs, such as the spleen and lungs.
Collapse
Affiliation(s)
- Juan Heredero
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Álvaro Peña
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Esther Broset
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Beatriz Blandín
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Diego de Miguel
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Teresa Alejo
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Alfonso Toro
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Elena Mata
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Alexandre López-Gavín
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Ana Gallego-Lleyda
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Diego Casabona
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Verónica Lampaya
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Ana Larraga
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Esther Pérez-Herrán
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - David Luna
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
- Department of Theoretical Physics, Faculty of Science, University of Zaragoza, Pedro Cerbuna s/n, 50009 Zaragoza, Spain
- Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, 50018 Zaragoza, Spain
| | - Irene Orera
- Proteomics Research Core Facility, Aragon Health Sciences Institute (IACS), 50009 Zaragoza, Spain;
| | - Eduardo Romanos
- Medical Imaging and Phenotyping Core Facility, Aragon Health Sciences Institute (IACS), 50009 Zaragoza, Spain; (E.R.); (A.G.)
| | - Alba García
- Medical Imaging and Phenotyping Core Facility, Aragon Health Sciences Institute (IACS), 50009 Zaragoza, Spain; (E.R.); (A.G.)
| | - Juan Martínez-Oliván
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| | - Javier Giménez-Warren
- Certest Pharma, Certest Biotec S.L., San Mateo de Gállego, 50840 Zaragoza, Spain; (J.H.); (Á.P.); (E.B.); (B.B.); (D.d.M.); (T.A.); (A.T.); (E.M.); (A.L.-G.); (A.G.-L.); (D.C.); (V.L.); (A.L.); (E.P.-H.); (D.L.)
| |
Collapse
|
15
|
Ojansivu M, Barriga HMG, Holme MN, Morf S, Doutch JJ, Andaloussi SEL, Kjellman T, Johnsson M, Barauskas J, Stevens MM. Formulation and Characterization of Novel Ionizable and Cationic Lipid Nanoparticles for the Delivery of Splice-Switching Oligonucleotides. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2419538. [PMID: 40091434 PMCID: PMC12038542 DOI: 10.1002/adma.202419538] [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: 12/12/2024] [Revised: 02/24/2025] [Indexed: 03/19/2025]
Abstract
Despite increasing knowledge about the mechanistic aspects of lipid nanoparticles (LNPs) as oligonucleotide carriers, the structure-function relationship in LNPs has been generally overlooked. Understanding this correlation is critical in the rational design of LNPs. Here, a materials characterization approach is utilized, applying structural information from small-angle X-ray scattering experiments to design novel LNPs focusing on distinct lipid organizations with a minimal compositional variation. The lipid phase structures are characterized in these LNPs and their corresponding bulk lipid mixtures with small-angle scattering techniques, and the LNP-cell interactions in vitro with respect to cytotoxicity, hemolysis, cargo delivery, cell uptake, and lysosomal swelling. An LNP is identified that outperforms Onpattro lipid composition using lipid components and molar ratios which differ from the gold standard clinical LNPs. The base structure of these LNPs has an inverse micellar phase organization, whereas the LNPs with inverted hexagonal phases are not functional, suggesting that this phase formation may not be needed for LNP-mediated oligonucleotide delivery. The importance of stabilizer choice for the LNP function is demonstrated and super-resolution microscopy highlights the complexity of the delivery mechanisms, where lysosomal swelling for the majority of LNPs is observed. This study highlights the importance of advanced characterization for the rational design of LNPs to enable the study of structure-function relationships.
Collapse
Affiliation(s)
- Miina Ojansivu
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteHuddingeStockholm171 77Sweden
| | - Hanna M. G. Barriga
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteHuddingeStockholm171 77Sweden
- Present address:
Division of NanobiotechnologyDepartment of Protein ScienceSciLifeLab, KTH Royal Institute of TechnologySolnaSweden
| | - Margaret N. Holme
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteHuddingeStockholm171 77Sweden
| | - Stefanie Morf
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteHuddingeStockholm171 77Sweden
| | - James J. Doutch
- ISIS Neutron and Muon SourceRutherford Appleton LaboratoryHarwell CampusOxfordshireOX11 0QXUK
| | - Samir EL Andaloussi
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstituteHuddinge14152StockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm141 86Sweden
- Karolinska ATMP CenterKarolinska InstituteHuddinge14152StockholmSweden
| | | | | | | | - Molly M. Stevens
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteHuddingeStockholm171 77Sweden
- Department of Physiology, Anatomy and GeneticsDepartment of Engineering ScienceKavli Institute for Nanoscience DiscoveryUniversity of OxfordOxfordOX1 3QUUK
| |
Collapse
|
16
|
Ogawa K, Tagami T, Miyake S, Ozeki T. Choice of organic solvent affects function of mRNA-LNP; pyridine produces highly functional mRNA-LNP. Int J Pharm 2025; 673:125367. [PMID: 39971169 DOI: 10.1016/j.ijpharm.2025.125367] [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: 12/17/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Lipid nanoparticles (LNPs) are well-known nanocarriers for mRNA delivery. mRNA-encapsulated LNPs (mRNA-LNPs) are prepared by alcohol dilution (broadly defined as solvent dilution) method, in which mRNA dissolved in acidic buffer is mixed with lipid dissolved in an organic solvent. Ethanol is the most commonly used organic solvent for dissolving lipids during the preparation of mRNA-LNPs. However, no studies have systematically investigated the effects of organic solvents that dissolve lipids during the preparation of mRNA-LNPs on the properties and functions of mRNA-LNPs. In this study, we prepared mRNA-LNPs by using a series of organic solvents and evaluated their characteristics. After screening, we discovered that pyridine, an organic solvent, improved the quality of mRNA-LNPs and their function in vitro and in vivo. Pyridine was applied versatilely to some lipid-composition combinations generally used in the preparation of mRNA-LNPs and can also be adapted to microfluidic-based preparation. Furthermore, with appropriate purification, the amount of pyridine remaining in the final preparation of the mRNA-LNPs was extremely low and did not affect safety. Although further mechanism-based studies are required, we conclude that pyridine is a solvent that can be applied to the production of mRNA-LNPs as a pharmaceutical product.
Collapse
Affiliation(s)
- Koki Ogawa
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Tatsuaki Tagami
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Satoru Miyake
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Tetsuya Ozeki
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan.
| |
Collapse
|
17
|
Mo Y, Keszei AFA, Kothari S, Liu H, Pan A, Kim P, Bu J, Kamanzi A, Dai DL, Mazhab-Jafari MT, Chen J, Leslie S, Zheng G. Lipid-siRNA Organization Modulates the Intracellular Dynamics of Lipid Nanoparticles. J Am Chem Soc 2025; 147:10430-10445. [PMID: 40068204 PMCID: PMC11951082 DOI: 10.1021/jacs.4c18308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/27/2025]
Abstract
Lipid nanoparticles (LNPs) are widely used for delivering therapeutic nucleic acids, yet the relationship between their internal structure and intracellular behavior, particularly before RNA release, remains unclear. Here, we elucidate how lipid-siRNA organization within LNPs can modulate their intracellular delivery dynamics. We use cryo-electron microscopy and photochemical assays to reveal that increased siRNA loading can reduce helper lipids' distribution to the LNP surface, while siRNA consistently localizes near the surface. These alterations in lipid-siRNA organization affect LNP membrane fluidity, enhancing LNP fusion with cellular membranes and promoting cytosolic siRNA delivery, primarily via macropinocytosis. Using photosensitive lipids and live cell imaging, we demonstrate that lipid-siRNA organization regulates LNP responsiveness to external stimuli, significantly affecting siRNA endosomal escape efficiency upon light activation. We further confirm this observation using convex lens-induced confinement microscopy and single-particle imaging. Overall, our findings provide critical insights into how lipid-siRNA organization shapes LNP intracellular dynamics, offering rational design principles for optimizing LNP-based RNA therapeutics.
Collapse
Affiliation(s)
- Yulin Mo
- Institute
of Medical Science, University of Toronto, Toronto, Ontario M5G 1L7, Canada
- Princess
Margaret Cancer Center, University Health
Network, Toronto, Ontario M5G 1L7, Canada
| | - Alexander F. A. Keszei
- Princess
Margaret Cancer Center, University Health
Network, Toronto, Ontario M5G 1L7, Canada
| | - Shagun Kothari
- Michael
Smith Laboratories and Department of Physics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Heyi Liu
- Princess
Margaret Cancer Center, University Health
Network, Toronto, Ontario M5G 1L7, Canada
| | - Anni Pan
- Princess
Margaret Cancer Center, University Health
Network, Toronto, Ontario M5G 1L7, Canada
| | - Paige Kim
- Princess
Margaret Cancer Center, University Health
Network, Toronto, Ontario M5G 1L7, Canada
| | - Jiachuan Bu
- Princess
Margaret Cancer Center, University Health
Network, Toronto, Ontario M5G 1L7, Canada
| | - Albert Kamanzi
- Michael
Smith Laboratories and Department of Physics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - David L. Dai
- Princess
Margaret Cancer Center, University Health
Network, Toronto, Ontario M5G 1L7, Canada
- Department
of Medical Biophysics, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Mohammad T. Mazhab-Jafari
- Princess
Margaret Cancer Center, University Health
Network, Toronto, Ontario M5G 1L7, Canada
- Department
of Medical Biophysics, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Juan Chen
- Princess
Margaret Cancer Center, University Health
Network, Toronto, Ontario M5G 1L7, Canada
| | - Sabrina Leslie
- Michael
Smith Laboratories and Department of Physics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Gang Zheng
- Institute
of Medical Science, University of Toronto, Toronto, Ontario M5G 1L7, Canada
- Princess
Margaret Cancer Center, University Health
Network, Toronto, Ontario M5G 1L7, Canada
- Department
of Medical Biophysics, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
18
|
Li T, Wang Y, Chen X, Cui H, Zhang L, Liu J, Wang J, Wang X, Zhao Y, Chen Q, Wang J. Direct Cytosolic Delivery of siRNA Conjugates: A Paradigm in Antiangiogenic Therapy for Choroidal Neovascularization. ACS NANO 2025; 19:11249-11262. [PMID: 40072892 DOI: 10.1021/acsnano.4c18924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Small interfering RNA (siRNA) has garnered tremendous interest as a potential therapeutic tool because of its intriguing gene-silencing ability. Toward the success in the manufacture of siRNA therapeutics for the potential treatment of choroidal neovascularization (CNV), siRNA conjugated with dual functional units of membrane-penetrating heptafluoropropyl and age-related macular degeneration-targeting cyclic Arg-Gly-Asp (RGD) peptide was attempted for transcellular transportation into the cell interiors. Of note, cyclic RGD allowed selective affinities toward the angiogenic endothelial cells in the pathological CNV. Noteworthy is the functional heptafluoropropyl group, due to its tempting lipophobic and hydrophobic properties, stimulating energy-independent transcellular trafficking behaviors to the cytoplasm directly from the extracellular compartments, namely, the nonendocytotic pathway. The behaviors manage to avoid the well-acknowledged drawback of endolysosomal entrapment, which is deemed to be the critical threat to the biovulnerable genomic therapeutics, thereby contributing to potent gene knockdown at the affected cells. Aiming for treatment of CNV, the siRNA duo was schemed with appropriate chemistry-based modifications for the targeted knockdown of both angiogenic VEGF-A and VEGF-R2. Subsequent investigations verified the potent reduction of vascular leakage, and our proposed siRNA duo accomplished a significant reduction of 67.3% in the mean area of the CNV lesion. Bioinformatic analysis has unveiled a multitude of therapeutic benefits conferred by anti-VEGF therapy, extending beyond the mere inhibition of angiogenesis, including the regulated leukocyte transendothelial migration, retinol metabolism, and estrogen signaling pathways. Hence, our proposed chemistry represents an interesting siRNA conjugate strategy accomplishing direct intracellular transportation of macromolecular biological payloads to the cytosol. Hence, this proposed fluorination strategy should be highlighted to encourage the development of appropriate prodrug chemistry in pursuit of transcellular trafficking of membrane-impermissible and biovulnerable biotherapeutics.
