1
|
López Espinar A, Mulder LM, Elkhashab M, Khan Z, Czarnocki-Cieciura M, Aburto MR, Vucen S, Kowalski PS. Tailoring Alkyl Side Chains of Ionizable Amino-Polyesters for Enhanced In Vivo mRNA Delivery. ACS APPLIED BIO MATERIALS 2025; 8:3958-3971. [PMID: 40293247 DOI: 10.1021/acsabm.5c00116] [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] [Indexed: 04/30/2025]
Abstract
Lipid nanoparticles (LNPs) containing ionizable lipids are the most clinically advanced platform for mRNA delivery, but their application beyond the liver remains challenging. Polymer-lipid hybrid nanoparticles offer a promising alternative, combining the synthetic versatility and unique properties of polymers with the biocompatibility of lipid excipients. While the significance of alkyl tail design is well-recognized for ionizable lipids, the impact of the polymer side chain composition on interactions with lipid excipients, mRNA delivery efficacy, and tissue specificity remains poorly understood. Here, we focus on a class of ionizable amino-polyesters (APEs) that exhibit features desired for potential clinical applications, including narrow molecular weight distribution and a good safety profile, and investigate the effect of polymer side chain composition on the formulation of APE lipid nanoparticles (APE-LNPs) for mRNA delivery. A library of 36 APEs was synthesized via ring-opening polymerization of chemically diverse tertiary amino-alcohols and lactone monomers with distinct alkyl side chain compositions, including variations in length and unsaturation. We show that optimal alkyl side chain length is critical for the assembly of stable mRNA nanoparticles and efficient mRNA delivery both in vitro and in vivo. Top-performing APE-LNPs display superior delivery efficacy in vitro and in extrahepatic tissues compared to benchmark LNPs, including DLin-MC3-DMA ionizable lipid. The polymer chain composition affects the tissue selectivity of APE-LNPs, with shorter side chains (4-5 carbons) effectively targeting the spleen and lungs, while longer chains (7-9 carbons) show enhanced liver delivery. We also explored the relevance of lipid excipients in APE-LNPs, demonstrating the essential role of unsaturated phospholipids in enhancing cellular uptake and mRNA delivery, and the limited relevance of cholesterol. These findings provide valuable insights into the design of polymers for use in the LNP context, which could aid the development of polymeric alternatives to ionizable lipids and expand the utility of mRNA LNP technology to nonliver tissues.
Collapse
Affiliation(s)
| | - Lianne M Mulder
- School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | | | - Zahra Khan
- School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Mariusz Czarnocki-Cieciura
- Laboratory of Protein Structure, International Institute of Molecular and Cell Biology, Warsaw 02-109, Poland
| | - Maria R Aburto
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 K8AF, Ireland
| | - Sonja Vucen
- SSPC, Research Ireland Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Piotr S Kowalski
- School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
| |
Collapse
|
2
|
Kim J, Yang J, Heo S, Poo H. Evaluation of mRNA Transfection Reagents for mRNA Delivery and Vaccine Efficacy via Intramuscular Injection in Mice. ACS APPLIED BIO MATERIALS 2025; 8:4315-4324. [PMID: 40263125 DOI: 10.1021/acsabm.5c00424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The selection of an effective delivery carrier is crucial to assessing mRNA-based vaccines and therapeutics in vivo. Although lipid nanoparticles (LNPs) are commonly used for mRNA delivery, the LNP-mRNA formulation process is laborious and time-consuming and requires a high-cost microfluidic device. Instead, mixing with commercial reagents may simplify mRNA transfection into cells. However, their potential as in vivo carriers in intramuscular vaccination in mouse models remains unclear. In this study, we used three types of commercial RNA transfection reagents, MessengerMAX (MAX; liposome), TransIT-mRNA (IT; cationic polymer), and Invivofectamine (IVF; LNP), to produce nanoparticles directly by pipetting. The particle characteristics and mRNA delivery efficacy of the mRNA-transfection reagent mixtures were analyzed. Additionally, immune responses to vaccine efficacy and protective immunity of the mRNA mixtures as vaccine antigens were evaluated in a mouse model. Although MAX and IT showed high in vitro transfection efficiencies, their in vivo performances were limited. In contrast, IVF exhibited notable particle stability and homogeneity, making it a promising delivery carrier. Intramuscular IVF injection significantly enhanced both innate and adaptive immune responses with a robust systemic protein expression. Notably, when using SARS-CoV-2 Spike mRNA, IVF showed robust humoral immune responses, including production of IgG and neutralizing antibodies, thereby resulting in complete protection against SARS-CoV-2 infection. Therefore, these findings position IVF as an accessible and efficient mRNA carrier for evaluating mRNA vaccines and therapeutic efficacy in basic research.
Collapse
Affiliation(s)
- Jungho Kim
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Jihyun Yang
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Suhyeon Heo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Haryoung Poo
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
3
|
Kim JW, Kang TI, Choi E, Kim H. Reverse Block Sequence in Self-Immolative Poly(benzyl ether)-Based Amphiphiles for Tailoring End Groups and Self-Assembly Behavior. Biomacromolecules 2025; 26:2934-2944. [PMID: 40305787 DOI: 10.1021/acs.biomac.4c01845] [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: 05/02/2025]
Abstract
This paper reports a modular design of self-immolative poly(benzyl ether) (PBE) amphiphiles that allows precise control over polymer chain structure, end-group placement, and degradation behavior. By tuning block sequences and exposing reactive end groups, these amphiphiles undergo efficient head-to-tail depolymerization upon external stimuli. Structural variations in the monomers enable micelle formation with end groups displayed on the surface, while the carboxylate content in the hydrophilic block influences global micelle morphology. The resulting micelles are degradable in aqueous environments and can transform into spherical structures when combined with conventional surfactants. As a proof of concept, small-molecule cargos were successfully loaded and released from the mixed micelles on demand. This design platform offers a versatile route to create functional, stimulus-responsive surfactants with tunable assembly, degradation, and controlled release capabilities.
Collapse
Affiliation(s)
- Ji Woo Kim
- School of Polymer Science and Engineering, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - Tae-Il Kang
- Korea Institute of Medical Microrobotics (KIMIRo), 43-26, Cheomdangwagi-ro 208-beon-gil, Buk-gu, Gwangju 61011, Korea
| | - Eunpyo Choi
- Department of Mechanical Engineering, Sogang University, 35, Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Hyungwoo Kim
- School of Polymer Science and Engineering, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| |
Collapse
|
4
|
Bhati V, Prasad S, Kabra A. RNA-based therapies for neurodegenerative disease: Targeting molecular mechanisms for disease modification. Mol Cell Neurosci 2025; 133:104010. [PMID: 40340000 DOI: 10.1016/j.mcn.2025.104010] [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: 03/06/2025] [Revised: 04/23/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD) are characterized by progressive neuronal damage, protein aggregation, and chronic inflammation, leading to cognitive and motor impairments. Despite symptomatic relief from current therapies, disease-modifying treatments targeting the core molecular mechanism are still lacking. RNA-based therapies offer a promising approach to treating neurodegenerative disease by targeting molecular mechanisms such as gene expression, protein synthesis, and neuroinflammation. Therapeutic strategies include Long non-coding RNA (lncRNA), Antisense oligonucleotides (ASOs), RNA interference (RNAi), small interfering RNA (siRNA) and short hairpin RNA (shRNA), messenger RNA (mRNA) therapies, and microRNA (miRNA)-based interventions. These therapies aim to decrease toxic protein accumulation, restore deficient proteins, and modulate inflammatory responses in conditions like AD, PD, and HD. Unlike conventional treatments that primarily manage symptoms, RNA-based therapies have the potential to modify disease progression by addressing its root causes. This review aims to provide a comprehensive overview of current RNA-based therapeutic strategies for neurodegenerative diseases, discussing their mechanism of action, preclinical and clinical advancement. It further explores innovative solutions, including nanocarrier-mediated delivery, chemical modifications to enhance RNA stability, and personalized medicine approaches guided by genetic profiling that are being developed to overcome these barriers. This review also underscores the therapeutic opportunities and current limitations of RNA-based interventions, highlighting their potential to transform the future of neurodegenerative disease management.
Collapse
Affiliation(s)
- Vishal Bhati
- University Institute of Pharma Sciences, Chandigarh University, Mohali-140413, Punjab, India
| | - Sonima Prasad
- University Institute of Pharma Sciences, Chandigarh University, Mohali-140413, Punjab, India
| | - Atul Kabra
- University Institute of Pharma Sciences, Chandigarh University, Mohali-140413, Punjab, India.
| |
Collapse
|
5
|
Pan M, Cao W, Zhai J, Zheng C, Xu Y, Zhang P. mRNA-based vaccines and therapies - a revolutionary approach for conquering fast-spreading infections and other clinical applications: a review. Int J Biol Macromol 2025; 309:143134. [PMID: 40233916 DOI: 10.1016/j.ijbiomac.2025.143134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025]
Abstract
Since the beginning of the COVID-19 pandemic, the development of messenger RNA (mRNA) vaccines has made significant progress in the pharmaceutical industry. The two COVID-19 mRNA vaccines from Moderna and Pfizer/BioNTech have been approved for marketing and have made significant contributions to preventing the spread of SARS-CoV-2. In addition, mRNA therapy has brought hope to some diseases that do not have specific treatment methods or are difficult to treat, such as the Zika virus and influenza virus infections, as well as the prevention and treatment of tumors. With the rapid development of in vitro transcription (IVT) technology, delivery systems, and adjuvants, mRNA therapy has also been applied to hereditary diseases such as Fabry's disease. This article reviews the recent development of mRNA vaccines for structural modification, treatment and prevention of different diseases; delivery carriers and adjuvants; and routes of administration to promote the clinical application of mRNA therapies.
Collapse
Affiliation(s)
- Mingyue Pan
- Department of Pharmacy, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China
| | - Weiling Cao
- Department of Pharmacy, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Yingying Xu
- Department of Pharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Peng Zhang
- Department of Pharmacy, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China.
| |
Collapse
|
6
|
Vlasova KY, Kerr A, Pennock ND, Jozic A, Sahel DK, Gautam M, Murthy NTV, Roberts A, Ali MW, MacDonald KD, Walker JM, Luxenhofer R, Sahay G. Synthesis of ionizable lipopolymers using split-Ugi reaction for pulmonary delivery of various size RNAs and gene editing. Nat Commun 2025; 16:4021. [PMID: 40301362 PMCID: PMC12041268 DOI: 10.1038/s41467-025-59136-z] [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] [Accepted: 04/11/2025] [Indexed: 05/01/2025] Open
Abstract
We present an efficient method for synthesizing cationic poly(ethylene imine) derivatives using the multicomponent split-Ugi reaction to create a library of functional ionizable lipopolymers. Here we show 155 polymers, formulated into polyplexes, to establish structure-activity relationships essential for endosomal escape and transfection. A lead structure is identified, and lipopolymer-lipid hybrid nanoparticles are developed to deliver mRNA to lung endothelium and immune cells, including T cells, with low in vivo toxicity. These nanoparticles show significant improvements in mRNA delivery to the lung compared to in vivo-JetPEI® and demonstrate effective delivery of therapeutic mRNA(s) of various sizes. IL-12 mRNA-loaded nanoparticles delay Lewis Lung cancer progression, while human CFTR mRNA restores CFTR protein function in CFTR knockout mice. Additionally, we demonstrate in vivo CRISPR-Cas9 mRNA delivery, achieving gene editing in lung tissue and successful PD-1 knockout in T cells in mice. These results highlight the platform's potential for systemic gene therapy delivery.
Collapse
Affiliation(s)
- K Yu Vlasova
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR, USA
| | - A Kerr
- Soft Matter Chemistry, Department of Chemistry and Helsinki Institute of Sustainability Science, Faculty of Science, University of Helsinki, Helsinki, Finland
| | - N D Pennock
- Department of Radiation Medicine, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - A Jozic
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR, USA
| | - D K Sahel
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR, USA
| | - M Gautam
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR, USA
| | - N T V Murthy
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR, USA
| | - A Roberts
- Department of Pediatrics, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - M W Ali
- Soft Matter Chemistry, Department of Chemistry and Helsinki Institute of Sustainability Science, Faculty of Science, University of Helsinki, Helsinki, Finland
| | - K D MacDonald
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR, USA
- Department of Pediatrics, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - J M Walker
- Department of Radiation Medicine, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental, and Cancer Biology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - R Luxenhofer
- Soft Matter Chemistry, Department of Chemistry and Helsinki Institute of Sustainability Science, Faculty of Science, University of Helsinki, Helsinki, Finland.
| | - G Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR, USA.
