1
|
Gurrola TE, Effah SN, Sariyer IK, Dampier W, Nonnemacher MR, Wigdahl B. Delivering CRISPR to the HIV-1 reservoirs. Front Microbiol 2024; 15:1393974. [PMID: 38812680 PMCID: PMC11133543 DOI: 10.3389/fmicb.2024.1393974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/22/2024] [Indexed: 05/31/2024] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection is well known as one of the most complex and difficult viral infections to cure. The difficulty in developing curative strategies arises in large part from the development of latent viral reservoirs (LVRs) within anatomical and cellular compartments of a host. The clustered regularly interspaced short palindromic repeats/ CRISPR-associated protein 9 (CRISPR/Cas9) system shows remarkable potential for the inactivation and/or elimination of integrated proviral DNA within host cells, however, delivery of the CRISPR/Cas9 system to infected cells is still a challenge. In this review, the main factors impacting delivery, the challenges for delivery to each of the LVRs, and the current successes for delivery to each reservoir will be discussed.
Collapse
Affiliation(s)
- Theodore E. Gurrola
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Samuel N. Effah
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ilker K. Sariyer
- Department of Microbiology, Immunology, and Inflammation and Center for Neurovirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Will Dampier
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Michael R. Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
2
|
Zhang X, Li Y, Zhou Z. Lipid Nanoparticle-Based Delivery System-A Competing Place for mRNA Vaccines. ACS OMEGA 2024; 9:6219-6234. [PMID: 38371811 PMCID: PMC10870384 DOI: 10.1021/acsomega.3c08353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/24/2023] [Accepted: 12/29/2023] [Indexed: 02/20/2024]
Abstract
mRNA, as one of the foci of biomedical research in the past decade, has become a candidate vaccine solution for various infectious diseases and tumors and for regenerative medicine and immunotherapy due to its high efficiency, safety, and effectiveness. A stable and effective delivery system is needed to protect mRNAs from nuclease degradation while also enhancing immunogenicity. The success of mRNA lipid nanoparticles in treating COVID-19, to a certain extent, marks a milestone for mRNA vaccines and also promotes further research on mRNA delivery systems. Here, we explore mRNA vaccine delivery systems, especially lipid nanoparticles (LNPs), considering the current research status, prospects, and challenges of lipid nanoparticles, and explore other mRNA delivery systems.
Collapse
Affiliation(s)
- Xinyu Zhang
- Research
Center for Infectious Diseases, Tianjin
University of Traditional Chinese Medicine, 300193 Tianjin, China
- Institute
for Biological Product Control, National
Institutes for Food and Drug Control (NIFDC) and WHO Collaborating
Center for Standardization and Evaluation of Biologicals, No.31 Huatuo Street, Daxing District, 102629 Beijing, China
- College
of Life Science, Jilin University, 130012 Changchun, China
| | - Yuanfang Li
- Department
of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, 361015 Xiamen, Fujian China
| | - Zehua Zhou
- Research
Center for Infectious Diseases, Tianjin
University of Traditional Chinese Medicine, 300193 Tianjin, China
| |
Collapse
|
3
|
Lin L, Su K, Cheng Q, Liu S. Targeting materials and strategies for RNA delivery. Theranostics 2023; 13:4667-4693. [PMID: 37649616 PMCID: PMC10465230 DOI: 10.7150/thno.87316] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
RNA-based therapeutics have shown great promise in various medical applications, including cancers, infectious diseases, and metabolic diseases. The recent success of mRNA vaccines for combating the COVID-19 pandemic has highlighted the medical value of RNA drugs. However, one of the major challenges in realizing the full potential of RNA drugs is to deliver RNA into specific organs and tissues in a targeted manner, which is crucial for achieving therapeutic efficacy, reducing side effects, and enhancing overall treatment efficacy. Numerous attempts have been made to pursue targeting, nonetheless, the lack of clear guideline and commonality elucidation has hindered the clinical translation of RNA drugs. In this review, we outline the mechanisms of action for targeted RNA delivery systems and summarize four key factors that influence the targeting delivery of RNA drugs. These factors include the category of vector materials, chemical structures of vectors, administration routes, and physicochemical properties of RNA vectors, and they all notably contribute to specific organ/tissue tropism. Furthermore, we provide an overview of the main RNA-based drugs that are currently in clinical trials, highlighting their design strategies and tissue tropism applications. This review will aid to understand the principles and mechanisms of targeted delivery systems, accelerating the development of future RNA drugs for different diseases.