Collapse
Affiliation(s)
- Tongqi Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| | - Yue Wang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
| | - Xiyi Chen
- School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, China
| | - Hongyan Cui
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Liuwei Zhang
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Jun Liu
- School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Jin Wang
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, No. 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Xiumei Wang
- School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Yan Zhao
- Department of Gastric Surgery, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
| | - Qixian Chen
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzheng 518120, China
| | - Jing Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| |
Collapse
|
19
|
Park JD, Shin HE, An YS, Jang HJ, Park J, Kim SN, Park CG, Park W. Advancing Natural Killer Cell Therapy: Genetic Engineering Strategies for Enhanced Cancer Immunotherapy. Ann Lab Med 2025; 45:146-159. [PMID: 39774132 PMCID: PMC11788708 DOI: 10.3343/alm.2024.0380] [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: 07/19/2024] [Revised: 10/06/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
Natural killer (NK) cells are pivotal innate immune system components that exhibit spontaneous cytolytic activity against abnormal cells, such as infected and tumor cells. NK cells have shown significant promise in adoptive cell therapy because of their favorable safety profiles and minimal toxicity in clinical settings. Despite their advantages, the therapeutic application of unmodified NK cells faces challenges, including limited in vivo persistence, particularly in the immunosuppressive tumor microenvironment. Recent advances in genetic engineering have enhanced the therapeutic potential of NK cells by addressing these limitations and improving their therapeutic efficacy. In this review, we have described various methodologies for the genetic modification of NK cells, including viral vectors, electroporation, and nanoparticle-based approaches. The ongoing research on nanomaterialbased approaches highlights their potential to overcome current limitations in NK cell therapy, paving the way for advanced cancer therapy and improved clinical outcomes. In this review, we also emphasize the potential of engineered NK cells in cancer immunotherapy and other clinical applications, highlighting the expanding scope of NK cell-based treatments and the critical role of innovative genetic engineering techniques.
Collapse
Affiliation(s)
- Joo Dong Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Ha Eun Shin
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Manoa, Honolulu, USA
| | - Yeon Su An
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Hye Jung Jang
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Manoa, Honolulu, USA
| | - Se-Na Kim
- Department of Industrial Cosmetic Science, Chungbuk National University, Cheongju, Korea
- Research and Development Center, MediArk Inc., Cheongju, Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, Institute for Cross-disciplinary Studies, Sungkyunkwan University, Suwon, Korea
- Department of Intelligent Precision Healthcare Convergence, Institute for Cross-disciplinary Studies, Sungkyunkwan University, Suwon, Korea
- Korea Institute of Science and Technology, Seoul, Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
- Korea Institute of Science and Technology, Seoul, Korea
- Department of MetaBioHealth, Institute for Cross-disciplinary Studies, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
20
|
Zhao R, Guo J, Liu Z, Zhang Y, Zuo J, Lv S, Li X, Yao W, Zhang X. Zwitterionic lipid nanoparticles for efficient siRNA delivery and hypercholesterolemia therapy with rational charge self-transformation. Theranostics 2025; 15:3693-3712. [PMID: 40093884 PMCID: PMC11905131 DOI: 10.7150/thno.111685] [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: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Rationale: Effective delivery of small interfering RNA (siRNA) remains a significant challenge in treating hypercholesterolemia due to biocompatibility, cellular uptake, and endosomal escape issues. Rational regulation of carrier surface charge contributes to efficient siRNA delivery in vivo. Methods: This study introduces zwitterionic lipid nanoparticles (ZwiLNPs) as a novel solution to these challenges. By leveraging the unique properties of zwitterionic polymers, we achieved robust siRNA encapsulation and targeted delivery. The design of ZwiLNPs facilitates charge self-transformation in response to physiological conditions, which enhances their biocompatibility and cellular uptake efficiency. Result: In vivo studies demonstrated significant liver-targeting capabilities of ZwiLNPs, with improved endosomal escape following cellular internalization. Comparative analyses confirmed that ZwiLNPs outperform conventional lipid nanoparticles in terms of both cellular uptake and endosomal release. Conclusion: These findings position ZwiLNPs as a promising platform for RNA interference therapies, particularly for hypercholesterolemia and other lipid-related disorders.
Collapse
Affiliation(s)
- Ruichen Zhao
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Jing Guo
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Ziqi Liu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Yusheng Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Jiamin Zuo
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Songzhang Lv
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Wenlong Yao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing, 100191, PR China
| | - Xin Zhang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
21
|
Luo C, Li Y, Liu H, He J, Yang X, Zhao E, Zi G, Liu L, Hong Y, Wang H, Li T, Yang Z, Wang R, Xu Y, Peng B. Intracellular trafficking of lipid nanoparticles is hindered by cholesterol. Int J Pharm 2025; 671:125240. [PMID: 39826779 DOI: 10.1016/j.ijpharm.2025.125240] [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: 10/29/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
The intracellular trafficking of lipid nanoparticles (LNPs) leading to endosomal escape is critical for delivery efficiency. How components of LNP affect its intracellular trafficking and delivery efficiency remains unknown. Here, we developed a highly sensitive LNP/nucleic acid tracking platform based on streptavidin-biotin-DNA complex and high throughput imaging. Naked nucleic acids were found to be retained in the endocytotic vesicles proportional to endocytosis activity. With the help of LNP, nucleic acids were transported along the endolysosomal pathway with N/P ratio as low as 2 amongst very weak nucleic acid and LNP interaction. As the N/P ratio increases (concomitant concentration increase of all lipids), the monophasic endocytosis of LNP-DNA demonstrated biphasic characteristics, as shown by accumulation of LNP-DNA trapped in early endosomes in the peripheral of cells. Through a series of specifically designed LNPs, we found increase in N/P ratio alone, i.e. increase of ionizable lipid content, had no effect on the formation of peripheral LNP-endosomes. Importantly, increase in cholesterol content, via dose or concentration increase, positively correlated with formation and aggregation of peripheral LNP-endosomes. Meanwhile, helper lipid such as DSPC alleviated the detrimental effect of cholesterol on aggregation of peripheral LNP-endosomes. The trapping of LNP-nucleic acids in peripheral early endosomes hindered their intracellular trafficking along the endolysosomal pathway, thus reducing their reach to releasing compartments and diminishing cargo delivery efficiency. Our results demonstrate that high cholesterol content hinders LNP intracellular trafficking, which is detrimental for intracellular delivery of cargo.
Collapse
Affiliation(s)
- Chengzhi Luo
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Yunfei Li
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Haidong Liu
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Jing He
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Xiaojuan Yang
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - E Zhao
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Guanghui Zi
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Li Liu
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Yujia Hong
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Hui Wang
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Ting Li
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Zhengyu Yang
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Rui Wang
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China
| | - Yuhong Xu
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China; Yunnan Key Laboratory of Screening and Research on Anti-pathogenic Plant Resources from Western Yunnan, Dali University, Xueren Road, Dali, Yunnan Province 671003, China.
| | - Baowei Peng
- College of Pharmacy, DaLi University, No. 2 Hongsheng Road, Dali, Yunnan Province 671003, China; Yunnan Key Laboratory of Screening and Research on Anti-pathogenic Plant Resources from Western Yunnan, Dali University, Xueren Road, Dali, Yunnan Province 671003, China; Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan Province 671003, China.
| |
Collapse
|
22
|
Ressnerova A, Heger Z, Pumera M. Translational nanorobotics breaking through biological membranes. Chem Soc Rev 2025; 54:1924-1956. [PMID: 39807638 DOI: 10.1039/d4cs00483c] [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: 01/16/2025]
Abstract
In the dynamic realm of translational nanorobotics, the endeavor to develop nanorobots carrying therapeutics in rational in vivo applications necessitates a profound understanding of the biological landscape of the human body and its complexity. Within this landscape, biological membranes stand as critical barriers to the successful delivery of therapeutic cargo to the target site. Their crossing is not only a challenge for nanorobotics but also a pivotal criterion for the clinical success of therapeutic-carrying nanorobots. Nevertheless, despite their urgency, strategies for membrane crossing in translational nanorobotics remain relatively underrepresented in the scientific literature, signaling an opportunity for further research and innovation. This review focuses on nanorobots with various propulsion mechanisms from chemical and physical to hybrid mechanisms, and it identifies and describes four essential biological membranes that represent the barriers needed to be crossed in the therapeutic journey of nanorobots in in vivo applications. First is the entry point into the blood stream, which is the skin or mucosa or intravenous injection; next is the exit from the bloodstream across the endothelium to the target site; further is the entry to the cell through the plasma membrane and, finally, the escape from the lysosome, which otherwise destroys the cargo. The review also discusses design challenges inherent in translating nanorobot technologies to real-world applications and provides a critical overview of documented membrane crossings. The aim is to underscore the need for further interdisciplinary collaborations between chemists, materials scientists and chemical biologists in this vital domain of translational nanorobotics that has the potential to revolutionize the field of precision medicine.