- Center for Innovative Drug Delivery and Imaging, College of Pharmacy, Oregon State University & Oregon Health & Sciences University, Portland, OR, USA.
| |
Collapse
|
7
|
Panda S, Eaton EJ, Muralikrishnan P, Stelljes EM, Seelig D, Leyden MC, Gilkey AK, Barnes JT, Morrissey DV, Sarupria S, Moriarity BS, Reineke TM. Machine Learning Reveals Amine Type in Polymer Micelles Determines mRNA Binding, In Vitro, and In Vivo Performance for Lung-Selective Delivery. JACS AU 2025; 5:1845-1861. [PMID: 40313817 PMCID: PMC12041957 DOI: 10.1021/jacsau.5c00084] [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] [Received: 01/23/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 05/03/2025]
Abstract
Cationic micelles, composed of amphiphilic block copolymers with polycationic coronas, offer a customizable platform for mRNA delivery. Here, we present a library of 30 cationic micelle nanoparticles (MNPs) formulated from diblock copolymers with reactive poly(pentafluorophenol acrylate) backbones modified with diverse amines. This library systematically varies in nitrogen-based cationic functionalities, exhibiting a spectrum of properties that encompass varied degrees of alkyl substitution (A1-A5), piperazine (A6), oligoamine (A7), guanidinium (A8), and hydroxylation (A9-A10) that vary in side-chain volume, substitution pattern, hydrophilicity, and pK a to assess parameter impact on mRNA delivery. In vitro delivery assays using GFP+ mRNA across multiple cell lines reveal that amine side-chain bulk and chemical structure critically affect performance. Using machine learning analysis via SHapley Additive exPlanations (SHAP) on 180 formulations (3780 experimental measurements), we mapped key relationships between amine chemistry and performance metrics, finding that amine-specific binding efficiency was a major determinant of mRNA delivery efficacy, cell viability, and GFP intensity. Micelles with stronger mRNA binding capabilities (A1 and A7) have higher cellular delivery performance, whereas those with intermediate binding tendencies deliver a higher amount of functional mRNA per cell (A2, A10). This indicates that balancing the binding strength is crucial for performance. Micelles with hydrophobic and bulky pendant groups (A3-A5) tend to induce necrosis during cellular delivery, highlighting the significance of chemical optimization. A7 amphiphile, displaying primary and secondary amine, consistently demonstrates the highest GFP expression across various cell types and in vivo achieves high delivery specificity to lung tissue upon intravenous administration. Moreover, we established a strong correlation between in vitro and in vivo performance using Multitask Gaussian Process models, underscoring the predictive power of in vitro models for anticipating in vivo outcomes. Overall, this innovative study integrates advanced data science with experimental design, demonstrating the pivotal role of chemical amine-dependent optimization for advancing targeted mRNA delivery to the lungs.
Collapse
Affiliation(s)
- Sidharth Panda
- Department
of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ella J. Eaton
- Department
of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department
of Pediatrics, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Masonic
Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Center
for Genome Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Praveen Muralikrishnan
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Erin M. Stelljes
- Department
of Pediatrics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Davis Seelig
- Department
of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, Minnesota 55108, United States
| | - Michael C. Leyden
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Alexandria K. Gilkey
- Department
of Pediatrics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jackson T. Barnes
- Department
of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - David V. Morrissey
- Pfizer
Research
and Development, 610
Main Street, Cambridge, Massachusetts 02139, United States
| | - Sapna Sarupria
- Department
of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Branden S. Moriarity
- Department
of Pediatrics, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Masonic
Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Center
for Genome Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M. Reineke
- Department
of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
8
|
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
|
9
|
Roy P, Kreofsky NW, Santa Chalarca CF, Reineke TM. Binary Copolymer Blending Enhances pDNA Delivery Performance and Colloidal Shelf Stability of Quinine-Based Polyplexes. Bioconjug Chem 2025; 36:770-781. [PMID: 40067683 DOI: 10.1021/acs.bioconjchem.5c00040] [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: 04/17/2025]
Abstract
Successful gene therapies require the efficient delivery of the therapeutic nucleic acids in the target cells, which is a major bottleneck. Our group has demonstrated that quinine-based polymers are effective and promising carriers for delivering nucleic acids, such as plasmid DNA (pDNA). However, the inherent hydrophobicity of quinine-based polymers makes the polymer-pDNA complexes (polyplexes) colloidally unstable leading to aggregation, which is relevant in clinical scenarios as larger particles (diameter >1000 nm) tend to perform poorly when administered systemically in vivo. Herein, we overcome the hydrophobicity-induced aggregation by using two types of quinine-based polymer systems to form polyplexes via a facile blending approach. We balanced desirable properties using quinine-based copolymers (HQ-X) as the pDNA binding component along with a quinine-based diblock copolymer (PHQ), having a polyethylene glycol chain, to provide colloidal stability to the particles. Using 5 polymer pairs, 5 mixing ratios, and 3 mixing sequences, we screened 66 formulations out of which 37 resulted in nonaggregating small polyplexes (diameter <300 nm) with colloidal stability tested up to 7 days at 4 °C. Furthermore, 18 out of these 37 colloidally stable formulations showed transfection performance better than or comparable to the commercial control, jetPEI. Our results clearly indicated that while the three mixing sequences generate polyplexes of similar characteristics, the best balance of transfection efficiency, toxicity, and colloidal stability is achieved at moderate PHQ % in the mixture when colloidal stability does not compromise payload binding. Our results showcase that polymer blending, in a manner similar to lipids, is an effective and parallel approach to achieving desirable polyplex characteristics, such as particle size, colloidal stability, and performance.
Collapse
Affiliation(s)
- Punarbasu Roy
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Nicholas W Kreofsky
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
10
|
Lv J, Fan Q, Zhang Y, Zhou X, Yu P, Yu X, Xin C, Hong J, Cheng Y. A Serum Resistant Polymer with Exceptional Endosomal Escape and mRNA Delivery Efficacy for CRISPR Gene Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413006. [PMID: 39921871 PMCID: PMC11967772 DOI: 10.1002/advs.202413006] [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: 10/17/2024] [Revised: 12/16/2024] [Indexed: 02/10/2025]
Abstract
Nanoparticle-based mRNA delivery offers a versatile platform for innovative therapies. However, most of the current delivery systems are limited by poor serum tolerance, suboptimal endosomal escape and mRNA delivery efficacy. Herein, a highly efficient mRNA-delivering material is identified from a library of fluoropolymers. The lead material FD17 shows exceptional serum stability and endosomal escape, enabling efficient mRNA delivery into various cell types, surpassing commercial mRNA delivery reagents such as Lipofectamine 3000. The formed mRNA nanoparticles adsorb abundant serum albumin on the surface, which facilitates cellular uptake via scavenger receptor-mediated endocytosis. FD17 enables the delivery of mRNAs encoding CRE, Cas9, and base editor hyCBE for efficient genome editing. The material mediates CRISPR/Cas9 gene therapy via intraocular injection effectively down-regulates vascular endothelial growth factor A in retinal pigment epithelial cells of mice, yielding promising therapeutic responses against laser-induced choroidal neovascularization. The discovered material in this study shows great promise for the development of mRNA therapeutics to combat a wide range of diseases.
Collapse
Affiliation(s)
- Jia Lv
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal UniversityShanghai200241China
| | - Qianqian Fan
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal UniversityShanghai200241China
- Department of General SurgeryCenter for Metabolism ResearchThe Fourth Affiliated Hospital of Zhejiang University School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
| | - Yirou Zhang
- Department of Ophthalmology and Vision ScienceEye, Ear, Nose, and Throat HospitalFudan UniversityShanghai200030China
| | - Xujiao Zhou
- Department of Ophthalmology and Vision ScienceEye, Ear, Nose, and Throat HospitalFudan UniversityShanghai200030China
| | - Panting Yu
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal UniversityShanghai200241China
| | - Xin Yu
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal UniversityShanghai200241China
| | - Changchang Xin
- Department of Ophthalmology and Vision ScienceEye, Ear, Nose, and Throat HospitalFudan UniversityShanghai200030China
| | - Jiaxu Hong
- Department of Ophthalmology and Vision ScienceEye, Ear, Nose, and Throat HospitalFudan UniversityShanghai200030China
- Department of OphthalmologyEye & ENT HospitalState Key Laboratory of Molecular Engineering of PolymersFudan UniversityShanghai200031China
- NHC Key laboratory of Myopia and Related Eye DiseasesShanghai200031China
- Shanghai Engineering Research Center of Synthetic ImmunologyShanghai200032China
- Department of OphthalmologyChildren's Hospital of Fudan UniversityNational Pediatric Medical Center of ChinaShanghai200031China
| | - Yiyun Cheng
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal UniversityShanghai200241China
| |
Collapse
|
11
|
Chen Q, Chang Y, He X, Ding Y, Wang R, Luo R, Yuan J, Chen J, Zhong G, Yang H, Chen J, Li J. Targeted Delivery of mRNA with Polymer-Lipid Nanoparticles for In Vivo Base Editing. ACS NANO 2025; 19:7835-7850. [PMID: 39962883 DOI: 10.1021/acsnano.4c14041] [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: 03/05/2025]
Abstract
Messenger RNA (mRNA) encoding base editors, along with single guide RNAs (sgRNAs), have emerged as a promising therapeutic approach for various disorders. However, there is still insufficient exploration in achieving targeted and efficient delivery of mRNA and sgRNA to multiple organs while ensuring high biocompatibility and stability in vivo. To address this challenge, we synthesized a library of 108 poly(β-amino) esters (PBAEs) by incorporating 100% hydrophobic side chains and end-caps with varying amines. These PBAEs were further formulated with other excipients, including helper lipids, cholesterol, and PEGylated lipids, to form polymer-lipid nanoparticles (PLNPs). Structure-function analysis revealed that eLog P of PBAEs could serve as a predictive parameter for determining the liver or lung tropism of PLNPs. The biocompatibility of PBAEs end-capped with monoamines was significantly higher compared to those end-capped with diamines. Leveraging these findings, we expanded the PBAE library and identified a leading PBAE (7C8C8) with mRNA delivery efficiency outperforming current FDA-approved ionizable lipids (ALC-0315, SM-102, and Dlin-MC3-DMA). The LD50 of the empty PLNPs (7C8C8) was determined to be 403.8 ± 49.46 mg/kg, indicating a significantly high safety profile. Additionally, PLNPs (7C8C8) demonstrated sustained transfection activity for at least 2 months when stored at -20 °C after freezing or at 4 °C following lyophilization. Subsequently, in vivo base editing using PLNPs (7C8C8) achieved an impressive editing efficiency of approximately 70% along with a significant reduction in protein levels exceeding 90%. Notably, synergistic effects were observed through simultaneous disruption of proprotein convertase subtilisin/kexin type 9 and angiopoietin-like protein 3 genes, resulting in a sustained low-density lipoprotein cholesterol reduction of over 60% for several months. These compelling findings provide strong support for the further development of PLNPs as promising platforms for mRNA-based therapies.