Collapse
Affiliation(s)
- Lixin Lin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kexin Su
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiang Cheng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Shuai Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
4
|
Karim ME, Haque ST, Al-Busaidi H, Bakhtiar A, Tha KK, Holl MMB, Chowdhury EH. Scope and challenges of nanoparticle-based mRNA delivery in cancer treatment. Arch Pharm Res 2022; 45:865-893. [DOI: 10.1007/s12272-022-01418-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022]
|
5
|
Zhang M, Hussain A, Yang H, Zhang J, Liang XJ, Huang Y. mRNA-based modalities for infectious disease management. NANO RESEARCH 2022; 16:672-691. [PMID: 35818566 PMCID: PMC9258466 DOI: 10.1007/s12274-022-4627-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 06/15/2023]
Abstract
The novel coronavirus disease 2019 (COVID-19) is still rampant all over the world, causing incalculable losses to the world. Major pharmaceutical organizations around the globe are focusing on vaccine research and drug development to prevent further damage caused by the pandemic. The messenger RNA (mRNA) technology has got ample of attention after the success of the two very effective mRNA vaccines during the recent pandemic of COVID-19. mRNA vaccine has been promoted to the core stage of pharmaceutical industry, and the rapid development of mRNA technology has exceeded expectations. Beyond COVID-19, the mRNA vaccine has been tested for various infectious diseases and undergoing clinical trials. Due to the ability of constant mutation, the viral infections demand abrupt responses and immediate production, and therefore mRNA-based technology offers best answers to sudden outbreaks. The need for mRNA-based vaccine became more obvious due to the recent emergence of new Omicron variant. In this review, we summarized the unique properties of mRNA-based vaccines for infectious diseases, delivery technologies, discussed current challenges, and highlighted the prospects of this promising technology in the future. We also discussed various clinical studies as well preclinical studies conducted on mRNA therapeutics for diverse infectious diseases.
Collapse
Affiliation(s)
- Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
| | - Haiyin Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry and Environmental Science, Hebei University, Baoding, 071002 China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
- School of Materials and the Environment, Beijing Institute of Technology, Zhuhai, 519085 China
| |
Collapse
|
6
|
Ripoll M, Bernard MC, Vaure C, Bazin E, Commandeur S, Perkov V, Lemdani K, Nicolaï MC, Bonifassi P, Kichler A, Frisch B, Haensler J. An imidazole modified lipid confers enhanced mRNA-LNP stability and strong immunization properties in mice and non-human primates. Biomaterials 2022; 286:121570. [PMID: 35576809 PMCID: PMC9078044 DOI: 10.1016/j.biomaterials.2022.121570] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022]
Abstract
The mRNA vaccine technology has promising applications to fight infectious diseases as demonstrated by the licensing of two mRNA-based vaccines, Comirnaty® (Pfizer/BioNtech) and Spikevax® (Moderna), in the context of the Covid-19 crisis. Safe and effective delivery systems are essential to the performance of these vaccines and lipid nanoparticles (LNPs) able to entrap, protect and deliver the mRNA in vivo are considered by many as the current "best in class". Nevertheless, current mRNA/LNP vaccine technology has still some limitations, one of them being thermostability, as evidenced by the ultracold distribution chain required for the licensed vaccines. We found that the thermostability of mRNA/LNP, could be improved by a novel imidazole modified lipid, DOG-IM4, in combination with standard helper lipids. DOG-IM4 comprises an ionizable head group consisting of imidazole, a dioleoyl lipid tail and a short flexible polyoxyethylene spacer between the head and tail. Here we describe the synthesis of DOG-IM4 and show that DOG-IM4 LNPs confer strong immunization properties to influenza HA mRNA in mice and macaques and a remarkable stability to the encapsulated mRNA when stored liquid in phosphate buffered saline at 4 °C. We speculate the increased stability to result from some specific attributes of the lipid's imidazole head group.