Collapse
Affiliation(s)
- Alzbeta Ressnerova
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00, Brno, Czech Republic.
- Research Group for Molecular Biology and Nanomedicine, Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic
| | - Zbynek Heger
- Research Group for Molecular Biology and Nanomedicine, Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic
- Center of Advanced Innovation Technologies, Faculty of Materials Science and Technology, VSB - Technical University of Ostrava, 17. Listopadu 2172/15, 70800 Ostrava, Czech Republic
| | - Martin Pumera
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00, Brno, Czech Republic.
- Advanced Nanorobots & Multiscale Robotics Laboratory, Faculty of Electrical Engineering and Computer Science, VSB - Technical University of Ostrava, 17. listopadu 2172/15, 70800 Ostrava, Czech Republic
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
- Department of Medical Research, China Medical University Hospital, China Medical University, No. 91 Hsueh-Shih Road, Taichung, Taiwan
| |
Collapse
|
23
|
Nong J, Gong X, Dang QM, Tiwari S, Patel M, Wu J, Hanna A, Park WJ, Atochina-Vasserman EN, Huang HT, Marcos-Contreras OA, Morris-Blanco KC, Miner JJ, Weissman D, Muzykantov VR, Gupta K, Issadore D, Myerson JW, Wang Z, Brenner JS. Multi-stage-mixing to control the supramolecular structure of lipid nanoparticles, thereby creating a core-then-shell arrangement that improves performance by orders of magnitude. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.12.623321. [PMID: 39605450 PMCID: PMC11601355 DOI: 10.1101/2024.11.12.623321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
As they became the dominant gene therapy platform, lipid nanoparticles (LNPs) experienced nearly all their innovation in varying the structure of individual molecules in LNPs. This ignored control of the spatial arrangement of molecules, which is suboptimal because supramolecular structure determines function in biology. To control LNPs' supramolecular structure, we introduce multi-stage-mixing (MSM) to successively add different molecules to LNPs. We first utilize MSM to create a core-then-shell (CTS) synthesis. CTS-LNPs display a clear core-shell structure, vastly lower frequency of LNPs containing no detectable mRNA, and improved mRNA-LNP expression. With DNA-loaded LNPs, which for decades lagged behind mRNA-LNPs due to low expression, CTS improved DNA-LNPs' protein expression by 2-3 orders of magnitude, bringing it within range of mRNA-LNPs. These results show that supramolecular arrangement is critical to LNP performance and can be controlled by mixing methodology. Further, MSM/CTS have finally made DNA-LNPs into a practical platform for long-term gene expression.
Collapse
|
24
|
Henke L, Ghorbani A, Mole SE. The use of nanocarriers in treating Batten disease: A systematic review. Int J Pharm 2025; 670:125094. [PMID: 39694161 DOI: 10.1016/j.ijpharm.2024.125094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/09/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
The neuronal ceroid lipofuscinoses, commonly known as Batten disease, are a group of lysosomal storage disorders affecting children. There is extensive central nervous system and retinal degeneration, resulting in seizures, vision loss and a progressive cognitive and motor decline. Enzyme replacement and gene therapies are being developed, and mRNA and oligonucleotide therapies are more recently being considered. Overcoming the challenges of the blood-brain barrier and blood-ocular barrier is crucial for effectively targeting the brain and eye, whatever the therapeutic approach. Nanoparticles and extracellular vesicles are small carriers that can encapsulate a cargo and pass through these cell barriers. They have been investigated as drug carriers for other pathologies and could be a promising treatment strategy for Batten disease. Their use in gene, enzyme, or mRNA replacement therapy of all lysosomal storage disorders, including Mucopolysaccharidoses, Niemann-Pick diseases, and Fabry disease, is investigated in this systematic review. Different nanocarriers can efficiently target the lysosome and cross the barriers into the brain and eyes. This supports continued exploration of nanocarriers as potential future treatment options for Batten disease.
Collapse
Affiliation(s)
- Larissa Henke
- Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Ali Ghorbani
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | - Sara E Mole
- Great Ormond Street Institute of Child Health, University College London, London WC1E 6BT, UK.
| |
Collapse
|
25
|
Tashima T. Non-Invasive Delivery of CRISPR/Cas9 Ribonucleoproteins (Cas9 RNPs) into Cells via Nanoparticles for Membrane Transport. Pharmaceutics 2025; 17:201. [PMID: 40006568 PMCID: PMC11859894 DOI: 10.3390/pharmaceutics17020201] [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: 01/09/2025] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
The clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) system is a promising biotechnology tool for genome editing. However, in living organisms, several pharmacokinetic challenges arise, including off-target side effects due to incorrect distribution, low bioavailability caused by membrane impermeability, and instability resulting from enzymatic degradation. Therefore, innovative delivery strategies must be developed to address these issues. Modified nanoparticles offer a potential solution for the non-invasive delivery of CRISPR/Cas9 ribonucleoproteins (Cas9 RNPs). Cas9 RNPs encapsulated in nanoparticles are protected from enzymatic degradation, similar to how microRNAs are shielded within exosomes. It is well-established that certain materials, including proteins, are expressed selectively in specific cell types. For example, the α-7 nicotinic receptor is expressed in endothelial and neuronal cells, while the αvβ3 integrin is expressed in cancer cells. These endogenous materials can facilitate receptor-mediated endocytosis or transcytosis. Nanoparticles encapsulating Cas9 RNPs and coated with ligands targeting such receptors may be internalized through receptor-mediated mechanisms. Once internalized, Cas9 RNPs could perform the desired gene editing in the nucleus after escaping the endosome through mechanisms such as the proton sponge effect or membrane fusion. In this review, I discuss the potential and advantages of delivering Cas9 RNP-encapsulated nanoparticles coated with ligands through receptor-mediated endocytosis or transcytosis.
Collapse
Affiliation(s)
- Toshihiko Tashima
- Tashima Laboratories of Arts and Sciences, 1239-5 Toriyama-cho, Kohoku-ku, Yokohama 222-0035, Japan
| |
Collapse
|
26
|
Bader J, Rüedi P, Mantella V, Geisshüsler S, Brigger F, Qureshi BM, Ortega Arroyo J, Montanari E, Leroux J. Loading of Extracellular Vesicles with Nucleic Acids via Hybridization with Non-Lamellar Liquid Crystalline Lipid Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404860. [PMID: 39741121 PMCID: PMC11848734 DOI: 10.1002/advs.202404860] [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: 05/06/2024] [Revised: 11/01/2024] [Indexed: 01/02/2025]
Abstract
The translation of cell-derived extracellular vesicles (EVs) into biogenic gene delivery systems is limited by relatively inefficient loading strategies. In this work, the loading of various nucleic acids into small EVs via their spontaneous hybridization with preloaded non-lamellar liquid crystalline lipid nanoparticles (LCNPs), forming hybrid EVs (HEVs) is described. It is demonstrated that LCNPs undergo pH-dependent structural transitions from inverse hexagonal (HII) phases at pH 5 to more disordered non-lamellar phases, possibly inverse micellar (L2) or sponge (L3) phases, at pH 7.4, which are particularly suitable for inducing a controlled hybridization process with EVs. State-of-the-art single-particle analysis techniques reveal that LCNPs interact with various EV subpopulations at physiological conditions and that ≈40% of HEVs are loaded with the genetic cargo. Importantly, this study demonstrates that EV membrane proteins remain accessible on HEV surfaces, with their intrinsic enzymatic activity unaffected after the hybridization process. Finally, HEVs show in vitro improved transfection efficiencies compared to unhybridized LCNPs. In summary, this versatile platform holds potential for loading various nucleic acid molecules into native EVs and may help developing EV-based therapeutics.
Collapse
Affiliation(s)
- Johannes Bader
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Pascal Rüedi
- Nanophotonic Systems LaboratoryDepartment of Mechanical and Process EngineeringETH ZurichZurich8092Switzerland
| | - Valeria Mantella
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Silvana Geisshüsler
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Finn Brigger
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Bilal Muhammad Qureshi
- Scientific Center for Optical and Electron Microscopy (ScopeM)ETH ZurichZurich8093Switzerland
| | - Jaime Ortega Arroyo
- Nanophotonic Systems LaboratoryDepartment of Mechanical and Process EngineeringETH ZurichZurich8092Switzerland
| | - Elita Montanari
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Jean‐Christophe Leroux
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| |
Collapse
|
27
|
Xu Z, Booth A, Rappolt M, Peckham M, Tyler AII, Beales PA. Topological and Morphological Membrane Dynamics in Giant Lipid Vesicles Driven by Monoolein Cubosomes. Angew Chem Int Ed Engl 2025; 64:e202414970. [PMID: 39348462 DOI: 10.1002/anie.202414970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/02/2024]
Abstract
Lipid nanoparticles have important applications as biomedical delivery platforms and broader engineering biology applications in artificial cell technologies. These emerging technologies often require changes in the shape and topology of biological or biomimetic membranes. Here we show that topologically-active lyotropic liquid crystal nanoparticles (LCNPs) can trigger such transformations in the membranes of giant unilamellar vesicles (GUVs). Monoolein (MO) LCNPs, cubosomes with an internal nanostructure of space groupI m 3 m ${Im3m}$ incorporate into 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) GUVs creating excess membrane area with stored curvature stress. Using time-resolved fluorescence confocal and lattice light sheet microscopy, we observe and characterise various life-like dynamic events in these GUVs, including growth, division, tubulation, membrane budding and fusion. Our results shed new light on the interactions of LCNPs with bilayer lipid membranes, providing insights relevant to how these nanoparticles might interact with cellular membranes during drug delivery and highlighting their potential as minimal triggers of topological transitions in artificial cells.