Collapse
Affiliation(s)
- Qimingxing Chen
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yan Chang
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoyan He
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yan Ding
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Runyuan Wang
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Ran Luo
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Jialu Yuan
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Jiabei Chen
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Guisheng Zhong
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Huiying Yang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jia Chen
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jianfeng Li
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
12
|
Vosoughi P, Naghib SM, Kangarshahi BM, Mozafari MR. A review of RNA nanoparticles for drug/gene/protein delivery in advanced therapies: Current state and future prospects. Int J Biol Macromol 2025; 295:139532. [PMID: 39765293 DOI: 10.1016/j.ijbiomac.2025.139532] [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: 09/22/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Nanotechnology involves the utilization of materials with exceptional properties at the nanoscale. Over the past few years, nanotechnologies have demonstrated significant potential in improving human health, particularly in medical treatments. The self-assembly characteristic of RNA is a highly effective method for designing and constructing nanostructures using a combination of biological, chemical, and physical techniques from different fields. There is great potential for the application of RNA nanotechnology in therapeutics. This review explores various nano-based drug delivery systems and their unique features through the impressive progress of the RNA field and their significant therapeutic promises due to their unique performance in the COVID-19 pandemic. However, a significant hurdle in fully harnessing the power of RNA drugs lies in effectively delivering RNA to precise organs and tissues, a critical factor for achieving therapeutic effectiveness, minimizing side effects, and optimizing treatment outcomes. There have been many efforts to pursue targeting, but the clinical translation of RNA drugs has been hindered by the lack of clear guidelines and shared understanding. A comprehensive understanding of various principles is essential to develop vaccines using nucleic acids and nanomedicine successfully. These include mechanisms of immune responses, functions of nucleic acids, nanotechnology, and vaccinations. Regarding this matter, the aim of this review is to revisit the fundamental principles of the immune system's function, vaccination, nanotechnology, and drug delivery in relation to the creation and manufacturing of vaccines utilizing nanotechnology and nucleic acids. RNA drugs have demonstrated significant potential in treating a wide range of diseases in both clinical and preclinical research. One of the reasons is their capacity to regulate gene expression and manage protein production efficiently. Different methods, like modifying chemicals, connecting ligands, and utilizing nanotechnology, have been essential in enabling the effective use of RNA-based treatments in medical environments. The article reviews stimuli-responsive nanotechnologies for RNA delivery and their potential in RNA medicines. It emphasizes the notable benefits of these technologies in improving the effectiveness of RNA and targeting specific cells and organs. This review offers a comprehensive analysis of different RNA drugs and how they work to produce therapeutic benefits. Recent progress in using RNA-based drugs, especially mRNA treatments, has shown that targeted delivery methods work well in medical treatments.
Collapse
Affiliation(s)
- Pegah Vosoughi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran.
| | - Babak Mikaeeli Kangarshahi
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
13
|
Zhang M, Wang Y, Li B, Yang B, Zhao M, Li B, Liu J, Hu Y, Wu Z, Ong Y, Han X, Ding L, Zhu K, Li J, Luo M, Chen S, Peng L, Zhang L, Chen X, Ni Q. STING-Activating Polymers Boost Lymphatic Delivery of mRNA Vaccine to Potentiate Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2412654. [PMID: 39713955 DOI: 10.1002/adma.202412654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/02/2024] [Indexed: 12/24/2024]
Abstract
The unprecedented success of mRNA vaccines against COVID-19 has inspired scientists to develop mRNA vaccines for cancer immunotherapy. However, using nucleoside modified mRNA as vaccine, though evading innate immune toxicity, diminishes its therapeutic efficacy for cancers. Here, we report a polyvalent stimulator of interferon genes (STING) activating polymer (termed as PD) to bolster the immunogenicity of mRNA vaccine. PD is made of tertiary amine units and conjugated with a biodegradable alkyl chain. Co-formulation of PDs bearing different number of tertiary amines with lipid materials and mRNA resulted in the lipid-like nanoparticles (PD LNPs) which effectively promoted lymphatic delivery and elicited robust immune activation via the STING signaling pathway. Notably, PD with eighteen tertiary amines (PD18) is predominant in balancing immune activity and tolerability. Subcutaneous administration of PD18 LNPs containing ovalbumin (OVA) mRNA enhanced the frequency of antigen specific CD8+ T cell with immune memory, leading to potent anticancer efficacy that surpassed 2'3'-cGAMP in both prophylactic and therapeutic cancer models. Additionally, PD18 LNP-based mRNA vaccine showed conferred resistance to cancer challenge for up to 60 days. Overall, this study offers a new perspective of using STING- activating polymer for imparting synergistic activity in mRNA vaccine-based cancer immunotherapy.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yongling Wang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Pharmacy, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, P. R. China
| | - Benhao Li
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Bowei Yang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Mengyao Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Bingyu Li
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jianping Liu
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yaxin Hu
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Zhaoming Wu
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yenhui Ong
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xiaolin Han
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lingwen Ding
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Kongfu Zhu
- Department of Biological Science, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117558, Singapore
| | - Jianwei Li
- Department of Biological Science, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117558, Singapore
| | - Min Luo
- Department of Biological Science, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117558, Singapore
| | - Shengqi Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Ling Peng
- Centre Interdisciplinaire de Nanoscience de Marseille, Aix-Marseille Universite, CNRS, UMR 7325, ́ "Equipe Labellisee Ligue ́Contre le Cancer", Marseille, 13288, France
| | - Longjiang Zhang
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Chemical and Biomolecular Engineering, and Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore, 117544, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 138667, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Qianqian Ni
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
14
|
Shi Y, Mao J, Wang S, Ma S, Luo L, You J. Pharmaceutical strategies for optimized mRNA expression. Biomaterials 2025; 314:122853. [PMID: 39342919 DOI: 10.1016/j.biomaterials.2024.122853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Messenger RNA (mRNA)-based immunotherapies and protein in situ production therapies hold great promise for addressing theoretically all the diseases characterized by aberrant protein levels. The safe, stable, and precise delivery of mRNA to target cells via appropriate pharmaceutical strategies is a prerequisite for its optimal efficacy. In this review, we summarize the structural characteristics, mode of action, development prospects, and limitations of existing mRNA delivery systems from a pharmaceutical perspective, with an emphasis on the impacts from formulation adjustments and preparation techniques of non-viral vectors on mRNA stability, target site accumulation and transfection efficiency. In addition, we introduce strategies for synergistical combination of mRNA and small molecules to augment the potency or mitigate the adverse effects of mRNA therapeutics. Lastly, we delve into the challenges impeding the development of mRNA drugs while exploring promising avenues for future advancements.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Jiapeng Mao
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Siyao Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, 166 Qiutaobei Road, Hangzhou, Zhejiang, 310017, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China; The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China; Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, 321299, PR China.
| |
Collapse
|
15
|
Guo R, Li Z, Shi J, Yong H, Wang C, Li J, Zhang C, Zhou D. Hyaluronic acid coating of poly(β-amino ester)/mRNA polyplexes enables ultra-high transfection efficiency. J Control Release 2025; 378:428-437. [PMID: 39701456 DOI: 10.1016/j.jconrel.2024.12.038] [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: 05/31/2024] [Revised: 10/30/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
mRNA holds significant potential for a wide range of biomedical applications, efficient and safe delivery of mRNA into target cells is essential to realize its therapeutic benefits. Linear poly(β-amino ester)s (LPAEs) have been utilized for mRNA delivery, yet there is a need to enhance their transfection efficiency and safety profile. In this study, a novel LPAE-based ternary mRNA delivery system with ultra-high transfection efficiency is developed by coating hyaluronic acid (HA) onto LPAE/mRNA binary polyplexes. Results demonstrate that across a broad range of HA doses, mRNA transfection and cell viability can be significantly enhanced. Intriguingly, pre-treating cells with HA further boosts the transfection efficiency up to 99.2 %. Mechanistic studies reveal that HA coating impacts the size, Zeta potential of the binary polyplexes, enhancing their interaction with the cell membrane and facilitating cellular uptake. Leveraging the unique biocompatibility and biodegradability of HA, this ternary mRNA delivery system emerges as a promising option for practical applications. The approach of coating binary polyplexes with natural biomacromolecules can be extended to other non-viral mRNA delivery vectors to achieve superior transfection efficiency and safety profiles.
Collapse
Affiliation(s)
- Rui Guo
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhili Li
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jiahao Shi
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Haiyang Yong
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Chenfei Wang
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Jianzhong Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| | - Chaoting Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital and Institute, Beijing 100142, China.
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
16
|
Liao M, Yao Y, Gan K, Su X, Zhao N, Zuckermann RN, Xuan S, Zhang Z. Self-promoted Controlled Ring-opening Polymerization via Side Chain-mediated Proton Transfer for the Synthesis of Tertiary Amine-pendant Polypeptoids. Angew Chem Int Ed Engl 2025; 64:e202417990. [PMID: 39410820 DOI: 10.1002/anie.202417990] [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: 09/18/2024] [Indexed: 11/10/2024]
Abstract
Proton transfer is essential in virtually all biochemical processes, with enzymes facilitating this transfer by optimizing the proximity and orientation of reactants through site-specific hydrogen bonds. Proton transfer is also crucial in the rate-determining step for the ring-opening polymerization of N-carboxyanhydrides (NCAs), widely used to prepare various peptidomimetic materials. This study utilizes side chain-assisted strategy to accelerate the rate of chain propagation by using NCAs with tertiary amine pendants. This moiety enables hydrogen bond formation between the incoming NCA and the polymer amino growing end. The tertiary amine side chain of the NCA forms a proton shuttle, via a less constrained transition state, to facilitate the proton transfer process. Moreover, the tertiary amine side chains enable the precipitation of NCA monomers through in situ protonation during the monomer synthesis. This greatly facilitates the synthesis of these unreported monomers, allowing the direct controlled synthesis of tertiary amine-pendant polypeptoids. This side chain-promoted polymerization has rarely been reported. Additionally, the tertiary amine side chains, as widely used functional groups, endow the polymers with unique properties including pH- and thermo-responsiveness, tunable pKas, and siRNA transfection capability. The self-promoted synthesis, facile monomer preparation, and attractive properties make tertiary amine-pendant polypeptoids promising materials for various applications.
Collapse
Affiliation(s)
- Mingzhen Liao
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Materials, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Yao Yao
- Suzhou GenePharma Co., Ltd., Suzhou, 215123, China
| | - Kunyu Gan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Materials, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Xianghua Su
- Suzhou GenePharma Co., Ltd., Suzhou, 215123, China
| | - Ning Zhao
- Suzhou GenePharma Co., Ltd., Suzhou, 215123, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Ronald N Zuckermann
- The Molecular Foundry, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, California, 94720, United States
| | - Sunting Xuan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Materials, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Zhengbiao Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Materials, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| |
Collapse
|
17
|
Chen S, Pinto Carneiro S, Merkel OM. Anionic polymer coating for enhanced delivery of Cas9 mRNA and sgRNA nanoplexes. Biomater Sci 2025; 13:659-676. [PMID: 39687993 PMCID: PMC11650648 DOI: 10.1039/d4bm01290a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024]
Abstract
Polymeric carriers have long been recognized as some of the most effective and promising systems for nucleic acid delivery. In this study, we utilized an anionic di-block co-polymer, PEG-PLE, to enhance the performance of lipid-modified PEI (C14-PEI) nanoplexes for delivering Cas9 mRNA and sgRNA targeting KRAS G12S mutations in lung cancer cells. Our results demonstrated that PEG-PLE, when combined with C14-PEI at a weight-to-weight ratio of 0.2, produced nanoplexes with a size of approximately 140 nm, a polydispersity index (PDI) of 0.08, and a zeta potential of around -1 mV. The PEG-PLE/C14-PEI nanoplexes at this ratio were observed to be both non-cytotoxic and effective in encapsulating Cas9 mRNA and sgRNA. Confocal microscopy imaging revealed efficient endosomal escape and intracellular distribution of the RNAs. Uptake pathway inhibition studies indicated that the internalization of PEG-PLE/C14-PEI primarily involves scavenger receptors and clathrin-mediated endocytosis. Compared to C14-PEI formulations, PEG-PLE/C14-PEI demonstrated a significant increase in luciferase mRNA expression and gene editing efficiency, as confirmed by T7EI and ddPCR, in A549 cells. Sanger sequencing identified insertions and/or deletions around the PAM sequence, with a total of 69% indels observed. Post-transfection, the KRAS-ERK pathway was downregulated, resulting in significant increases in cell apoptosis and inhibition of cell migration. Taken together, this study reveals a new and promising formulation for CRISPR delivery as potential lung cancer treatment.