Collapse
Affiliation(s)
- Manon Ripoll
- Sanofi R&D, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280, Marcy l'Etoile, France; Laboratoire de Conception et Application de Molécules Bioactives, Equipe 3Bio (Biovectorisation, Bioconjugaison, Biomatériaux), UMR 7199 - CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, BP 60024, 67401, Illkirch Cedex, France.
| | | | - Céline Vaure
- Sanofi R&D, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280, Marcy l'Etoile, France.
| | - Emilie Bazin
- Sanofi R&D, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280, Marcy l'Etoile, France.
| | - Sylvie Commandeur
- Sanofi R&D, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280, Marcy l'Etoile, France.
| | - Vladimir Perkov
- Sanofi R&D, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280, Marcy l'Etoile, France.
| | - Katia Lemdani
- Sanofi R&D, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280, Marcy l'Etoile, France; Neovacs, 3 impasse Reille, 75014 Paris, France.
| | - Marie-Claire Nicolaï
- Sanofi R&D, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280, Marcy l'Etoile, France.
| | - Patrick Bonifassi
- Sanofi R&D, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280, Marcy l'Etoile, France.
| | - Antoine Kichler
- Laboratoire de Conception et Application de Molécules Bioactives, Equipe 3Bio (Biovectorisation, Bioconjugaison, Biomatériaux), UMR 7199 - CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, BP 60024, 67401, Illkirch Cedex, France.
| | - Benoit Frisch
- Laboratoire de Conception et Application de Molécules Bioactives, Equipe 3Bio (Biovectorisation, Bioconjugaison, Biomatériaux), UMR 7199 - CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, BP 60024, 67401, Illkirch Cedex, France.
| | - Jean Haensler
- Sanofi R&D, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280, Marcy l'Etoile, France.
| |
Collapse
|
7
|
Hu A, Zhu J, Zeng C, Lin CH, Yu J, Liu JQ, Lynch K, Talebian F, Pan X, Yan J, Dong Y, Li Z, Bai XF. IL-27 Induces CCL5 Production by T Lymphocytes, Which Contributes to Antitumor Activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2239-2245. [PMID: 35418466 PMCID: PMC9050872 DOI: 10.4049/jimmunol.2100885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/25/2022] [Indexed: 04/15/2023]
Abstract
IL-27 is a pleiotropic cytokine that exhibits stimulatory/regulatory functions on multiple lineages of immune cells including T lymphocytes. In this study, we demonstrate that IL-27 directly induces CCL5 production by T lymphocytes, particularly CD8+ T cells in vitro and in vivo. IL-27-induced CCL5 production is IL-27R-dependent. In CD4+ T cells, IL-27-induced CCL5 production was primarily dependent on Stat1 activation, whereas in CD8+ T cells, Stat1 deficiency does not abrogate CCL5 induction. A chromatin immunoprecipitation assay revealed that in the CCL5 promoter region, both putative Stat3 binding sites exhibit significant binding to Stat3, whereas only one out of four Stat1 binding sites displays moderate binding to Stat1. In tumor-bearing mice, IL-27 induced dramatic production of CCL5 in tumor-infiltrating T cells. IL-27-induced CCL5 appears to contribute to an IL-27-mediated antitumor effect. This is signified by diminished tumor inhibition in anti-CCL5- and IL-27-treated mice. Additionally, intratumor delivery of CCL5 mRNA using lipid nanoparticles significantly inhibited tumor growth. Thus, IL-27 induces robust CCL5 production by T cells, which contributes to antitumor activity.