Collapse
Affiliation(s)
- Zexi Xu
- School of Food Science and Nutrition, University of Leeds, Leeds, LS2 9JT, United Kingdom
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Andrew Booth
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Michael Rappolt
- School of Food Science and Nutrition, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Michelle Peckham
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Arwen I I Tyler
- School of Food Science and Nutrition, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Paul A Beales
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| |
Collapse
|
28
|
Padilla MS, Shepherd SJ, Hanna AR, Kurnik M, Zhang X, Chen M, Byrnes J, Joseph RA, Yamagata HM, Ricciardi AS, Mrksich K, Issadore D, Gupta K, Mitchell MJ. Solution biophysics identifies lipid nanoparticle non-sphericity, polydispersity, and dependence on internal ordering for efficacious mRNA delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.19.629496. [PMID: 39763759 PMCID: PMC11702722 DOI: 10.1101/2024.12.19.629496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Lipid nanoparticles (LNPs) are the most advanced delivery system currently available for RNA therapeutics. Their development has accelerated since the success of Patisiran, the first siRNA-LNP therapeutic, and the mRNA vaccines that emerged during the COVID-19 pandemic. Designing LNPs with specific targeting, high potency, and minimal side effects is crucial for their successful clinical use. However, our understanding of how the composition and mixing method influences the structural, biophysical, and biological properties of the resulting LNPs remains limited, hindering the development of LNPs. Our lack of structural understanding extends from the physical and compositional polydispersity of LNPs, which render traditional characterization methods, such as dynamic light scattering (DLS), unable to accurately quantitate the physicochemical characteristics of LNPs. In this study, we address the challenge of structurally characterizing polydisperse LNP formulations using emerging solution-based biophysical methods that have higher resolution and provide biophysical data beyond size and polydispersity. These techniques include sedimentation velocity analytical ultracentrifugation (SV-AUC), field-flow fractionation followed by multi-angle light scattering (FFF-MALS), and size-exclusion chromatography in-line with synchrotron small-angle X-ray scattering (SEC-SAXS). Here, we show that the LNPs have intrinsic polydispersity in size, RNA loading, and shape, and that these parameters are dependent on both the formulation technique and lipid composition. Lastly, we demonstrate that these biophysical methods can be employed to predict transfection in human primary T cells, intravenous administration, and intramuscular administration by examining the relationship between mRNA translation and physicochemical characteristics. We envision that employing solution-based biophysical methods will be essential for determining LNP structure-function relationships, facilitating the creation of new design rules for LNPs.
Collapse
Affiliation(s)
- Marshall S. Padilla
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah J. Shepherd
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew R. Hanna
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Xujun Zhang
- Wyatt Technology, LLC, Goleta, CA 93117, USA
| | | | - James Byrnes
- National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, NY 11973, USA
| | - Ryann A. Joseph
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hannah M. Yamagata
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Adele S. Ricciardi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Surgery, University of Pennsylvania Health System, Philadelphia, PA 19104, USA
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kaitlin Mrksich
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Issadore
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kushol Gupta
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, 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
| |
Collapse
|
29
|
Torres C, Mancinelli G, Chen JWE, Cordoba-Chacon J, Pins D, Saeed S, McKinney R, Castellanos K, Orsi G, Singhal M, Patel A, Acebedo J, Coleman A, Heneche J, Yalagala PCR, Subbaiah PV, Leal C, Grimaldo S, Ortuno FM, Bishehsari F, Grippo PJ. Cell Membrane Fatty Acids and PIPs Modulate the Etiology of Pancreatic Cancer by Regulating AKT. Nutrients 2024; 17:150. [PMID: 39796583 PMCID: PMC11722924 DOI: 10.3390/nu17010150] [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: 10/24/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is one of the worst solid malignancies in regard to outcomes and metabolic dysfunction leading to cachexia. It is alarming that PDAC incidence rates continue to increase and warrant the need for innovative approaches to combat this disease. Due to its relatively slow progression (10-20 years), prevention strategies represent an effective means to improve outcomes. One of the risk factors for many cancers and for pancreatic cancer in particular is diet. Hence, our objective is to understand how a diet rich in ω3 and ω6 polyunsaturated fatty acids affects the progression of this disease. Methods: We investigated polyunsaturated fatty acid (PUFA) effects on disease progression employing both in vitro (PDAC cell lines) and in vivo (EL-Kras and KC mice) approaches. Also, we gathered data from the National Health and Nutrition Examination Survey (NHANES) and the National Cancer Institute (NCI) from 1999 to 2017 for a retrospective observational study. Results: The consumption of PUFAs in a patient population correlates with increased PDAC incidence, particularly when the ω3 intake increases to a lesser extent than ω6. Our data demonstrate dietary PUFAs can be incorporated into plasma membrane lipids affecting PI3K/AKT signaling and support the emergence of membrane-targeted therapies. Moreover, we show that the phospholipid composition of a lipid nanoparticle (LNP) can impact the cell membrane integrity and, ultimately, cell viability after administration of these LNPs. Conclusions: Cancer prevention is impactful particularly for those with very poor prognosis, including pancreatic cancer. Our results point to the importance of dietary intervention in this disease when detected early and the potential to improve the antiproliferative effect of drug efficacy when combined with these regimens in later stages of pancreatic cancer.
Collapse
Affiliation(s)
- Carolina Torres
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto de Investigacion Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Georgina Mancinelli
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Jee-Wei Emily Chen
- Department of Materials Science & Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; (J.-W.E.C.)
| | - Jose Cordoba-Chacon
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Danielle Pins
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Sara Saeed
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Ronald McKinney
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Karla Castellanos
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | | | - Megha Singhal
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Akshar Patel
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Jose Acebedo
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Adonis Coleman
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Jorge Heneche
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Poorna Chandra Rao Yalagala
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Papasani V. Subbaiah
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Cecilia Leal
- Department of Materials Science & Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; (J.-W.E.C.)
| | - Sam Grimaldo
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Francisco M. Ortuno
- Department of Computer Architecture and Computer Technology, University of Granada, 18071 Granada, Spain
| | - Faraz Bishehsari
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Paul J. Grippo
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois Chicago, 840 S. Wood Street, CSB 708, Chicago, IL 60612, USA
| |
Collapse
|
30
|
Deng S, Shao H, Shang H, Pang L, Chen X, Cao J, Wang Y, Zhao Z. Development of a Cationic Polymeric Micellar Structure with Endosomal Escape Capability Enables Enhanced Intramuscular Transfection of mRNA-LNPs. Vaccines (Basel) 2024; 13:25. [PMID: 39852804 PMCID: PMC11768556 DOI: 10.3390/vaccines13010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: The endosomal escape of lipid nanoparticles (LNPs) is crucial for efficient mRNA-based therapeutics. Here, we present a cationic polymeric micelle (cPM) as a safe and potent co-delivery system with enhanced endosomal escape capabilities. Methods: We synthesized a cationic and ampholytic di-block copolymer, poly (poly (ethylene glycol)4-5 methacrylatea-co-hexyl methacrylateb)X-b-poly(butyl methacrylatec-co-dimethylaminoethyl methacrylated-co-propyl acrylatee)Y (p(PEG4-5MAa-co-HMAb)X-b-p(BMAc-co-DMAEMAd-co-PAAe)Y), via reversible addition-fragmentation chain transfer polymerization. The cPMs were then formulated using the synthesized polymer by the dispersion-diffusion method and characterized by dynamic light scattering (DLS) and cryo-transmission electron microscopy (CryoTEM). The membrane-destabilization activity of the cPMs was evaluated by a hemolysis assay. We performed an in vivo functional assay of firefly luciferase (Fluc) mRNA using two of the most commonly studied LNPs, SM102 LNP and Dlin-MC3-DMA LNPs. Results: With a particle size of 61.31 ± 0.68 nm and a zeta potential of 37.76 ± 2.18 mV, the cPMs exhibited a 2-3 times higher firefly luciferase signal at the injection site compared to the control groups without cPMs following intramuscular injection in mice, indicating the high potential of cPMs to enhance the endosomal escape efficiency of mRNA-LNPs. Conclusions: The developed cPM, with enhanced endosomal escape capabilities, presents a promising strategy to improve the expression efficiency of delivered mRNAs. This approach offers a novel alternative strategy with no modifications to the inherent properties of mRNA-LNPs, preventing any unforeseeable changes in formulation characteristics. Consequently, this polymer-based nanomaterial holds immense potential for clinical applications in mRNA-based vaccines.
Collapse
Affiliation(s)
- Siyuan Deng
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| | - Han Shao
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| | - Hongtao Shang
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| | - Lingjin Pang
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| | - Xiaomeng Chen
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| | - Jingyi Cao
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
- NeoCura Bio-Medical Technology Co., Ltd., 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China
| | - Yi Wang
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
- NeoCura Bio-Medical Technology Co., Ltd., 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China
| | - Zhao Zhao
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| |
Collapse
|
31
|
Wu S, Yang Y, Lian X, Zhang F, Hu C, Tsien J, Chen Z, Sun Y, Vaidya A, Kim M, Sung YC, Xiao Y, Bian X, Wang X, Tian Z, Guerrero E, Robinson J, Basak P, Qin T, Siegwart DJ. Isosteric 3D Bicyclo[1.1.1]Pentane (BCP) Core-Based Lipids for mRNA Delivery and CRISPR/Cas Gene Editing. J Am Chem Soc 2024; 146:34733-34742. [PMID: 39655603 PMCID: PMC11717372 DOI: 10.1021/jacs.4c13154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Lipid nanoparticles (LNPs) are an essential component of messenger RNA (mRNA) vaccines and genome editing therapeutics. Ionizable amino lipids, which play the most crucial role in enabling mRNA to overcome delivery barriers, have, to date, been restricted to two-dimensional (2D) architectures. Inspired by improved physicochemical properties resulting from the incorporation of three-dimensionality (3D) into small-molecule drugs, we report the creation of 3D ionizable lipid designs through the introduction of bicyclo[1.1.1]pentane (BCP) core motifs. BCP-based lipids enabled efficient in vivo mRNA delivery to the liver and spleen with significantly greater performance over 2D benzene- and cyclohexane-based analogues. Notably, lead BCP-NC2-C12 LNPs mediated ∼90% reduction in the PCSK9 serum protein level via CRISPR/Cas9 gene knockout, outperforming 2D controls and clinically used DLin-MC3-DMA LNPs at the same dose. Here, we introduce BCP-based designs with superior in vivo activity, thereby expanding the chemical scope of ionizable amino lipids from 2D to 3D and offering a promising avenue to improve mRNA and gene editing efficiency for the continued development of genetic medicines.