Collapse
Affiliation(s)
- Siyu Chen
- Ludwig-Maximilians-University, Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Butenandtstraße 5-13, Munich, 81377, Germany.
| | - Simone Pinto Carneiro
- Ludwig-Maximilians-University, Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Butenandtstraße 5-13, Munich, 81377, Germany.
| | - Olivia M Merkel
- Ludwig-Maximilians-University, Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Butenandtstraße 5-13, Munich, 81377, Germany.
| |
Collapse
|
18
|
Liao HC, Liu SJ. Advances in nucleic acid-based cancer vaccines. J Biomed Sci 2025; 32:10. [PMID: 39833784 PMCID: PMC11748563 DOI: 10.1186/s12929-024-01102-w] [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: 07/07/2024] [Accepted: 11/05/2024] [Indexed: 01/22/2025] Open
Abstract
Nucleic acid vaccines have emerged as crucial advancements in vaccine technology, particularly highlighted by the global response to the COVID-19 pandemic. The widespread administration of mRNA vaccines against COVID-19 to billions globally marks a significant milestone. Furthermore, the approval of an mRNA vaccine for Respiratory Syncytial Virus (RSV) this year underscores the versatility of this technology. In oncology, the combination of mRNA vaccine encoding neoantigens and immune checkpoint inhibitors (ICIs) has shown remarkable efficacy in eliciting protective responses against diseases like melanoma and pancreatic cancer. Although the use of a COVID-19 DNA vaccine has been limited to India, the inherent stability at room temperature and cost-effectiveness of DNA vaccines present a viable option that could benefit developing countries. These advantages may help DNA vaccines address some of the challenges associated with mRNA vaccines. Currently, several trials are exploring the use of DNA-encoded neoantigens in combination with ICIs across various cancer types. These studies highlight the promising role of nucleic acid-based vaccines as the next generation of immunotherapeutic agents in cancer treatment. This review will delve into the recent advancements and current developmental status of both mRNA and DNA-based cancer vaccines.
Collapse
Affiliation(s)
- Hung-Chun Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 307378, Taiwan.
| |
Collapse
|
19
|
Gao Y, Zhang L, Wang Z, Bai H, Wu C, Shuai Q, Yan Y. Enhanced Pulmonary and Splenic mRNA Delivery Using DOTAP-Incorporated Poly(β-Amino Ester)-Lipid Nanoparticles. Biomacromolecules 2025; 26:623-634. [PMID: 39746921 DOI: 10.1021/acs.biomac.4c01445] [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: 01/04/2025]
Abstract
mRNA-based therapies hold tremendous promise for treating various diseases, yet their clinical success is hindered by delivery challenges. This study developed a library of 140 lipocationic Poly(β-amino ester)s (PBAEs) and formulated lipid-polymer hybrid nanoparticles (LPHs) with four helper lipids, including 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), to enhance mRNA delivery. Initial in vitro screening of four representative PBAEs identified the D/P4-1 formulation (DOTAP/PBAE molar ratio of 4:1) as the most effective. Further screening of the library using this formulation identified eight top-performing LPHs. In vivo experiments confirmed high luciferase expression in the spleen and lungs of mice following intravenous administration of Luc mRNA-loaded LPHs. Detailed analysis revealed that DOTAP incorporation influenced LPH properties, including apparent pKa, surface charge, and internal hydrophobicity, enabling enhanced mRNA release and cellular uptake. This study demonstrates potent approaches to modulate PBAE-lipid nanoparticle properties by altering PBAE structures and nanoparticle composition, offering insights for designing effective hybrid carriers for mRNA therapeutics.
Collapse
Affiliation(s)
- Yuduo Gao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Luwei Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Ziyue Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Hao Bai
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Chengfan Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Qi Shuai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
20
|
Yu M, Lin L, Zhou D, Liu S. Interaction design in mRNA delivery systems. J Control Release 2025; 377:413-426. [PMID: 39580076 DOI: 10.1016/j.jconrel.2024.11.038] [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: 10/11/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024]
Abstract
Following the coronavirus disease 2019 (COVID-19) pandemic, mRNA technology has made significant breakthroughs, emerging as a potential universal platform for combating various diseases. To address the challenges associated with mRNA delivery, such as instability and limited delivery efficacy, continuous advancements in genetic engineering and nanotechnology have led to the exploration and refinement of various mRNA structural modifications and delivery platforms. These achievements have significantly broadened the clinical applications of mRNA therapies. Despite the progress, the understanding of the interactions in mRNA delivery systems remains limited. These interactions are complex and multi-dimensional, occurring between mRNA and vehicles as well as delivery materials and helper ingredients. Resultantly, stability of the mRNA delivery systems and their delivery efficiency can be both significantly affected. This review outlines the current state of mRNA delivery strategies and summarizes the interactions in mRNA delivery systems. The interactions include the electrostatic interactions, hydrophobic interactions, hydrogen bonding, π-π stacking, coordination interactions, and so on. This interaction understanding provides guideline for future design of next-generation mRNA delivery systems, thereby offering new perspectives and strategies for developing diverse mRNA therapeutics.
Collapse
Affiliation(s)
- Mengyao Yu
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Lixin Lin
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Shuai Liu
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
21
|
Xu L, Li C, Liao R, Xiao Q, Wang X, Zhao Z, Zhang W, Ding X, Cao Y, Cai L, Rosenecker J, Guan S, Tang J. From Sequence to System: Enhancing IVT mRNA Vaccine Effectiveness through Cutting-Edge Technologies. Mol Pharm 2025; 22:81-102. [PMID: 39601789 DOI: 10.1021/acs.molpharmaceut.4c00863] [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: 11/29/2024]
Abstract
The COVID-19 pandemic has spotlighted the potential of in vitro transcribed (IVT) mRNA vaccines with their demonstrated efficacy, safety, cost-effectiveness, and rapid manufacturing. Numerous IVT mRNA vaccines are now under clinical trials for a range of targets, including infectious diseases, cancers, and genetic disorders. Despite their promise, IVT mRNA vaccines face hurdles such as limited expression levels, nonspecific targeting beyond the liver, rapid degradation, and unintended immune activation. Overcoming these challenges is crucial to harnessing the full therapeutic potential of IVT mRNA vaccines for global health advancement. This review provides a comprehensive overview of the latest research progress and optimization strategies for IVT mRNA molecules and delivery systems, including the application of artificial intelligence (AI) models and deep learning techniques for IVT mRNA structure optimization and mRNA delivery formulation design. We also discuss recent development of the delivery platforms, such as lipid nanoparticles (LNPs), polymers, and exosomes, which aim to address challenges related to IVT mRNA protection, cellular uptake, and targeted delivery. Lastly, we offer insights into future directions for improving IVT mRNA vaccines, with the hope to spur further progress in IVT mRNA vaccine research and development.
Collapse
Affiliation(s)
- Lifeng Xu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Chao Li
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Rui Liao
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Qin Xiao
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Xiaoran Wang
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Xiaoyan Ding
- Department of Pediatrics, Ludwig-Maximilians University of Munich, Munich 80337, Germany
| | - Yuxue Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Larry Cai
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Joseph Rosenecker
- Department of Pediatrics, Ludwig-Maximilians University of Munich, Munich 80337, Germany
| | - Shan Guan
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Jie Tang
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| |
Collapse
|
22
|
Wang H, Cheng Y. Polymers for mRNA Delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70002. [PMID: 39763235 DOI: 10.1002/wnan.70002] [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: 05/30/2024] [Revised: 10/22/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
mRNA delivery has emerged as a transformative approach in biotechnology and medicine, offering a versatile platform for the development of novel therapeutics. Unlike traditional small molecule drugs or protein-based biologics, mRNA therapeutics have the unique ability to direct cells to generate therapeutic proteins, allowing for precise modulation of biological processes. The delivery of mRNA into target cells is a critical step in realizing the therapeutic potential of this technology. In this review, our focus is on the latest advancements in designing functional polymers to achieve efficient mRNA delivery. Biodegradable polymers and low molecular weight polymers in addressing the balance in mRNA binding and release are summarized. Benefiting from the excellent performance of lipid nanoparticles in mRNA delivery, polymer/lipid hybrid nanostructures are also included. Finally, the challenges and future prospects in the development of polymer-based mRNA delivery systems are discussed.
Collapse
Affiliation(s)
- Hui Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, China
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
23
|
Ariga K. Materials Nanoarchitectonics for Advanced Devices. MATERIALS (BASEL, SWITZERLAND) 2024; 17:5918. [PMID: 39685353 DOI: 10.3390/ma17235918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024]
Abstract
Advances in nanotechnology have made it possible to observe and evaluate structures down to the atomic and molecular level. The next step in the development of functional materials is to apply the knowledge of nanotechnology to materials sciences. This is the role of nanoarchitectonics, which is a concept of post-nanotechnology. Nanoarchitectonics is defined as a methodology to create functional materials using nanounits such as atoms, molecules, and nanomaterials as building blocks. Nanoarchitectonics is very general and is not limited to materials or applications, and thus nanoarchitecture is applied in many fields. In particular, in the evolution from nanotechnology to nanoarchitecture, it is useful to consider the contribution of nanoarchitecture in device applications. There may be a solution to the widely recognized problem of integrating top-down and bottom-up approaches in the design of functional systems. With this in mind, this review discusses examples of nanoarchitectonics in developments of advanced devices. Some recent examples are introduced through broadly dividing them into organic molecular nanoarchitectonics and inorganic materials nanoarchitectonics. Examples of organic molecular nanoarchitecture include a variety of control structural elements, such as π-conjugated structures, chemical structures of complex ligands, steric hindrance effects, molecular stacking, isomerization and color changes due to external stimuli, selective control of redox reactions, and doping control of organic semiconductors by electron transfer reactions. Supramolecular chemical processes such as association and intercalation of organic molecules are also important in controlling device properties. The nanoarchitectonics of inorganic materials often allows for control of size, dimension, and shape, and their associated physical properties can also be controlled. In addition, there are specific groups of materials that are suitable for practical use, such as nanoparticles and graphene. Therefore, nanoarchitecture of inorganic materials also has a more practical aspect. Based on these aspects, this review finally considers the future of materials nanoarchitectonics for further advanced devices.
Collapse
Affiliation(s)
- Katsuhiko Ariga
- Research Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa 277-8561, Chiba, Japan
| |
Collapse
|
24
|
Bai L, Chen X, Li C, Zhou H, Li Y, Xiao J, Zhang F, Cheng H, Zhou M. Mannose/stearyl chloride doubly functionalized polyethylenimine as a nucleic acid vaccine carrier to promote macrophage uptake. Drug Deliv 2024; 31:2427138. [PMID: 39540234 PMCID: PMC11565675 DOI: 10.1080/10717544.2024.2427138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Transmembrane transport remains a significant challenge for nucleic acid vaccine vectors. Promoting the ability of immune cells, such as macrophages, to capture foreign stimuli is also an effective approach to improving cross-presentation. In addition, polyethyleneimine (PEI) has gained attention in the field of nucleic acid vaccine carriers due to its excellent gene transfection efficiency and unique proton buffering effect. However, although high molecular weight PEI exhibits high efficiency, its high-density positive charges make it highly toxic, which limits its application. In this study, mannose/stearyl chloride functionalized polyethylenimine (SA-Man-PEI) was prepared by functionalizing PEI (molecular weight of 25 kDa) with mannose with immunomodulatory and phagocyte targeting effects, and an alkyl hydrophobic chain segment, which could easily promote cell uptake. Moreover, the functionalized-PEI retains a strong proton buffering effect, which helps the carrier escape from the lysosome. The particle sizes of the composite particles formed by SA-Man-PEI and ovalbumin (OVA) were below 200 nm, with good storage stability at both 4 °C and 37 °C. At a drug concentration of 2 μg/mL, the cell survival rate of functionalized-PEI was 19.2% higher than that of unfunctionalized PEI. In vitro macrophage endocytosis experiments showed that SA-Man-PEI could significantly enhance the macrophage uptake of composite particles, compared to unfunctionalized PEI or single-functionalized PEI. This study offers a new approach for developing PEI as a nucleic acid vaccine carrier, which could simultaneously enhance cell targeting and promote cell uptake.