Collapse
Affiliation(s)
- Aiyan Hu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Jianmin Zhu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Chunxi Zeng
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Cho-Hao Lin
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Jianyu Yu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Jin-Qing Liu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Kimberly Lynch
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Fatemeh Talebian
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH
| | - Xueliang Pan
- Center for Biostatistics, College of Medicine, The Ohio State University, Columbus, OH
| | - Jingyue Yan
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH; and
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH; and
| | - Zihai Li
- Institute for Immuno-Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Xue-Feng Bai
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH;
- Institute for Immuno-Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| |
Collapse
|
8
|
Liu JQ, Zhang C, Zhang X, Yan J, Zeng C, Talebian F, Lynch K, Zhao W, Hou X, Du S, Kang DD, Deng B, McComb DW, Bai XF, Dong Y. Intratumoral delivery of IL-12 and IL-27 mRNA using lipid nanoparticles for cancer immunotherapy. J Control Release 2022; 345:306-313. [PMID: 35301053 PMCID: PMC9133152 DOI: 10.1016/j.jconrel.2022.03.021] [Citation(s) in RCA: 106] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 02/05/2022] [Accepted: 03/10/2022] [Indexed: 11/21/2022]
Abstract
Cytokines are important immunotherapeutics with approved drugs for the treatment of human cancers. However, systemic administration of cytokines often fails to achieve adequate concentrations to immune cells in tumors due to dose-limiting toxicity. Thus, developing localized therapy that directly delivers immune-stimulatory cytokines to tumors may improve the therapeutic efficacy. In this study, we generated novel lipid nanoparticles (LNPs) encapsulated with mRNAs encoding cytokines including IL-12, IL-27 and GM-CSF, and tested their anti-tumor activity. We first synthesized ionizable lipid materials containing di-amino groups with various head groups (DALs). The novel DAL4-LNP effectively delivered different mRNAs in vitro to tumor cells and in vivo to tumors. Intratumoral injection of DAL4-LNP loaded with IL-12 mRNA was most potent in inhibiting B16F10 melanoma tumor growth compared to IL-27 or GM-CSF mRNAs in monotherapy. Furthermore, intratumoral injection of dual DAL4-LNP-IL-12 mRNA and IL-27 mRNA showed a synergistic effect in suppressing tumor growth without causing systematic toxicity. Most importantly, intratumoral delivery of IL-12 and IL-27 mRNAs induced robust infiltration of immune effector cells, including IFN-γ and TNF-α producing NK and CD8+ T cells into tumors. Thus, intratumoral administration of DAL-LNP loaded with IL-12 and IL-27 mRNA provides a new treatment strategy for cancer.