Collapse
Affiliation(s)
- Shiying Wu
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Yangyang Yang
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Xizhen Lian
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Fangyu Zhang
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Chao Hu
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Jet Tsien
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Zexiang Chen
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Yehui Sun
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Amogh Vaidya
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Minjeong Kim
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Yun-Chieh Sung
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Yufen Xiao
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Xiaoyan Bian
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Xu Wang
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Zeru Tian
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Erick Guerrero
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Joshua Robinson
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Pratima Basak
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Tian Qin
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| | - Daniel J Siegwart
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390, United States
| |
Collapse
|
32
|
Haque MA, Shrestha A, Mikelis CM, Mattheolabakis G. Comprehensive analysis of lipid nanoparticle formulation and preparation for RNA delivery. Int J Pharm X 2024; 8:100283. [PMID: 39309631 PMCID: PMC11415597 DOI: 10.1016/j.ijpx.2024.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/21/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Nucleic acid-based therapeutics are a common approach that is increasingly popular for a wide spectrum of diseases. Lipid nanoparticles (LNPs) are promising delivery carriers that provide RNA stability, with strong transfection efficiency, favorable and tailorable pharmacokinetics, limited toxicity, and established translatability. In this review article, we describe the lipid-based delivery systems, focusing on lipid nanoparticles, the need of their use, provide a comprehensive analysis of each component, and highlight the advantages and disadvantages of the existing manufacturing processes. We further summarize the ongoing and completed clinical trials utilizing LNPs, indicating important aspects/questions worth of investigation, and analyze the future perspectives of this significant and promising therapeutic approach.
Collapse
Affiliation(s)
- Md. Anamul Haque
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Archana Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Constantinos M. Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| |
Collapse
|
33
|
Ma Y, Li S, Lin X, Chen Y. A perspective of lipid nanoparticles for RNA delivery. EXPLORATION (BEIJING, CHINA) 2024; 4:20230147. [PMID: 39713203 PMCID: PMC11655307 DOI: 10.1002/exp.20230147] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/07/2024] [Indexed: 12/24/2024]
Abstract
Over the last two decades, lipid nanoparticles (LNPs) have evolved as an effective biocompatible and biodegradable RNA delivery platform in the fields of nanomedicine, biotechnology, and drug delivery. They are novel bionanomaterials that can be used to encapsulate a wide range of biomolecules, such as mRNA, as demonstrated by the current successes of COVID-19 mRNA vaccines. Therefore, it is important to provide a perspective on LNPs for RNA delivery, which further offers useful guidance for researchers who want to work in the RNA-based LNP field. This perspective first summarizes the approaches for the preparation of LNPs, followed by the introduction of the key characterization parameters. Then, the in vitro cell experiments to study LNP performance, including cell selection, cell viability, cellular association/uptake, endosomal escape, and their efficacy, were summarized. Finally, the in vivo animal experiments in the aspects of animal selection, administration, dosing and safety, and their therapeutic efficacy were discussed. The authors hope this perspective can offer valuable guidance to researchers who enter the field of RNA-based LNPs and help them understand the crucial parameters that RNA-based LNPs demand.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Shiyao Li
- School of ScienceRMIT UniversityBundooraVictoriaAustralia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical EngineeringThe University of MelbourneParkvilleVictoriaAustralia
| | - Xin Lin
- Department of Cell BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Yupeng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
34
|
Patel S, Salaman SD, Kapoor DU, Yadav R, Sharma S. Latest developments in biomaterial interfaces and drug delivery: challenges, innovations, and future outlook. Z NATURFORSCH C 2024:znc-2024-0208. [PMID: 39566511 DOI: 10.1515/znc-2024-0208] [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: 09/25/2024] [Accepted: 11/03/2024] [Indexed: 11/22/2024]
Abstract
An ideal drug carrier system should demonstrate optimal payload and release characteristics, thereby ensuring prolonged therapeutic index while minimizing adverse effects. The field of drug delivery has undergone significant advancements, particularly within the last two decades, owing to the revolutionary impact of biomaterials. The use of biomaterials presents significant due to their biocompatibility and biodegradability, which must be addressed in order to achieve effective drug delivery. The properties of the biomaterial and its interface are primarily influenced by their physicochemical attributes, physiological barriers, cellular trafficking, and immunomodulatory effects. By attuning these barriers, regulating the physicochemical properties, and masking the immune system's response, the bio interface can be effectively modulated, leading to the development of innovative supramolecular structures with enhanced effectiveness. With a comprehensive understanding of these technologies, there is a growing demand for repurposing existing drugs for new therapeutic indications within this space. This review aims to provide a substantial body of evidence showcasing the productiveness of biomaterials and their interface in drug delivery, as well as methods for mitigating and modulating barriers and physicochemical properties along with an examination of future prospects in this field.
Collapse
Affiliation(s)
- Saraswati Patel
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, 602105, Tamil Nadu, India
| | - Samsi D Salaman
- Apollo College of Pharmacy, Mevaloorkuppam, Kanchipuram, 602105, Tamil Nadu, India
| | - Devesh U Kapoor
- Dr. Dayaram Patel Pharmacy College, Sardar Baug, Station Road, 394601 Bardoli, Gujarat, India
| | - Richa Yadav
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O., Rajasthan, 304022, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O., Rajasthan, 304022, India
| |
Collapse
|
35
|
Kim B, Subraveti SN, Liu JX, Nayagam SK, Merghoub S, Caggiano NJ, Amelemah DF, Jiang T, Bizmark N, Conway JM, Tsourkas A, Prud'homme RK. Diblock Copolymer Targeted Lipid Nanoparticles: Next-Generation Nucleic Acid Delivery System Produced by Confined Impinging Jet Mixers. ACS APPLIED BIO MATERIALS 2024; 7:7595-7607. [PMID: 39480746 DOI: 10.1021/acsabm.4c01176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Despite the recent advances and clinical demonstration of lipid nanoparticles (LNPs) for therapeutic and prophylactic applications, the extrahepatic delivery of nucleic acids remains a significant challenge in the field. This limitation arises from the rapid desorption of lipid-PEG in the bloodstream and clearance to the liver, which hinders extrahepatic delivery. In response, we explore the substitution of lipid-PEG with biodegradable block copolymers (BCPs), specifically poly(ε-caprolactone)-block-poly(ethylene glycol) (PCL-b-PEG). BCPs offer strong anchoring for large macromolecules, potentially enhancing cell-specific targeting. To develop and optimize BCP-stabilized LNPs (BCP-LNPs), we employed a Design of Experiment (DOE) approach. Through a systematic exploration, we identified optimal formulations for BCP-LNPs, achieving desirable physicochemical properties and encapsulation efficiency. Notably, BCP-LNPs exhibit surprising trends in transfection efficiency, with certain formulations showing up to a 40-fold increase in transfection in Hela cells, while maintaining minimal cytotoxicity. The lipid compositions that optimized PCL-b-PEG LNP transfection were different from the compositions that optimized PEG-lipid LNP transfection. Furthermore, our study confirms the versatility of BCP-LNPs in encapsulating and delivering both mRNA and pDNA, demonstrating their cargo-agnostic nature. Lastly, we showcased the targeted BCP-LNPs using a Cetuximab-conjugated formulation. These targeted LNPs show significant promise in delivering cargo specific to EGFR-overexpressing cells (A549 cells), with up to 2.4 times higher transfection compared to nontargeted LNPs. This finding underscores the potential of BCP-LNPs in targeted gene therapy, especially in challenging scenarios such as tumor targeting. Overall, our study establishes the viability of BCP-LNPs as a versatile, efficient, and targeted delivery platform for nucleic acids, opening avenues for advanced therapeutic applications.
Collapse
Affiliation(s)
- Bumjun Kim
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Sai Nikhil Subraveti
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Jason X Liu
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Satya K Nayagam
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Safaa Merghoub
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Nicholas J Caggiano
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - David F Amelemah
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Ting Jiang
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Navid Bizmark
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Jonathan M Conway
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Robert K Prud'homme
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
36
|
Ogawa K, Aikawa O, Tagami T, Ito T, Tahara K, Kawakami S, Ozeki T. Stable and inhalable powder formulation of mRNA-LNPs using pH-modified spray-freeze drying. Int J Pharm 2024; 665:124632. [PMID: 39182740 DOI: 10.1016/j.ijpharm.2024.124632] [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: 05/14/2024] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
A powder formulation for mucosal administration of mRNA-encapsulated lipid nanoparticles (mRNA-LNPs) is expected to be useful for respiratory diseases. Although freeze-drying is widely used to obtain solid formulations of mRNA-LNPs, highly hydrosoluble cryoprotectants, such as sucrose are necessary. However, sucrose is not a suitable excipient for inhalation powders because of its hygroscopic and deliquescence properties. Spray freeze-drying (SFD) is a method to produce inhalable powder formulation. In this study, we prepared inhalable powder formulations of mRNA-LNPs without deliquescence excipients using pH-modified SFD, which strengthens the interaction between mRNA and ionizable lipids of LNPs by acidic pH modifier, leading to retention of the encapsulated structure of mRNA-LNPs even after SFD. Powdered mRNA-LNPs were suitable for inhalation, and mRNA was encapsulated in LNPs after SFD. The mRNA encapsulation efficiency and mRNA transfection efficiency of pH-modified SFD-mediated powdered mRNA-LNPs were higher than those of conventional SFD, although they were significantly lower than those of liquid intact mRNA-LNPs. However, after long-term storage, the powdered formulation of the mRNA-LNPs exhibited higher mRNA transfection efficiency than liquid mRNA-LNP. Powdered mRNA-LNPs also exerted their function in air-liquid interface cultivation and in vivo intratracheal administration. Collectively, the powder formulation of mRNA-LNPs especially prepared by SFD is expected to be applied for dry powder inhalers.