Collapse
Affiliation(s)
- Lu Bai
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Xiaoqi Chen
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Chengyu Li
- School of Materials Science and Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei Province, China
| | - Haijun Zhou
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Yantao Li
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Jijun Xiao
- School of Materials Science and Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei Province, China
| | - Fen Zhang
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Hua Cheng
- Institute of Biology, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Mengmeng Zhou
- Shijiazhuang Polymer Composite Technological Innovation Center; Shijiazhuang Key Laboratory of Low Carbon Energy Materials, College of Chemical Engineering, Shijiazhuang University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
25
|
Li Q, Byun J, Kim D, Wu Y, Lee J, Oh YK. Cell membrane-coated mRNA nanoparticles for enhanced delivery to dendritic cells and immunotherapy. Asian J Pharm Sci 2024; 19:100968. [PMID: 39640052 PMCID: PMC11617980 DOI: 10.1016/j.ajps.2024.100968] [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: 02/16/2024] [Revised: 06/30/2024] [Accepted: 07/24/2024] [Indexed: 12/07/2024] Open
Abstract
Cationic polymers such as polyethylenimine have been considered promising carriers for mRNA vaccines. However, their application is hindered by their inherent toxicity and a lack of targeted delivery capability. These issues need to be addressed to develop effective cancer vaccines. In this study, we investigated whether dendritic cell membrane-coated polyethylenimine/mRNA nanoparticles (DPN) could effectively deliver mRNA to dendritic cells and induce immune responses. For comparison, we employed red blood cell membrane-coated polyethylenimine/mRNA (RPN) and plain polyethylenimine/mRNA polyplex (PN). The dendritic cell membrane coating altered the zeta potential values and surface protein patterns of PN. DPN demonstrated significantly higher uptake in dendritic cells compared to PN and RPN, and it also showed greater mRNA expression within these cells. DPN, carrying mRNA encoding luciferase, enhanced green fluorescent protein, or ovalbumin (OVA), exhibited higher protein expression in dendritic cells than the other groups. Additionally, DPN exhibited favorable mRNA escape from lysosomes post-internalization into dendritic cells. In mice, subcutaneous administration of DPN containing ovalbumin mRNA (DPNOVA) elicited higher titers of anti-OVA IgG antibodies and a greater population of OVA-specific CD8+ T cells than the other groups. In a B16F10-OVA tumor model, DPNOVA treatment resulted in the lowest tumor growth among the treated groups. Moreover, the population of OVA-specific CD8+ T cells was the highest in the DPNOVA-treated group. While we demonstrated DPN's feasibility as an mRNA delivery system in a tumor model, the potential of DPN can be broadly extended for immunotherapeutic treatments of various diseases through mRNA delivery to antigen-presenting cells.
Collapse
Affiliation(s)
| | | | - Dongyoon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
26
|
Laila UE, An W, Xu ZX. Emerging prospects of mRNA cancer vaccines: mechanisms, formulations, and challenges in cancer immunotherapy. Front Immunol 2024; 15:1448489. [PMID: 39654897 PMCID: PMC11625737 DOI: 10.3389/fimmu.2024.1448489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/18/2024] [Indexed: 12/12/2024] Open
Abstract
Cancer continues to pose an alarming threat to global health, necessitating the need for the development of efficient therapeutic solutions despite massive advances in the treatment. mRNA cancer vaccines have emerged as a hopeful avenue, propelled by the victory of mRNA technology in COVID-19 vaccines. The article delves into the intricate mechanisms and formulations of cancer vaccines, highlighting the ongoing efforts to strengthen mRNA stability and ensure successful translation inside target cells. Moreover, it discusses the design and mechanism of action of mRNA, showcasing its potential as a useful benchmark for developing efficacious cancer vaccines. The significance of mRNA therapy and selecting appropriate tumor antigens for the personalized development of mRNA vaccines are emphasized, providing insights into the immune mechanism. Additionally, the review explores the integration of mRNA vaccines with other immunotherapies and the utilization of progressive delivery platforms, such as lipid nanoparticles, to improve immune responses and address challenges related to immune evasion and tumor heterogeneity. While underscoring the advantages of mRNA vaccines, the review also addresses the challenges associated with the susceptibility of RNA to degradation and the difficulty in identifying optimum tumor-specific antigens, along with the potential solutions. Furthermore, it provides a comprehensive overview of the ongoing research efforts aimed at addressing these hurdles and enhancing the effectiveness of mRNA-based cancer vaccines. Overall, this review is a focused and inclusive impression of the present state of mRNA cancer vaccines, outlining their possibilities, challenges, and future predictions in the fight against cancer, ultimately aiding in the development of more targeted therapies against cancer.
Collapse
Affiliation(s)
| | | | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| |
Collapse
|
27
|
Tani H. Recent Advances and Prospects in RNA Drug Development. Int J Mol Sci 2024; 25:12284. [PMID: 39596348 PMCID: PMC11594839 DOI: 10.3390/ijms252212284] [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: 10/23/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
RNA therapeutics have undergone remarkable evolution since their inception in the late 1970s, revolutionizing medicine by offering new possibilities for treating previously intractable diseases. The field encompasses various modalities, including antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs), each with unique mechanisms and applications. The foundation was laid in 1978 with the discovery that synthetic oligonucleotides could inhibit viral replication, followed by pivotal developments such as RNA interference's discovery in 1998. The COVID-19 pandemic marked a crucial turning point, demonstrating the potential of mRNA vaccines and accelerating interest in RNA-based approaches. However, significant challenges remain, including stability issues, delivery to target tissues, potential off-target effects, and immunogenicity concerns. Recent advancements in chemical modifications, delivery systems, and the integration of AI technologies are addressing these challenges. The field has seen notable successes, such as approved treatments for spinal muscular atrophy and hereditary transthyretin-mediated amyloidosis. Looking ahead, RNA therapeutics show promise for personalized medicine approaches, particularly in treating genetic disorders and cancer. The continued evolution of this field, driven by technological innovations and deeper understanding of RNA biology, suggests a transformative impact on future medical treatments. The purpose of this review is to provide a comprehensive overview of the evolution, current state, and prospects of RNA therapeutics.
Collapse
Affiliation(s)
- Hidenori Tani
- Department of Health Pharmacy, Yokohama University of Pharmacy, 601 Matano, Totsuka, Yokohama 245-0066, Japan
| |
Collapse
|
28
|
Shi Y, Shi M, Wang Y, You J. Progress and prospects of mRNA-based drugs in pre-clinical and clinical applications. Signal Transduct Target Ther 2024; 9:322. [PMID: 39543114 PMCID: PMC11564800 DOI: 10.1038/s41392-024-02002-z] [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: 06/02/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 11/17/2024] Open
Abstract
In the last decade, messenger ribonucleic acid (mRNA)-based drugs have gained great interest in both immunotherapy and non-immunogenic applications. This surge in interest can be largely attributed to the demonstration of distinct advantages offered by various mRNA molecules, alongside the rapid advancements in nucleic acid delivery systems. It is noteworthy that the immunogenicity of mRNA drugs presents a double-edged sword. In the context of immunotherapy, extra supplementation of adjuvant is generally required for induction of robust immune responses. Conversely, in non-immunotherapeutic scenarios, immune activation is unwanted considering the host tolerability and high expression demand for mRNA-encoded functional proteins. Herein, mainly focused on the linear non-replicating mRNA, we overview the preclinical and clinical progress and prospects of mRNA medicines encompassing vaccines and other therapeutics. We also highlight the importance of focusing on the host-specific variations, including age, gender, pathological condition, and concurrent medication of individual patient, for maximized efficacy and safety upon mRNA administration. Furthermore, we deliberate on the potential challenges that mRNA drugs may encounter in the realm of disease treatment, the current endeavors of improvement, as well as the application prospects for future advancements. Overall, this review aims to present a comprehensive understanding of mRNA-based therapies while illuminating the prospective development and clinical application of mRNA drugs.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Meixing Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, P. R. China.
- The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, P. R. China.
- Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, P. R. China.
| |
Collapse
|
29
|
Parvin N, Mandal TK, Joo SW. The Impact of COVID-19 on RNA Therapeutics: A Surge in Lipid Nanoparticles and Alternative Delivery Systems. Pharmaceutics 2024; 16:1366. [PMID: 39598489 PMCID: PMC11597542 DOI: 10.3390/pharmaceutics16111366] [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: 10/02/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
The COVID-19 pandemic has significantly accelerated progress in RNA-based therapeutics, particularly through the successful development and global rollout of mRNA vaccines. This review delves into the transformative impact of the pandemic on RNA therapeutics, with a strong focus on lipid nanoparticles (LNPs) as a pivotal delivery platform. LNPs have proven to be critical in enhancing the stability, bioavailability, and targeted delivery of mRNA, facilitating the unprecedented success of vaccines like those developed by Pfizer-BioNTech and Moderna. Beyond vaccines, LNP technology is being explored for broader therapeutic applications, including treatments for cancer, rare genetic disorders, and infectious diseases. This review also discusses emerging RNA delivery systems, such as polymeric nanoparticles and viral vectors, which offer alternative strategies to overcome existing challenges related to stability, immune responses, and tissue-specific targeting. Additionally, we examine the pandemic's influence on regulatory processes, including the fast-tracked approvals for RNA therapies, and the surge in research funding that has spurred further innovation in the field. Public acceptance of RNA-based treatments has also grown, laying the groundwork for future developments in personalized medicine. By providing an in-depth analysis of these advancements, this review highlights the long-term impact of COVID-19 on the evolution of RNA therapeutics and the future of precision drug delivery technologies.
Collapse
Affiliation(s)
| | - Tapas K. Mandal
- School of Mechanical Engineering, School of Basic Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Sang-Woo Joo
- School of Mechanical Engineering, School of Basic Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
30
|
Dong C, Tan D, Sun H, Li Z, Zhang L, Zheng Y, Liu S, Zhang Y, He Q. Interleukin-12 Delivery Strategies and Advances in Tumor Immunotherapy. Curr Issues Mol Biol 2024; 46:11548-11579. [PMID: 39451566 PMCID: PMC11506767 DOI: 10.3390/cimb46100686] [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] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Interleukin-12 (IL-12) is considered to be a promising cytokine for enhancing an antitumor immune response; however, recombinant IL-12 has shown significant toxicity and limited efficacy in early clinical trials. Recently, many strategies for delivering IL-12 to tumor tissues have been developed, such as modifying IL-12, utilizing viral vectors, non-viral vectors, and cellular vectors. Previous studies have found that the fusion of IL-12 with extracellular matrix proteins, collagen, and immune factors is a way to enhance its therapeutic potential. In addition, studies have demonstrated that viral vectors are a good platform, and a variety of viruses such as oncolytic viruses, adenoviruses, and poxviruses have been used to deliver IL-12-with testing previously conducted in various cancer models. The local expression of IL-12 in tumors based on viral delivery avoids systemic toxicity while inducing effective antitumor immunity and acting synergistically with other therapies without compromising safety. In addition, lipid nanoparticles are currently considered to be the most mature drug delivery system. Moreover, cells are also considered to be drug carriers because they can effectively deliver therapeutic substances to tumors. In this article, we will systematically discuss the anti-tumor effects of IL-12 on its own or in combination with other therapies based on different delivery strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qing He
- State Key Laboratory of Drug Regulatory Sciences, National Institutes for Food and Drug Control, Beijing 102629, China; (C.D.); (D.T.); (H.S.); (Z.L.); (L.Z.); (Y.Z.); (S.L.); (Y.Z.)
| |
Collapse
|
31
|
Garcia-Atutxa I, Mondragon-Teran P, Huerta-Saquero A, Villanueva-Flores F. Advancements in monkeypox vaccines development: a critical review of emerging technologies. Front Immunol 2024; 15:1456060. [PMID: 39464881 PMCID: PMC11502315 DOI: 10.3389/fimmu.2024.1456060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/20/2024] [Indexed: 10/29/2024] Open
Abstract
Monkeypox (mpox) is a zoonotic illness caused by the monkeypox virus (MPXV), with higher health concerns among people who are pregnant, children, and persons who are immunocompromised, including people with untreated and advanced HIV disease. Significant progress has been made in developing vaccines against mpox, yet critical challenges and limitations persist in ensuring their effectiveness, safety, and accessibility. The pertinence of this review is highlighted by the World Health Organization's declaration of a global health emergency on August 14, 2024, due to the recent mpox outbreak, underscoring the critical necessity for effective vaccine solutions in the face of a rapidly evolving virus. Here, we comprehensively analyze various vaccine platforms utilized in mpox prevention, including attenuated and non-replicating virus vaccines, viral vector-based vaccines, recombinant protein vaccines, and DNA and mRNA vaccines. We evaluate the advantages and limitations of each platform, highlighting the urgent need for ongoing research and innovation to enhance vaccine efficacy and safety. Recent advancements, such as incorporating immunostimulatory sequences, improved delivery systems, and developing polyvalent vaccines, are explored for their potential to offer broader protection against diverse orthopoxvirus strains. This work underscores the need to optimize currently available vaccines and investigate novel vaccination strategies to address future public health emergencies effectively. By focusing on these advanced methodologies, we aim to contribute to the development of robust and adaptable vaccine solutions for mpox and other related viral threats.
Collapse
Affiliation(s)
- Igor Garcia-Atutxa
- Computer Science Department, Universidad Católica de Murcia (UCAM), Murcia, Spain
| | - Paul Mondragon-Teran
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| | - Alejandro Huerta-Saquero
- Departamento de Bionanotecnología, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México (UNAM), Ensenada, Mexico
| | - Francisca Villanueva-Flores
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| |
Collapse
|
32
|
Ma D, Xie A, Lv J, Min X, Zhang X, Zhou Q, Gao D, Wang E, Gao L, Cheng L, Liu S. Engineered extracellular vesicles enable high-efficient delivery of intracellular therapeutic proteins. Protein Cell 2024; 15:724-743. [PMID: 38518087 PMCID: PMC11443452 DOI: 10.1093/procel/pwae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 02/19/2024] [Indexed: 03/24/2024] Open
Abstract
Developing an intracellular delivery system is of key importance in the expansion of protein-based therapeutics acting on cytosolic or nuclear targets. Recently, extracellular vesicles (EVs) have been exploited as next-generation delivery modalities due to their natural role in intercellular communication and biocompatibility. However, fusion of protein of interest to a scaffold represents a widely used strategy for cargo enrichment in EVs, which could compromise the stability and functionality of cargo. Herein, we report intracellular delivery via EV-based approach (IDEA) that efficiently packages and delivers native proteins both in vitro and in vivo without the use of a scaffold. As a proof-of-concept, we applied the IDEA to deliver cyclic GMP-AMP synthase (cGAS), an innate immune sensor. The results showed that cGAS-carrying EVs activated interferon signaling and elicited enhanced antitumor immunity in multiple syngeneic tumor models. Combining cGAS EVs with immune checkpoint inhibition further synergistically boosted antitumor efficacy in vivo. Mechanistically, scRNA-seq demonstrated that cGAS EVs mediated significant remodeling of intratumoral microenvironment, revealing a pivotal role of infiltrating neutrophils in the antitumor immune milieu. Collectively, IDEA, as a universal and facile strategy, can be applied to expand and advance the development of protein-based therapeutics.
Collapse
Affiliation(s)
- Ding Ma
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - An Xie
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Jiahui Lv
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Xiaolin Min
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Xinye Zhang
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Qian Zhou
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Daxing Gao
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Enyu Wang
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
| | - Lei Gao
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
| | - Linzhao Cheng
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Senquan Liu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
33
|
Norimatsu J, Mizuno HL, Watanabe T, Obara T, Nakakido M, Tsumoto K, Cabral H, Kuroda D, Anraku Y. Triphenylphosphonium-modified catiomers enhance in vivo mRNA delivery through stabilized polyion complexation. MATERIALS HORIZONS 2024; 11:4711-4721. [PMID: 38988276 DOI: 10.1039/d4mh00325j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Nanocarriers based on cationic materials play a central role in the success of mRNA-based therapies. Traditionally, amine-bearing lipids and polymers have been successfully employed for creating mRNA-loaded nanocarriers, though they still present challenges, such as physical and biological instability, limiting both delivery efficiency and therapeutic potential. Non-amine cations could be a promising avenue in addressing these limitations. However, such alternatives remain notably underexplored. Herein, we introduced triphenylphosphonium (TPP) as an alternative cationic moiety for mRNA delivery, leveraging its advantageous properties for nucleic acid complexation. Through the modification of amine-bearing catiomers, we replaced traditional amine-based counterparts with TPP to create innovative polymeric micelles as mRNA nanocarriers. A comprehensive analysis, encompassing physicochemical, thermodynamic, and computational approaches, revealed that the TPP substitution significantly influenced polymer self-assembly, mRNA binding, and the overall stability of mRNA-loaded polymeric micelles. Upon intravenous injection, TPP-bearing micelles demonstrated a remarkable increase in mRNA bioavailability, facilitating efficient protein production in solid tumors. These findings provide a compelling rationale for substituting amines with TPP, emphasizing their potential for advancing mRNA therapeutics.
Collapse
Affiliation(s)
- Jumpei Norimatsu
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Hayato L Mizuno
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo 152-8550, Japan.
| | - Takayoshi Watanabe
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Takumi Obara
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Makoto Nakakido
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- The Institute of Medical Sciences, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kanagawa 210-0821, Japan
| | - Daisuke Kuroda
- Research Center of Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan.
| | - Yasutaka Anraku
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo 152-8550, Japan.
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kanagawa 210-0821, Japan
| |
Collapse
|
34
|
Neill B, Romero AR, Fenton OS. Advances in Nonviral mRNA Delivery Materials and Their Application as Vaccines for Melanoma Therapy. ACS APPLIED BIO MATERIALS 2024; 7:4894-4913. [PMID: 37930174 PMCID: PMC11220486 DOI: 10.1021/acsabm.3c00721] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Messenger RNA (mRNA) vaccines are promising platforms for cancer immunotherapy because of their potential to encode for a variety of tumor antigens, high tolerability, and capacity to induce strong antitumor immune responses. However, the clinical translation of mRNA cancer vaccines can be hindered by the inefficient delivery of mRNA in vivo. In this review, we provide an overview of mRNA cancer vaccines by discussing their utility in treating melanoma. Specifically, we begin our review by describing the barriers that can impede mRNA delivery to target cells. We then review native mRNA structure and discuss various modification methods shown to enhance mRNA stability and transfection. Next, we outline the advantages and challenges of three nonviral carrier platforms (lipid nanoparticles, polymeric nanoparticles, and lipopolyplexes) frequently used for mRNA delivery. Last, we summarize preclinical and clinical studies that have investigated nonviral mRNA vaccines for the treatment of melanoma. In writing this review, we aim to highlight innovative nonviral strategies designed to address mRNA delivery challenges while emphasizing the exciting potential of mRNA vaccines as next-generation therapies for the treatment of cancers.
Collapse
Affiliation(s)
- Bevin Neill
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adriana Retamales Romero
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Owen S. Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
35
|
Isaac I, Bhatia M, Bhattacharya C. Recent Advances in Biomaterials for mRNA Delivery to Immune Cells. ACS APPLIED BIO MATERIALS 2024; 7:5136-5146. [PMID: 39058246 DOI: 10.1021/acsabm.4c00734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Modulating the function of immune cells by targeting the cells themselves has become a key strategy in immunotherapy for combating various diseases such as cancer, autoimmune disorders, and infectious ailments. The use of mRNA (mRNA) is a powerful tool for transiently inducing protein expression, which is often used for genetic manipulation. However, its inherent instability and inability to precisely reach target cells necessitate the use of biomaterials for safe and effective delivery. Additionally, transfecting immune cells is difficult and complex due to their resistance mechanisms, signaling pathways, and cellular interactions. This review focuses on the recent development of biomaterials for mRNA delivery to immune cells, including lipid nanoparticles and polymeric carriers. It also outlines the challenges of targeting and delivering therapeutic payloads to immune cells, providing commentary and outlook on the design of next-generation materials. Finally, this approach has the potential to significantly enhance the precision and effectiveness of therapeutic interventions for various diseases, shaping the future of mRNA delivery for immune conditions.
Collapse
Affiliation(s)
- Ivan Isaac
- Department of Chemistry and Biochemistry, University of Nevada─Las Vegas 4505 S. Maryland Parkway, Las Vegas, Nevada 89154, United States
| | - Mayurakkhi Bhatia
- Department of Chemistry and Biochemistry, University of Nevada─Las Vegas 4505 S. Maryland Parkway, Las Vegas, Nevada 89154, United States
| | - Chandrabali Bhattacharya
- Department of Chemistry and Biochemistry, University of Nevada─Las Vegas 4505 S. Maryland Parkway, Las Vegas, Nevada 89154, United States
- Interdisciplinary Biomedical Engineering Program, University of Nevada─Las Vegas, Las Vegas, Nevada 89154, United States
| |
Collapse
|
36
|
Li J, Zhang Y, Yang YG, Sun T. Advancing mRNA Therapeutics: The Role and Future of Nanoparticle Delivery Systems. Mol Pharm 2024; 21:3743-3763. [PMID: 38953708 DOI: 10.1021/acs.molpharmaceut.4c00276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The coronavirus (COVID-19) pandemic has underscored the critical role of mRNA-based vaccines as powerful, adaptable, readily manufacturable, and safe methodologies for prophylaxis. mRNA-based treatments are emerging as a hopeful avenue for a plethora of conditions, encompassing infectious diseases, cancer, autoimmune diseases, genetic diseases, and rare disorders. Nonetheless, the in vivo delivery of mRNA faces challenges due to its instability, suboptimal delivery, and potential for triggering undesired immune reactions. In this context, the development of effective drug delivery systems, particularly nanoparticles (NPs), is paramount. Tailored with biophysical and chemical properties and susceptible to surface customization, these NPs have demonstrated enhanced mRNA delivery in vivo and led to the approval of several NPs-based formulations for clinical use. Despite these advancements, the necessity for developing a refined, targeted NP delivery system remains imperative. This review comprehensively surveys the biological, translational, and clinical progress in NPs-mediated mRNA therapeutics for both the prevention and treatment of diverse diseases. By addressing critical factors for enhancing existing methodologies, it aims to inform the future development of precise and efficacious mRNA-based therapeutic interventions.
Collapse
Affiliation(s)
- Jiaxuan Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
- International Center of Future Science, Jilin University, Changchun, Jilin 130021, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
- International Center of Future Science, Jilin University, Changchun, Jilin 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
37
|
Wu Z, Sun W, Qi H. Recent Advancements in mRNA Vaccines: From Target Selection to Delivery Systems. Vaccines (Basel) 2024; 12:873. [PMID: 39203999 PMCID: PMC11359327 DOI: 10.3390/vaccines12080873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 09/03/2024] Open
Abstract
mRNA vaccines are leading a medical revolution. mRNA technologies utilize the host's own cells as bio-factories to produce proteins that serve as antigens. This revolutionary approach circumvents the complicated processes involved in traditional vaccine production and empowers vaccines with the ability to respond to emerging or mutated infectious diseases rapidly. Additionally, the robust cellular immune response elicited by mRNA vaccines has shown significant promise in cancer treatment. However, the inherent instability of mRNA and the complexity of tumor immunity have limited its broader application. Although the emergence of pseudouridine and ionizable cationic lipid nanoparticles (LNPs) made the clinical application of mRNA possible, there remains substantial potential for further improvement of the immunogenicity of delivered antigens and preventive or therapeutic effects of mRNA technology. Here, we review the latest advancements in mRNA vaccines, including but not limited to target selection and delivery systems. This review offers a multifaceted perspective on this rapidly evolving field.