Collapse
Affiliation(s)
- Koki Ogawa
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Otowa Aikawa
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Tatsuaki Tagami
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Takaaki Ito
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, 1-25-4 Daigaku-Nishi, Gifu 501-1196, Japan
| | - Kohei Tahara
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, 1-25-4 Daigaku-Nishi, Gifu 501-1196, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, Japan
| | - Tetsuya Ozeki
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan.
| |
Collapse
|
37
|
Milanesi L, Tomas S. The interaction of a self-assembled nanoparticle and a lipid membrane: Binding, disassembly and re-distribution. Heliyon 2024; 10:e39681. [PMID: 39524779 PMCID: PMC11550047 DOI: 10.1016/j.heliyon.2024.e39681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Here we report a detailed study of the interactions of nanoparticles, formed by the self-assembly of cholesterol-containing porphyrins, with lipid membranes. We show that the interaction is a two-step process: first, the docking and fusion, then, the redistribution of the building blocks of the self-assembled nanoparticles (SANs henceforth). Analysis of the binding and structural data is consistent with the docking step being driven by a multivalence cooperative effect and with the formation of SAN aggregates on the membrane, whilst the solubility of the cholesterol anchor in the membrane is key to both the fusion and redistribution of the SANs building blocks. The tendency of the SAN to aggregate in the membrane helps explain the photosensitizer properties of the SANs, essential to their anti-microbial activity. The solubility of the cholesteryl anchors drives fusion to the membrane and de-assembly of the SAN, explaining the capability of the SANs to deliver therapeutic cargos at the lipid interface. The subsequent redistribution of the SANs building blocks offer a plausible pathway to body clearance that is not immediately available to hard nanoparticles. These properties, and the modularity of the synthesis, point to the SANs being an excellent platform for the development of nanomedicines. An unexpected consequence of unraveling the mechanism of membrane interaction of these SANs is that it allows us to derive a value of the free energy of binding of cholesterol (the membrane anchor of the SAN building blocks) to a lipid membrane, that is consistent with the literature values. This is an additional property that can be exploited to determine the affinity of a variety of membrane anchors to membranes of various compositions.
Collapse
Affiliation(s)
- Lilia Milanesi
- Department of Chemistry, University of the Balearic Islands, Ctra. Valldemossa, Km 7.5. 07122, Palma de Mallorca, Spain
| | - Salvador Tomas
- Department of Chemistry, University of the Balearic Islands, Ctra. Valldemossa, Km 7.5. 07122, Palma de Mallorca, Spain
| |
Collapse
|
38
|
Garaizar A, Díaz-Oviedo D, Zablowsky N, Rissanen S, Köbberling J, Sun J, Steiger C, Steigemann P, Mann FA, Meier K. Toward understanding lipid reorganization in RNA lipid nanoparticles in acidic environments. Proc Natl Acad Sci U S A 2024; 121:e2404555121. [PMID: 39475644 PMCID: PMC11551392 DOI: 10.1073/pnas.2404555121] [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: 03/05/2024] [Accepted: 10/02/2024] [Indexed: 11/13/2024] Open
Abstract
The use of lipid nanoparticles (LNPs) for therapeutic RNA delivery has gained significant interest, particularly highlighted by recent milestones such as the approval of Onpattro and two mRNA-based SARS-CoV-2 vaccines. However, despite substantial advancements in this field, our understanding of the structure and internal organization of RNA-LNPs -and their relationship to efficacy, both in vitro and in vivo- remains limited. In this study, we present a coarse-grained molecular dynamics (MD) approach that allows for the simulations of full-size LNPs. By analyzing MD-derived structural characteristics in conjunction with cellular experiments, we investigate the effect of critical parameters, such as pH and composition, on LNP structure and potency. Additionally, we examine the mobility and chemical environment within LNPs at a molecular level. Our findings highlight the significant impact that LNP composition and internal molecular mobility can have on key stages of LNP-based intracellular RNA delivery.
Collapse
Affiliation(s)
- Adiran Garaizar
- Drug Discovery Sciences, Bayer Pharmaceuticals, Wuppertal42113, Germany
- Computational Life Science, Bayer Crop Science, Monheim am Rhein40789, Germany
| | - David Díaz-Oviedo
- Drug Discovery Sciences, Bayer Pharmaceuticals, Wuppertal42113, Germany
| | - Nina Zablowsky
- Lead Discovery, Nuvisan Innovation Campus Berlin, Berlin13353, Germany
| | - Sami Rissanen
- Chemical and Pharmaceutical Development, Bayer Pharmaceuticals, Turku20210, Finland
| | | | - Jiawei Sun
- Chemical and Pharmaceutical Development, Bayer Pharmaceuticals, Berlin13353, Germany
| | - Christoph Steiger
- Chemical and Pharmaceutical Development, Bayer Pharmaceuticals, Berlin13353, Germany
| | | | - Florian A. Mann
- Chemical and Pharmaceutical Development, Bayer Pharmaceuticals, Berlin13353, Germany
| | - Katharina Meier
- Drug Discovery Sciences, Bayer Pharmaceuticals, Wuppertal42113, Germany
| |
Collapse
|
39
|
Liu X, Min Q, Li Y, Chen S. Enhanced Cellular Immunity for Hepatitis B Virus Vaccine: A Novel Polyinosinic-Polycytidylic Acid-Incorporated Adjuvant Leveraging Cytoplasmic Retinoic Acid-Inducible Gene-Like Receptor Activation and Increased Antigen Uptake. Biomater Res 2024; 28:0096. [PMID: 39469105 PMCID: PMC11513446 DOI: 10.34133/bmr.0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
Conventional aluminum adjuvants exhibit limited cellular immunity. Polyinosinic-polycytidylic acid (poly I:C) activates cytoplasmic retinoic acid-inducible gene-like receptor (RLR), triggering strong T cell activation and cellular responses. However, when applied as an adjuvant, its limited endocytosis and restricted cytoplasmic delivery diminish its effectiveness and increase its toxicity. Hybrid polymer-lipid nanoparticle (PLNP) possesses numerous benefits such as good stability, reduced drug leakage, simple fabrication, easy property modulation, and excellent reproducibility compared to the lipid nanoparticle or the polymeric vector. Here, we developed a novel cationic polymer-lipid hybrid adjuvant capable of incorporating poly I:C to enhance cellular immunity. The hepatitis B surface antigen (HBsAg) was immobilized onto poly I:C-incorprated PLNP (PPLNP) via electrostatic interactions, forming the HBsAg/PPLNP vaccine formulation. The PPLNP adjuvant largely enhanced the cellular endocytosis and cytoplasmic transport of poly I:C, activating RLR followed by promoting type I interferon (IFN) secretion. Meanwhile, PPLNP obviously enhanced the antigen uptake, prolonged antigen retention at the site of administration, and facilitated enhanced transport of antigens to lymph nodes. The HBsAg/PPLNP nanovaccine led to amplified concentrations of antigen-specific immunoglobulin G (IgG), IFN-γ, granzyme B, and an enhanced IgG2a/IgG1 ratio, alongside the FasL+/CD8+ T cell activation, favoring a T helper 1 (TH1)-driven immune response. PPLNP, distinguished by its biocompatibility, ease of fabrication, and effectiveness in augmenting cellular immunity, holds significant promise as a new adjuvant.
Collapse
Affiliation(s)
- Xuhan Liu
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Institute for Inheritance-Based Innovation of Chinese Medicine, Marshall Laboratory of Biomedical Engineering, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Qiuxia Min
- Department of Pharmacy, First People’s Hospital of Yunnan Province,
Kunming University of Science and Technology, Kunming, 650034 Yunnan, China
| | - Yihui Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Siyuan Chen
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816 China
| |
Collapse
|
40
|
Wu S, Su K, Yan X, Shi L, Lin L, Ren E, Zhou J, Zhang C, Song Y, Liu S. Paracyclophane-based ionizable lipids for efficient mRNA delivery in vivo. J Control Release 2024; 376:395-401. [PMID: 39424104 DOI: 10.1016/j.jconrel.2024.10.028] [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: 08/22/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
mRNA therapeutics utilizing lipid nanoparticle (LNP) delivery technology represent a medical innovation for the treatment of various diseases. Amine-derived ionizable cationic lipids have been regarded as the pivotal component of LNPs, which often utilize commercially available small amine molecules as their cores. Given that even minor changes in the structure of ionizable lipids can result in significant differences in the delivery performance, there is a growing need to redesign the lipid amine-cores to optimize mRNA therapy. Here, we rationally design and synthesize a library of 198 paracyclophane-based ionizable lipids (PILs), which are then formulated into LNPs for mRNA delivery in vitro and in vivo. The resulting PIL LNPs display favorable characteristics, including appropriate particle sizes, zeta potentials, mRNA binding capability, efficacious endosomal escape, and robust mRNA delivery in vitro. Tailoring the PIL structures further enables mRNA expression specifically in the liver or simultaneously across multi-organs in vivo. Notably, the optimized PIL LNPs demonstrate superior efficacy compared to the U.S. Food and Drug Administration (FDA) approved DLin-MC3-DMA LNPs following intravenous administration. Additionally, when administered intramuscularly, our PIL LNPs exhibit higher efficacy than the SM-102 and ALC-0315 LNPs that are employed in the coronavirus disease 2019 (COVID-19) mRNA vaccines. These findings demonstrate the potential of paracyclophane-based ionizable lipids in advancing mRNA therapeutics, particularly for liver-targeted drugs and vaccines.
Collapse
Affiliation(s)
- Shiqi Wu
- College of Pharmaceutical Sciences, Liangzhu Laboratory, Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Kexin Su
- College of Pharmaceutical Sciences, Liangzhu Laboratory, Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinxin Yan
- College of Pharmaceutical Sciences, Liangzhu Laboratory, Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lu Shi
- College of Pharmaceutical Sciences, Liangzhu Laboratory, Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lixin Lin
- College of Pharmaceutical Sciences, Liangzhu Laboratory, Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - En Ren
- College of Pharmaceutical Sciences, Liangzhu Laboratory, Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jingjing Zhou
- Cosychem Technology (Tianjin) Co., Ltd., Tianjin 300450, China
| | - Chao Zhang
- Cosychem Technology (Tianjin) Co., Ltd., Tianjin 300450, China
| | - Yanmin Song
- Cosychem Technology (Tianjin) Co., Ltd., Tianjin 300450, China
| | - Shuai Liu
- College of Pharmaceutical Sciences, Liangzhu Laboratory, Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
41
|
Jeon T, Goswami R, Nagaraj H, Cicek YA, Lehot V, Welton J, Bell CJ, Park J, Luther DC, Im J, Rotello CM, Mager J, Rotello VM. Engineered zwitterionic diblock copolymer-siRNA polyplexes provide highly effective treatment of triple-negative breast cancer in a 4T1 murine model. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2406763. [PMID: 40017807 PMCID: PMC11864752 DOI: 10.1002/adfm.202406763] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Indexed: 03/01/2025]
Abstract
Self-assembly of siRNA with a block copolymer featuring guanidinium and zwitterion functionalized blocks generates core-shell-like nanovectors that provide cytosolic access to siRNA and efficiently evade phagocytic clearance. The guanidinium-functionalized inner block complexes siRNA in the nanovector interior and enables cytosolic delivery. The zwitterionic outer block provides a non-interacting shell on the nanovectors that reduces macrophage uptake in vitro and phagocytic clearance and enhances tumor localization in vivo. These nanovectors were used to treat a 4T1 (murine) model of triple-negative breast cancer (TNBC). The nanovectors deliver siRNA efficiently to 4T1 triple-negative breast cancer cells in vitro, with high selectivity relative to macrophages. This efficiency and selectivity translate into in vivo efficacy: diblock nanovectors evaded phagocytic clearance and efficiently localized in an aggressive murine 4T1 orthotopic model, with a ~3-fold increase of vector residing in the tumor compared to the homopolymer nanovectors. This increased localization efficiently knocked down STAT3 (~80%) and provided tumorostasis (100% growth inhibition) at a low dose of 0.14 mg/kg. The in vitro and in vivo efficacy of these nanovectors demonstrate the potential of engineered polymer architectures to generate effective self-assembled siRNA therapeutics that avoid phagocytic clearance for the treatment of diseases requiring systemic administration.
Collapse
Affiliation(s)
- Taewon Jeon
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, Massachusetts, 01003, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Yagiz Anil Cicek
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Victor Lehot
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Janelle Welton
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Charlotte J Bell
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Jungmi Park
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - David C Luther
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Jungkyun Im
- Department of Chemical Engineering, and Department of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, 22 Soonchunhyangro, Asan, 31538, Republic of Korea
| | - Caren M Rotello
- Department of Psychological and Brain Science, University of Massachusetts, Amherst, 135 Hicks Way, MA, 01003, USA
| | - Jesse Mager
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Vincent M Rotello
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, Massachusetts, 01003, USA
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
42
|
Wang J, Ding Y, Chong K, Cui M, Cao Z, Tang C, Tian Z, Hu Y, Zhao Y, Jiang S. Recent Advances in Lipid Nanoparticles and Their Safety Concerns for mRNA Delivery. Vaccines (Basel) 2024; 12:1148. [PMID: 39460315 PMCID: PMC11510967 DOI: 10.3390/vaccines12101148] [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: 09/13/2024] [Revised: 10/03/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
INTRODUCTION The advent of lipid nanoparticles (LNPs) as a delivery platform for mRNA therapeutics has revolutionized the biomedical field, particularly in treating infectious diseases, cancer, genetic disorders, and metabolic diseases. Recent Advances in Therapeutic LNPs: LNPs, composed of ionizable lipids, phospholipids, cholesterol, and polyethylene glycol (PEG) lipids, facilitate efficient cellular uptake and cytosolic release of mRNA while mitigating degradation by nucleases. However, as synthetic entities, LNPs face challenges that alter their therapeutic efficacy and safety concerns. Toxicity/Reactogenicity/Immunogenicity: This review provides a comprehensive overview of the latest advancements in LNP research, focusing on preclinical safety assessments encompassing toxicity, reactogenicity, and immunogenicity. Summary and Outlook: Additionally, it outlines potential strategies for addressing these challenges and offers insights into future research directions for enhancing the application of LNPs in mRNA therapeutics.
Collapse
Affiliation(s)
- Jialiang Wang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yaopeng Ding
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Kellie Chong
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; (K.C.)
| | - Meng Cui
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Zeyu Cao
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Chenjue Tang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Zhen Tian
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yuping Hu
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; (K.C.)
| | - Yu Zhao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Shaoyi Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
43
|
Serpico L, Zhu Y, Maia RF, Sumedha S, Shahbazi MA, Santos HA. Lipid nanoparticles-based RNA therapies for breast cancer treatment. Drug Deliv Transl Res 2024; 14:2823-2844. [PMID: 38831199 PMCID: PMC11384647 DOI: 10.1007/s13346-024-01638-2] [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] [Accepted: 05/21/2024] [Indexed: 06/05/2024]
Abstract
Breast cancer (BC) prevails as a major burden on global healthcare, being the most prevalent form of cancer among women. BC is a complex and heterogeneous disease, and current therapies, such as chemotherapy and radiotherapy, frequently fall short in providing effective solutions. These treatments fail to mitigate the risk of cancer recurrence and cause severe side effects that, in turn, compromise therapeutic responses in patients. Over the last decade, several strategies have been proposed to overcome these limitations. Among them, RNA-based technologies have demonstrated their potential across various clinical applications, notably in cancer therapy. However, RNA therapies are still limited by a series of critical issues like off-target effect and poor stability in circulation. Thus, novel approaches have been investigated to improve the targeting and bioavailability of RNA-based formulations to achieve an appropriate therapeutic outcome. Lipid nanoparticles (LNPs) have been largely proven to be an advantageous carrier for nucleic acids and RNA. This perspective explores the most recent advances on RNA-based technology with an emphasis on LNPs' utilization as effective nanocarriers in BC therapy and most recent progresses in their clinical applications.
Collapse
Affiliation(s)
- Luigia Serpico
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.
| | - Yuewen Zhu
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Renata Faria Maia
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Sumedha Sumedha
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
44
|
Lokras AG, Bobak TR, Baghel SS, Sebastiani F, Foged C. Advances in the design and delivery of RNA vaccines for infectious diseases. Adv Drug Deliv Rev 2024; 213:115419. [PMID: 39111358 DOI: 10.1016/j.addr.2024.115419] [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: 03/01/2024] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
RNA medicines represent a paradigm shift in treatment and prevention of critical diseases of global significance, e.g., infectious diseases. The highly successful messenger RNA (mRNA) vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were developed at record speed during the coronavirus disease 2019 pandemic. A consequence of this is exceptionally shortened vaccine development times, which in combination with adaptability makes the RNA vaccine technology highly attractive against infectious diseases and for pandemic preparedness. Here, we review state of the art in the design and delivery of RNA vaccines for infectious diseases based on different RNA modalities, including linear mRNA, self-amplifying RNA, trans-amplifying RNA, and circular RNA. We provide an overview of the clinical pipeline of RNA vaccines for infectious diseases, and present analytical procedures, which are paramount for characterizing quality attributes and guaranteeing their quality, and we discuss future perspectives for using RNA vaccines to combat pathogens beyond SARS-CoV-2.
Collapse
Affiliation(s)
- Abhijeet Girish Lokras
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Thomas Rønnemoes Bobak
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Saahil Sandeep Baghel
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Federica Sebastiani
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark; Division of Physical Chemistry, Department of Chemistry, Lund University, 22100, Lund, Sweden
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| |
Collapse
|
45
|
Heshmati N, Chakka LRJ, Zhang Y, Maniruzzaman M. Fabrication of mRNA encapsulated lipid nanoparticles using state of the art SMART-MaGIC technology and transfection in vitro. Sci Rep 2024; 14:22714. [PMID: 39349578 PMCID: PMC11442764 DOI: 10.1038/s41598-024-73804-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/20/2024] [Indexed: 10/02/2024] Open
Abstract
The messenger ribose nucleic acid (mRNA) in the form of Corona virus of 2019 (COVID-19) vaccines were effectively delivered through lipid nanoparticles (LNP) proving its use as effective carriers in clinical applications. In the present work, mRNA (erythropoietin (EPO)) encapsulated LNPs were prepared using a next generation state-of-the-art patented, Sprayed Multi Absorbed-droplet Reposing Technology (SMART) coupled with Multi-channeled and Guided Inner-Controlling printheads (MaGIC) technologies. The LNP-mRNA were synthesized at different N/P ratios and the particles were characterized for particle size and zeta potential (Zetasizer), encapsulation or complexation (gel retardation assay) and transfection (Fluorescence microscopy and ELISA) in MG63 sarcoma cells in vitro. The results showed a narrow distribution of mRNA-lipid particles of 200 nm when fabricated with SMART alone and then the size was reduced to approximately 50 nm with the combination of SMART-MaGIC technologies. The gel retardation assay showed that the N/P > 1 exhibited strong encapsulation of mRNA with lipid. The in vitro results showed the toxicity profile of the lipids where N/P ratio of 5 was optimized with > 50% cell viability. It can be concluded that the functional LNP-mRNA prepared and analyzed with SMART-MaGIC technologies, could be a potential new fabrication method of mRNA loaded LNPs for point-of-service or distributed manufacturing.
Collapse
Affiliation(s)
- Niloofar Heshmati
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Leela Raghava Jaidev Chakka
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Yu Zhang
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Mohammed Maniruzzaman
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA.
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University, MS, 38677, USA.
| |
Collapse
|
46
|
Aayush A, Darji S, Estes KM, Yeh E, Thompson DH. Development of an Elastin-like Polypeptide-Based Nucleic Acid Delivery System Targeted to EGFR+ Bladder Cancer Cells Using a Layer-by-Layer Approach. Biomacromolecules 2024; 25:5729-5744. [PMID: 39185801 PMCID: PMC11388462 DOI: 10.1021/acs.biomac.4c00165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/27/2024]
Abstract
Nucleic acid (NA)-based therapies are revolutionizing biomedical research through their ability to control cellular functions at the genetic level. This work demonstrates a versatile elastin-like polypeptide (ELP) carrier system using a layer-by-layer (LbL) formulation approach that delivers NA cargos ranging in size from siRNA to plasmids. The components of the system can be reconfigured to modulate the biochemical and biophysical characteristics of the carrier for engaging the unique features of the biological target. We show the physical characterization and biological performance of LbL ELP nucleic acid nanoparticles (LENNs) in murine and human bladder tumor cell lines. Targeting bladder tumors is difficult owing to the constant influx of urine into the bladder, leading to low contact times (typically <2 h) for therapeutic agents delivered via intravesical instillation. LENN complexes bind to bladder tumor cells within 30 min and become rapidly internalized to release their NA cargo within 60 min. Our data show that a readily adaptable NA-delivery system has been created that is flexible in its targeting ability, cargo size, and disassembly kinetics. This approach provides an alternative path to either lipid nanoparticle formulations that suffer from inefficiency and physicochemical instability or viral vectors that are plagued by manufacturing and immune rejection challenges. This agile ELP-based nanocarrier provides an alternative route for nucleic acid delivery using a biomanufacturable, biodegradable, biocompatible, and highly tunable vehicle capable of targeting cells via engagement with overexpressed cell surface receptors.