Collapse
Affiliation(s)
- Zhongyan Wu
- Newish Biological R&D Center, Beijing 100101, China;
| | - Weilu Sun
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK;
| | - Hailong Qi
- Newish Biological R&D Center, Beijing 100101, China;
| |
Collapse
|
38
|
Almeida B, Dias TR, Cruz P, Sousa-Pimenta M, Teixeira AL, Pereira CE, Costa-Silva B, Oliveira J, Medeiros R, Dias F. Plasma EV-miRNAs as Potential Biomarkers of COVID-19 Vaccine Immune Response in Cancer Patients. Vaccines (Basel) 2024; 12:848. [PMID: 39203974 PMCID: PMC11359428 DOI: 10.3390/vaccines12080848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024] Open
Abstract
Cancer patients, prone to severe COVID-19, face immune challenges due to their disease and treatments. Identifying biomarkers, particularly extracellular vesicle (EV)-derived microRNAs (miRNAs), is vital for comprehending their response to COVID-19 vaccination. Therefore, this study aimed to investigate specific EV-miRNAs in the plasma of cancer patients under active treatment who received the COVID-19 booster vaccine. The selected miRNAs (EV-hsa-miR-7-5p, EV-hsa-miR-15b-5p, EV-hsa-miR-24-3p, EV-hsa-miR-145- 5p, and EV-hsa-miR-223-3p) are involved in regulating SARS-CoV-2 spike protein and cytokine release, making them potential biomarkers for vaccination response. The study involved 54 cancer patients. Plasma and serum samples were collected at pre-boost vaccination, and at 3 and 6 months post-boost vaccination. Anti-spike antibody levels were measured. Additionally, RNA was extracted from EVs isolated from plasma and the expression levels of miRNAs were assessed. The results showed a significantly positive antibody response after COVID-19 boost vaccination. The expression levels of EV-hsa-miR-7-5p, EV-hsa-miR-15b-5p, EV-hsa-miR-24-3p, and EV-hsa-miR-223-3p increased significantly after 6 months of COVID-19 booster vaccination. Interestingly, an increased expression of certain EV-hsa-miRNAs was positively correlated. Bioinformatic analysis revealed that these correlated miRNAs play a critical role in regulating the targets present in antiviral responses and cytokine production. These findings suggest that EV-hsa-miR-15b-5p, EV-hsa-miR-24-3p, and EV-hsa-miR-223-3p may be crucial in immune response induced by mRNA vaccines.
Collapse
Affiliation(s)
- Beatriz Almeida
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) & RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto. CCC), 4200-072 Porto, Portugal; (B.A.); (T.R.D.); (A.L.T.); (R.M.)
- Research Department, Portuguese League Against Cancer Northern Branch (LPCC-NRN), 4200-172 Porto, Portugal
| | - Tânia R. Dias
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) & RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto. CCC), 4200-072 Porto, Portugal; (B.A.); (T.R.D.); (A.L.T.); (R.M.)
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-523 Porto, Portugal
| | - Pedro Cruz
- Department of Oncology, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (Porto. CCC), 4200-072 Porto, Portugal; (P.C.); (J.O.)
| | - Mário Sousa-Pimenta
- Department of Onco-Hematology, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (Porto. CCC), 4200-072 Porto, Portugal;
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) & RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto. CCC), 4200-072 Porto, Portugal; (B.A.); (T.R.D.); (A.L.T.); (R.M.)
| | - Catarina Esteves Pereira
- Systems Oncology Group, Champalimaud Research, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal; (C.E.P.); (B.C.-S.)
| | - Bruno Costa-Silva
- Systems Oncology Group, Champalimaud Research, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal; (C.E.P.); (B.C.-S.)
| | - Júlio Oliveira
- Department of Oncology, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (Porto. CCC), 4200-072 Porto, Portugal; (P.C.); (J.O.)
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) & RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto. CCC), 4200-072 Porto, Portugal; (B.A.); (T.R.D.); (A.L.T.); (R.M.)
- Research Department, Portuguese League Against Cancer Northern Branch (LPCC-NRN), 4200-172 Porto, Portugal
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-523 Porto, Portugal
- Laboratory Medicine, Clinical Pathology Department, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (Porto. CCC), 4200-072 Porto, Portugal
- Biomedicine Research Center (CEBIMED), Research Innovation and Development Institute (FP-I3ID), 4249-004 Porto, Portugal
| | - Francisca Dias
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) & RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto. CCC), 4200-072 Porto, Portugal; (B.A.); (T.R.D.); (A.L.T.); (R.M.)
| |
Collapse
|
39
|
Ariga K. Liquid-Liquid and Liquid-Solid Interfacial Nanoarchitectonics. Molecules 2024; 29:3168. [PMID: 38999120 PMCID: PMC11243083 DOI: 10.3390/molecules29133168] [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: 06/14/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
Nanoscale science is becoming increasingly important and prominent, and further development will necessitate integration with other material chemistries. In other words, it involves the construction of a methodology to build up materials based on nanoscale knowledge. This is also the beginning of the concept of post-nanotechnology. This role belongs to nanoarchitectonics, which has been rapidly developing in recent years. However, the scope of application of nanoarchitectonics is wide, and it is somewhat difficult to compile everything. Therefore, this review article will introduce the concepts of liquid and interface, which are the keywords for the organization of functional material systems in biological systems. The target interfaces are liquid-liquid interface, liquid-solid interface, and so on. Recent examples are summarized under the categories of molecular assembly, metal-organic framework and covalent organic framework, and living cell. In addition, the latest research on the liquid interfacial nanoarchitectonics of organic semiconductor film is also discussed. The final conclusive section summarizes these features and discusses the necessary components for the development of liquid interfacial nanoarchitectonics.
Collapse
Affiliation(s)
- Katsuhiko Ariga
- Research Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), Ibaraki 305-0044, Japan;
- Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan
| |
Collapse
|
40
|
Narasipura EA, Fenton OS. Advances in non-viral mRNA delivery to the spleen. Biomater Sci 2024; 12:3027-3044. [PMID: 38712531 PMCID: PMC11175841 DOI: 10.1039/d4bm00038b] [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: 05/08/2024]
Abstract
Developing safe and effective delivery strategies for localizing messenger RNA (mRNA) payloads to the spleen is an important goal in the field of genetic medicine. Accomplishing this goal is challenging due to the instability, size, and charge of mRNA payloads. Here, we provide an analysis of non-viral delivery technologies that have been developed to deliver mRNA payloads to the spleen. Specifically, our review begins by outlining the unique anatomy and potential targets for mRNA delivery within the spleen. Next, we describe approaches in mRNA sequence engineering that can be used to improve mRNA delivery to the spleen. Then, we describe advances in non-viral carrier systems that can package and deliver mRNA payloads to the spleen, highlighting key advances in the literature in lipid nanoparticle (LNP) and polymer nanoparticle (PNP) technology platforms. Finally, we provide commentary and outlook on how splenic mRNA delivery may afford next-generation treatments for autoimmune disorders and cancers. In undertaking this approach, our goal with this review is to both establish a fundamental understanding of drug delivery challenges associated with localizing mRNA payloads to the spleen, while also broadly highlighting the potential to use these genetic medicines to treat disease.
Collapse
Affiliation(s)
- Eshan A Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
41
|
Puccetti M, Pariano M, Schoubben A, Ricci M, Giovagnoli S. Engineering carrier nanoparticles with biomimetic moieties for improved intracellular targeted delivery of mRNA therapeutics and vaccines. J Pharm Pharmacol 2024; 76:592-605. [PMID: 38092697 DOI: 10.1093/jpp/rgad089] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/12/2023] [Indexed: 06/07/2024]
Abstract
Biological membrane-engineered lipid nanoparticles (LNP) have shown enormous potential as vehicles for drug delivery due to their outstanding biomimetic properties. To make these nanoparticles more adaptable to complex biological systems, several methods and cellular sources have been adopted to introduce biomembrane-derived moieties onto LNP and provide the latter with more functions while preserving their intrinsic nature. In this review, we focus on LNP decoration with specific regard to mRNA therapeutics and vaccines. The bio-engineering approach exploits a variety of biomembranes for functionalization, such as those derived from red blood cells, white blood cells, cancer cells, platelets, exosomes, and others. Biomembrane engineering could greatly enhance efficiency in targeted drug delivery, treatment, and diagnosis of cancer, inflammation, immunological diseases, and a variety of pathologic conditions. These membrane-modification techniques are expected to advance biomembrane-derived LNP into wider applications in the future.
Collapse
Affiliation(s)
- Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Aurelie Schoubben
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
42
|
Wu Y, Luo L, Hao Z, Liu D. DNA-based nanostructures for RNA delivery. MEDICAL REVIEW (2021) 2024; 4:207-224. [PMID: 38919398 PMCID: PMC11195427 DOI: 10.1515/mr-2023-0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/28/2024] [Indexed: 06/27/2024]
Abstract
RNA-based therapeutics have emerged as a promising approach for the treatment of various diseases, including cancer, genetic disorders, and infectious diseases. However, the delivery of RNA molecules into target cells has been a major challenge due to their susceptibility to degradation and inefficient cellular uptake. To overcome these hurdles, DNA-based nano technology offers an unprecedented opportunity as a potential delivery platform for RNA therapeutics. Due to its excellent characteristics such as programmability and biocompatibility, these DNA-based nanostructures, composed of DNA molecules assembled into precise and programmable structures, have garnered significant attention as ideal building materials for protecting and delivering RNA payloads to the desired cellular destinations. In this review, we highlight the current progress in the design and application of three DNA-based nanostructures: DNA origami, lipid-nanoparticle (LNP) technology related to frame guided assembly (FGA), and DNA hydrogel for the delivery of RNA molecules. Their biomedical applications are briefly discussed and the challenges and future perspectives in this field are also highlighted.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Beijing SupraCirc Biotechnology Co., Ltd, Beijing, China
| | - Liangzhi Luo
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Ziyang Hao
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Dongsheng Liu
- Department of Chemistry, Engineering Research Center of Advanced Rare Earth Materials, (Ministry of Education), Tsinghua University, Beijing, China
| |
Collapse
|
43
|
Tang Y, Liu B, Zhang Y, Liu Y, Huang Y, Fan W. Interactions between nanoparticles and lymphatic systems: Mechanisms and applications in drug delivery. Adv Drug Deliv Rev 2024; 209:115304. [PMID: 38599495 DOI: 10.1016/j.addr.2024.115304] [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: 12/12/2023] [Revised: 03/08/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
The lymphatic system has garnered significant attention in drug delivery research due to the advantages it offers, such as enhancing systemic exposure and enabling lymph node targeting for nanomedicines via the lymphatic delivery route. The journey of drug carriers involves transport from the administration site to the lymphatic vessels, traversing the lymph before entering the bloodstream or targeting specific lymph nodes. However, the anatomical and physiological barriers of the lymphatic system play a pivotal role in influencing the behavior and efficiency of carriers. To expedite research and subsequent clinical translation, this review begins by introducing the composition and classification of the lymphatic system. Subsequently, we explore the routes and mechanisms through which nanoparticles enter lymphatic vessels and lymph nodes. The review further delves into the interactions between nanomedicine and body fluids at the administration site or within lymphatic vessels. Finally, we provide a comprehensive overview of recent advancements in lymphatic delivery systems, addressing the challenges and opportunities inherent in current systems for delivering macromolecules and vaccines.
Collapse
Affiliation(s)
- Yisi Tang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Bao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuting Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| | - Wufa Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
44
|
Kutikuppala LVS, Kourampi I, Kanagala RSD, Bhattacharjee P, Boppana SH. Prospects and Challenges in Developing mRNA Vaccines for Infectious Diseases and Oncogenic Viruses. Med Sci (Basel) 2024; 12:28. [PMID: 38804384 PMCID: PMC11130901 DOI: 10.3390/medsci12020028] [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: 10/16/2023] [Revised: 02/12/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
mRNA vaccines have emerged as an optimistic technological platform for vaccine innovation in this new scientific era. mRNA vaccines have dramatically altered the domain of vaccinology by offering a versatile and rapid approach to combating infectious diseases and virus-induced cancers. Clinical trials have demonstrated efficacy rates of 94-95% in preventing COVID-19, and mRNA vaccines have been increasingly recognized as a powerful vaccine platform. Although mRNA vaccines have played an essential role in the COVID-19 pandemic, they still have several limitations; their instability and degradation affect their storage, delivery, and over-all efficiency. mRNA is typically enclosed in a transport mechanism to facilitate its entry into the target cell because it is an unstable and negatively charged molecule. For instance, mRNA that is given using lipid-nanoparticle-based vaccine delivery systems (LNPs) solely enters cells through endocytosis, establishing an endosome without damaging the cell membrane. The COVID-19 pandemic has accelerated the development of mRNA vaccine platforms used to treat and prevent several infectious diseases. This technology has the potential to change the future course of the disease by providing a safe and effective way to combat infectious diseases and cancer. A single-stranded genetic sequence found in mRNA vaccines instructs host cells to produce proteins inside ribosomes to elicit immunological responses and prepare the immune system to fight infections or cancer cells. The potential applications of mRNA vaccine technology are vast and can lead to the development of a preferred vaccine pattern. As a result, a new generation of vaccinations has gradually gained popularity and access to the general population. To adapt the design of an antigen, and even combine sequences from different variations in response to new changes in the viral genome, mRNA vaccines may be used. Current mRNA vaccines provide adequate safety and protection, but the duration of that protection can only be determined if further clinical research is conducted.