Collapse
Affiliation(s)
- Aayush Aayush
- Department of Chemistry &
Purdue Institute for Cancer Research, Purdue
University, Bindley Bioscience Center, West Lafayette, Indiana 47907, United States
| | - Saloni Darji
- Department of Chemistry &
Purdue Institute for Cancer Research, Purdue
University, Bindley Bioscience Center, West Lafayette, Indiana 47907, United States
| | - Kiera M. Estes
- Department of Chemistry &
Purdue Institute for Cancer Research, Purdue
University, Bindley Bioscience Center, West Lafayette, Indiana 47907, United States
| | - Emily Yeh
- Department of Chemistry &
Purdue Institute for Cancer Research, Purdue
University, Bindley Bioscience Center, West Lafayette, Indiana 47907, United States
| | - David H. Thompson
- Department of Chemistry &
Purdue Institute for Cancer Research, Purdue
University, Bindley Bioscience Center, West Lafayette, Indiana 47907, United States
| |
Collapse
|
47
|
Jang M, Yeom K, Han J, Fagan E, Park JH. Inhalable mRNA Nanoparticle with Enhanced Nebulization Stability and Pulmonary Microenvironment Infiltration. ACS NANO 2024; 18:24204-24218. [PMID: 39174871 DOI: 10.1021/acsnano.4c05653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
The delivery of mRNA into the lungs is the key to solving infectious and intractable diseases that frequently occur in the lungs. Since inhalation using a nebulizer is the most promising method for mRNA delivery into the lungs, there have been many attempts toward adapting lipid nanoparticles for mRNA inhalation. However, conventional lipid nanoparticles, which have shown great effectiveness for systemic delivery of mRNA and intramuscular vaccination, are not effective for pulmonary delivery due to their structural instability during nebulization and their inability to adapt to the pulmonary microenvironment. To address these issues, we developed an ionizable liposome-mRNA lipocomplex (iLPX). iLPX has a highly ordered lipid bilayer structure, which increases stability during nebulization, and its poly(ethylene glycol)-free composition allows it to infiltrate the low serum environment and the pulmonary surfactant layer in the lungs. We selected an inhalation-optimized iLPX (IH-iLPX) using a multistep screening procedure that mimics the pulmonary delivery process of inhaled nanoparticles. The IH-iLPX showed a higher transfection efficiency in the lungs compared to conventional lipid nanoparticles after inhalation with no observed toxicity in vivo. Furthermore, analysis of lung distribution revealed even protein expression in the deep lungs, with effective delivery to epithelial cells. This study provides insights into the challenges and solutions related to the development of inhaled mRNA pulmonary therapeutics.
Collapse
Affiliation(s)
- Mincheol Jang
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Kyunghwan Yeom
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Junhee Han
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Erinn Fagan
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
48
|
Alzahrani MS, Almutairy B, Althobaiti YS, Alsaab HO. Recent Advances in RNA Interference-Based Therapy for Hepatocellular Carcinoma: Emphasis on siRNA. Cell Biochem Biophys 2024; 82:1947-1964. [PMID: 38987439 DOI: 10.1007/s12013-024-01395-6] [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] [Accepted: 06/30/2024] [Indexed: 07/12/2024]
Abstract
Even though RNA treatments were first proposed as a way to change aberrant signaling in cancer, research in this field is currently ongoing. The term "RNAi" refers to the use of several RNAi technologies, including ribozymes, riboswitches, Aptamers, small interfering RNA (siRNA), antisense oligonucleotides (ASOs), and CRISPR/Cas9 technology. The siRNA therapy has already achieved a remarkable feat by revolutionizing the treatment arena of cancers. Unlike small molecules and antibodies, which need administration every three months or even every two years, RNAi may be given every quarter to attain therapeutic results. In order to overcome complex challenges, delivering siRNAs to the targeted tissues and cells effectively and safely and improving the effectiveness of siRNAs in terms of their action, stability, specificity, and potential adverse consequences are required. In this context, the three primary techniques of siRNA therapies for hepatocellular carcinoma (HCC) are accomplished for inhibiting angiogenesis, decreasing cell proliferation, and promoting apoptosis, are discussed in this review. We also deliberate targeting issues, immunogenic reactions to siRNA therapy, and the difficulties with their intrinsic chemistry and transportation.
Collapse
Affiliation(s)
- Mohammad S Alzahrani
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif21944, Saudi Arabia
| | - Bandar Almutairy
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Yusuf S Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif21944, Saudi Arabia
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif21944, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O. Box 11099, Taif21944, Saudi Arabia.
| |
Collapse
|
49
|
Eygeris Y, Henderson MI, Curtis AG, Jozic A, Stoddard J, Reynaga R, Chirco KR, Su GLN, Neuringer M, Lauer AK, Ryals RC, Sahay G. Preformed Vesicle Approach to LNP Manufacturing Enhances Retinal mRNA Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400815. [PMID: 38738752 PMCID: PMC11661498 DOI: 10.1002/smll.202400815] [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: 02/01/2024] [Revised: 04/20/2024] [Indexed: 05/14/2024]
Abstract
Complete encapsulation of nucleic acids by lipid-based nanoparticles (LNPs) is often thought to be one of the main prerequisites for successful nucleic acid delivery, as the lipid environment protects mRNA from degradation by external nucleases and assists in initiating delivery processes. However, delivery of mRNA via a preformed vesicle approach (PFV-LNPs) defies this precondition. Unlike traditional LNPs, PFV-LNPs are formed via a solvent-free mixing process, leading to a superficial mRNA localization. While demonstrating low encapsulation efficiency in the RiboGreen assay, PFV-LNPs improved delivery of mRNA to the retina by up to 50% compared to the LNP analogs across several benchmark formulations, suggesting the utility of this approach regardless of the lipid composition. Successful mRNA and gene editors' delivery is observed in the retinal pigment epithelium and photoreceptors and validated in mice, non-human primates, and human retinal organoids. Deploying PFV-LNPs in gene editing experiments result in a similar extent of gene editing compared to analogous LNP (up to 3% on genomic level) in the Ai9 reporter mouse model; but, remarkably, retinal tolerability is significantly improved for PFV-LNP treatment. The study findings indicate that the LNP formulation process can greatly influence mRNA transfection and gene editing outcomes, improving LNP treatment safety without sacrificing efficacy.
Collapse
Affiliation(s)
- Yulia Eygeris
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201 USA
| | - Michael I. Henderson
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201 USA
| | - Allison G. Curtis
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
| | - Antony Jozic
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201 USA
| | - Jonathan Stoddard
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Rene Reynaga
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Kathleen R. Chirco
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
| | - Grace Li-Na Su
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
| | - Martha Neuringer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Andreas K. Lauer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Renee C. Ryals
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201 USA
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon 97201
| |
Collapse
|
50
|
Fan Y, Rigas D, Kim LJ, Chang FP, Zang N, McKee K, Kemball CC, Yu Z, Winkler P, Su WC, Jessen P, Hura GL, Chen T, Koenig SG, Nagapudi K, Leung D, Yen CW. Physicochemical and structural insights into lyophilized mRNA-LNP from lyoprotectant and buffer screenings. J Control Release 2024; 373:727-737. [PMID: 39059500 DOI: 10.1016/j.jconrel.2024.07.052] [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: 06/06/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
The surge in RNA therapeutics has revolutionized treatments for infectious diseases like COVID-19 and shows the potential to expand into other therapeutic areas. However, the typical requirement for ultra-cold storage of mRNA-LNP formulations poses significant logistical challenges for global distribution. Lyophilization serves as a potential strategy to extend mRNA-LNP stability while eliminating the need for ultra-cold supply chain logistics. Although recent advancements have demonstrated the promise of lyophilization, the choice of lyoprotectant is predominately focused on sucrose, and there remains a gap in comprehensive evaluation and comparison of lyoprotectants and buffers. Here, we aim to systematically investigate the impact of a diverse range of excipients including oligosaccharides, polymers, amino acids, and various buffers, on the quality and performance of lyophilized mRNA-LNPs. From the screening of 45 mRNA-LNP formulations under various lyoprotectant and buffer conditions for lyophilization, we identified previously unexplored formulation compositions, e.g., polyvinylpyrrolidone (PVP) in Tris or acetate buffers, as promising alternatives to the commonly used oligosaccharides to maintain the physicochemical stability of lyophilized mRNA-LNPs. Further, we delved into how physicochemical and structural properties influence the functionality of lyophilized mRNA-LNPs. Leveraging high-throughput small-angle X-ray scattering (SAXS) and cryogenic transmission electron microscopy (cryo-TEM), we showed that there is complex interplay between mRNA-LNP structural features and cellular translation efficacy. We also assessed innate immune responses of the screened mRNA-LNPs in human peripheral blood mononuclear cells (PBMCs), and showed minimal alterations of cytokine secretion profiles induced by lyophilized formulations. Our results provide valuable insights into the structure-activity relationship of lyophilized formulations of mRNA-LNP therapeutics, paving the way for rational design of these formulations. This work creates a foundation for a comprehensive understanding of mRNA-LNP properties and in vitro performance change resulting from lyophilization.
Collapse
Affiliation(s)
- Yuchen Fan
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Diamanda Rigas
- Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Lee Joon Kim
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Lab, Berkeley, CA 94020, USA
| | - Feng-Peng Chang
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Nanzhi Zang
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kristina McKee
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Christopher C Kemball
- Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zhixin Yu
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Pascal Winkler
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Wan-Chih Su
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Pierce Jessen
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Greg L Hura
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Lab, Berkeley, CA 94020, USA; Chemistry and Biochemistry Department, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Tao Chen
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Stefan G Koenig
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Karthik Nagapudi
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Dennis Leung
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Chun-Wan Yen
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|