Collapse
Affiliation(s)
| | - Islam Kourampi
- Department of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ramya S. D. Kanagala
- Department of Medicine, Dr. KNR University of Health Sciences, Warangal 506007, India;
| | | | - Sri Harsha Boppana
- Department of Anesthesia and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
| |
Collapse
|
45
|
Opsomer L, Jana S, Mertens I, Cui X, Hoogenboom R, Sanders NN. Efficient in vitro and in vivo transfection of self-amplifying mRNA with linear poly(propylenimine) and poly(ethylenimine-propylenimine) random copolymers as non-viral carriers. J Mater Chem B 2024; 12:3927-3946. [PMID: 38563779 DOI: 10.1039/d3tb03003b] [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: 04/04/2024]
Abstract
Messenger RNA (mRNA) based vaccines have been introduced worldwide to combat the Covid-19 pandemic. These vaccines consist of non-amplifying mRNA formulated in lipid nanoparticles (LNPs). Consequently, LNPs are considered benchmark non-viral carriers for nucleic acid delivery. However, the formulation and manufacturing of these mRNA-LNP nanoparticles are expensive and time-consuming. Therefore, we used self-amplifying mRNA (saRNA) and synthesized novel polymers as alternative non-viral carrier platform to LNPs, which enable a simple, rapid, one-pot formulation of saRNA-polyplexes. Our novel polymer-based carrier platform consists of randomly concatenated ethylenimine and propylenimine comonomers, resulting in linear, poly(ethylenimine-ran-propylenimine) (L-PEIx-ran-PPIy) copolymers with controllable degrees of polymerization. Here we demonstrate in multiple cell lines, that our saRNA-polyplexes show comparable to higher in vitro saRNA transfection efficiencies and higher cell viabilities compared to formulations with Lipofectamine MessengerMAX™ (LFMM), a commercial, lipid-based carrier considered to be the in vitro gold standard carrier. This is especially true for our in vitro best performing saRNA-polyplexes with N/P 5, which are characterised with a size below 100 nm, a positive zeta potential, a near 100% encapsulation efficiency, a high retention capacity and the ability to protect the saRNA from degradation mediated by RNase A. Furthermore, an ex vivo hemolysis assay with pig red blood cells demonstrated that the saRNA-polyplexes exhibit negligible hemolytic activity. Finally, a bioluminescence-based in vivo study was performed over a 35-day period, and showed that the polymers result in a higher and prolonged bioluminescent signal compared to naked saRNA and L-PEI based polyplexes. Moreover, the polymers show different expression profiles compared to those of LNPs, with one of our new polymers (L-PPI250) demonstrating a higher sustained expression for at least 35 days after injection.
Collapse
Affiliation(s)
- Lisa Opsomer
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium.
| | - Somdeb Jana
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, 9000 Ghent, Belgium.
| | - Ine Mertens
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, 9000 Ghent, Belgium.
| | - Xiaole Cui
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium.
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, 9000 Ghent, Belgium.
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium.
- Cancer Research Institute (CRIG), Ghent University, B-9000 Ghent, Belgium
| |
Collapse
|
46
|
Chen P, Yang W, Mochida Y, Li S, Hong T, Kinoh H, Kataoka K, Cabral H. Selective Intracellular Delivery of Antibodies in Cancer Cells with Nanocarriers Sensing Endo/Lysosomal Enzymatic Activity. Angew Chem Int Ed Engl 2024; 63:e202317817. [PMID: 38342757 DOI: 10.1002/anie.202317817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
The differential enzymatic activity in the endo/lysosomes of particular cells could trigger targeted endosomal escape functions, enabling selective intracellular protein delivery. However, this strategy may be jeopardized due to protein degradation during endosomal trafficking. Herein, using custom made fluorescent probes to assess the endosomal activity of cathepsin B (CTSB) and protein degradation, we found that certain cancer cells with hyperacidified endosomes grant a spatiotemporal window where CTSB activity surpass protein digestion. This inspired the engineering of antibody-loaded polymeric nanocarriers having CTSB-activatable endosomal escape ability. The nanocarriers selectively escaped from the endo/lysosomes in the cells with high endosomal CTSB activity and delivered active antibodies to intracellular targets. This study provides a viable strategy for cell-specific protein delivery using stimuli-responsive nanocarriers with controlled endosomal escape.
Collapse
Affiliation(s)
- Pengwen Chen
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Wenqian Yang
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yuki Mochida
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki, 210-0821, Japan
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Shangwei Li
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Taehun Hong
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroaki Kinoh
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki, 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki, 210-0821, Japan
| | - Horacio Cabral
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
47
|
Kim J, Eygeris Y, Ryals RC, Jozić A, Sahay G. Strategies for non-viral vectors targeting organs beyond the liver. NATURE NANOTECHNOLOGY 2024; 19:428-447. [PMID: 38151642 DOI: 10.1038/s41565-023-01563-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 11/01/2023] [Indexed: 12/29/2023]
Abstract
In recent years, nanoparticles have evolved to a clinical modality to deliver diverse nucleic acids. Rising interest in nanomedicines comes from proven safety and efficacy profiles established by continuous efforts to optimize physicochemical properties and endosomal escape. However, despite their transformative impact on the pharmaceutical industry, the clinical use of non-viral nucleic acid delivery is limited to hepatic diseases and vaccines due to liver accumulation. Overcoming liver tropism of nanoparticles is vital to meet clinical needs in other organs. Understanding the anatomical structure and physiological features of various organs would help to identify potential strategies for fine-tuning nanoparticle characteristics. In this Review, we discuss the source of liver tropism of non-viral vectors, present a brief overview of biological structure, processes and barriers in select organs, highlight approaches available to reach non-liver targets, and discuss techniques to accelerate the discovery of non-hepatic therapies.
Collapse
Affiliation(s)
- Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | - Yulia Eygeris
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Renee C Ryals
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| | - Antony Jozić
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA.
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA.
- Department of Biomedical Engineering, Robertson Life Sciences Building, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
48
|
Wang J, Zhu H, Gan J, Liang G, Li L, Zhao Y. Engineered mRNA Delivery Systems for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308029. [PMID: 37805865 DOI: 10.1002/adma.202308029] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/05/2023] [Indexed: 10/09/2023]
Abstract
Messenger RNA (mRNA)-based therapeutic strategies have shown remarkable promise in preventing and treating a staggering range of diseases. Optimizing the structure and delivery system of engineered mRNA has greatly improved its stability, immunogenicity, and protein expression levels, which has led to a wider range of uses for mRNA therapeutics. Herein, a thorough analysis of the optimization strategies used in the structure of mRNA is first provided and delivery systems are described in great detail. Furthermore, the latest advancements in biomedical engineering for mRNA technology, including its applications in combatting infectious diseases, treating cancer, providing protein replacement therapy, conducting gene editing, and more, are summarized. Lastly, a perspective on forthcoming challenges and prospects concerning the advancement of mRNA therapeutics is offered. Despite these challenges, mRNA-based therapeutics remain promising, with the potential to revolutionize disease treatment and contribute to significant advancements in the biomedical field.
Collapse
Affiliation(s)
- Ji Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Haofang Zhu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jingjing Gan
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Gaofeng Liang
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, 450009, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, 450009, China
| |
Collapse
|
49
|
DeLong RK, Nava-Chavez J, Kumar R, Mathew EN, Mwangi W, Yoon S. Enhancing RNA Payload and Temperature Stability and Activity with Cationic Peptide-Coated Zinc Oxide Nanoparticles. ACS Pharmacol Transl Sci 2024; 7:707-715. [PMID: 38481696 PMCID: PMC10928881 DOI: 10.1021/acsptsci.3c00280] [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: 10/13/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/09/2025]
Abstract
The lipid nanoparticle (LNP) mRNA vaccine was first tested through clinic but suffered from relatively low RNA payloads and poor temperature stability. Our lab patented a protamine-coated particle approach for temperature-stabilizing DNA vaccines, translating this successfully to the clinic. In subsequent work, we have characterized RNA interaction and delivery by zinc oxide nanoparticles, filing a patent most recently entitled RNA-stabilizing nanoparticles, similarly utilizing protamine-coated zinc oxide nanoparticles for RNA. Here, we present this data for the first time. Briefly, ZnO, ZnO-protamine, and ZnO-protamine-RNA were characterized by size and zeta potential analyses and the RNA-loaded nanoparticles were visualized by transmission electron microscopy. UV spectroscopic analysis demonstrated up to 95-98% loading efficiency with protamine and approximately 75% loading efficiency with LL37, another cationic antiviral peptide. Elution of the RNA isolated from the particles afforded a calculation in three independent trials where RNA payloads ranged from 18 to 45 μg of RNA per 0.5 mg of coated particles. Circular dichroism (CD) analysis indicated that binding of RNA to ZnO NPs stabilized, enhancing the pattern with a clear dependence on the RNA:ZnO stoichiometry. Enhanced temperature stability was shown by differential scanning calorimetry (DSC), gel electrophoresis, and in vitro mRNA expression analysis. Using poly I:C RNA with a well-defined melting point (64.3 ± 0.32 °C), formation of the ZnO:RNA complex increased the RNA melting point (70.9 ± 0.62 °C). After refrigerated or room-temperature storage or incubation at 30, 40, or 50 °C, RNA comigration with the control RNA was recovered from all samples, exposed to either 14 or 100 nm ZnO, and coated with protamine. Furthermore, the ZnO-protamine-mRNA samples retained significantly higher expression activity when incubated at these elevated temperatures. Finally, the ZnO-protamine-mRNA was functionally active for in vitro translation, in cell extracts, and in cells for expression of GFP, luciferase, and COVID spike protein. These data support further preclinical development of ZnO-protamine-mRNA.
Collapse
Affiliation(s)
- Robert K. DeLong
- Innovation
Development Laboratory, Landmark Bio, 300 North Beacon Street, Watertown, Massachusetts 02472, United States
| | - Juliet Nava-Chavez
- Department
of Biology, Kansas State University, Manhattan , Kansas66502, United States
| | - Rakshith Kumar
- Department
of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan , Kansas66052, United States
| | - Elza Neelima Mathew
- Department
of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan , Kansas66052, United States
- Department
of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan , Kansas66502, United States
| | - Waithaka Mwangi
- Department
of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan , Kansas66052, United States
| | - Sunyoung Yoon
- Kansas
State University, Manhattan , Kansas66502, United States
| |
Collapse
|
50
|
Ariga K. Confined Space Nanoarchitectonics for Dynamic Functions and Molecular Machines. MICROMACHINES 2024; 15:282. [PMID: 38399010 PMCID: PMC10892885 DOI: 10.3390/mi15020282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024]
Abstract
Nanotechnology has advanced the techniques for elucidating phenomena at the atomic, molecular, and nano-level. As a post nanotechnology concept, nanoarchitectonics has emerged to create functional materials from unit structures. Consider the material function when nanoarchitectonics enables the design of materials whose internal structure is controlled at the nanometer level. Material function is determined by two elements. These are the functional unit that forms the core of the function and the environment (matrix) that surrounds it. This review paper discusses the nanoarchitectonics of confined space, which is a field for controlling functional materials and molecular machines. The first few sections introduce some of the various dynamic functions in confined spaces, considering molecular space, materials space, and biospace. In the latter two sections, examples of research on the behavior of molecular machines, such as molecular motors, in confined spaces are discussed. In particular, surface space and internal nanospace are taken up as typical examples of confined space. What these examples show is that not only the central functional unit, but also the surrounding spatial configuration is necessary for higher functional expression. Nanoarchitectonics will play important roles in the architecture of such a total system.
Collapse
Affiliation(s)
- Katsuhiko Ariga
- Research Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan;
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa 277-8561, Japan
| |
Collapse
|