Collapse
Affiliation(s)
- Jin-Qing Liu
- Department of Pathology, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Chengxiang Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Xinfu Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Jingyue Yan
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Chunxi Zeng
- Department of Pathology, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States; Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Fatemeh Talebian
- Department of Pathology, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Kimberly Lynch
- Department of Pathology, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Weiyu Zhao
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Xucheng Hou
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Shi Du
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Diana D Kang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Binbin Deng
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, United States
| | - David W McComb
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, United States; Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States
| | - Xue-Feng Bai
- Department of Pathology, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States; Department of Radiation Oncology, Department of Biomedical Engineering, The Center for Clinical and Translational Science, The Comprehensive Cancer Center, Dorothy M. Davis Heart & Lung Research Institute, Center for Cancer Engineering, Center for Cancer Metabolism, Pelotonia Institute for Immune-Oncology, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
9
|
Hauck ES, Hecker JG. Non-Viral Delivery of RNA Gene Therapy to the Central Nervous System. Pharmaceutics 2022; 14:165. [PMID: 35057059 PMCID: PMC8779867 DOI: 10.3390/pharmaceutics14010165] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/01/2022] [Accepted: 01/05/2022] [Indexed: 02/05/2023] Open
Abstract
Appropriate gene delivery systems are essential for successful gene therapy in clinical medicine. Lipid-mediated nucleic acid delivery is an alternative to viral vector-mediated gene delivery and has the following advantages. Lipid-mediated delivery of DNA or mRNA is usually more rapid than viral-mediated delivery, offers a larger payload, and has a nearly zero risk of incorporation. Lipid-mediated delivery of DNA or RNA is therefore preferable to viral DNA delivery in those clinical applications that do not require long-term expression for chronic conditions. Delivery of RNA may be preferable to non-viral DNA delivery in some clinical applications, since transit across the nuclear membrane is not necessary, and onset of expression with RNA is therefore even faster than with DNA, although both are faster than most viral vectors. Delivery of RNA to target organ(s) has previously been challenging due to RNA's rapid degradation in biological systems, but cationic lipids complexed with RNA, as well as lipid nanoparticles (LNPs), have allowed for delivery and expression of the complexed RNA both in vitro and in vivo. This review will focus on the non-viral lipid-mediated delivery of RNAs, including mRNA, siRNA, shRNA, and microRNA, to the central nervous system (CNS), an organ with at least two unique challenges. The CNS contains a large number of slowly dividing or non-dividing cell types and is protected by the blood brain barrier (BBB). In non-dividing cells, RNA-lipid complexes demonstrated increased transfection efficiency relative to DNA transfection. The efficiency, timing of the onset, and duration of expression after transfection may determine which nucleic acid is best for which proposed therapy. Expression can be seen as soon as 1 h after RNA delivery, but duration of expression has been limited to 5-7 h. In contrast, transfection with a DNA lipoplex demonstrates protein expression within 5 h and lasts as long as several weeks after transfection.
Collapse
Affiliation(s)
- Ellen S. Hauck
- Department of Anesthesiology, Lewis Katz School of Medicine, Temple University, 3401 N Broad St., Philadelphia, PA 19140, USA
| | - James G. Hecker
- Department of Anesthesiology, Harborview Medical Center, University of Washington, P.O. Box 359724, 329 Ninth Ave, Seattle, WA 98104, USA;
| |
Collapse
|
10
|
Abstract
RNA-based therapeutics have shown great promise in treating a broad spectrum of diseases through various mechanisms including knockdown of pathological genes, expression of therapeutic proteins, and programmed gene editing. Due to the inherent instability and negative-charges of RNA molecules, RNA-based therapeutics can make the most use of delivery systems to overcome biological barriers and to release the RNA payload into the cytosol. Among different types of delivery systems, lipid-based RNA delivery systems, particularly lipid nanoparticles (LNPs), have been extensively studied due to their unique properties, such as simple chemical synthesis of lipid components, scalable manufacturing processes of LNPs, and wide packaging capability. LNPs represent the most widely used delivery systems for RNA-based therapeutics, as evidenced by the clinical approvals of three LNP-RNA formulations, patisiran, BNT162b2, and mRNA-1273. This review covers recent advances of lipids, lipid derivatives, and lipid-derived macromolecules used in RNA delivery over the past several decades. We focus mainly on their chemical structures, synthetic routes, characterization, formulation methods, and structure-activity relationships. We also briefly describe the current status of representative preclinical studies and clinical trials and highlight future opportunities and challenges.
Collapse
Affiliation(s)
- Yuebao Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Changzhen Sun
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Chang Wang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Katarina E Jankovic
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Department of Biomedical Engineering, The Center for Clinical and Translational Science, The Comprehensive Cancer Center, Dorothy M. Davis Heart & Lung Research Institute, Department of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
11
|
mRNA delivery via non-viral carriers for biomedical applications. Int J Pharm 2021; 607:121020. [PMID: 34416327 DOI: 10.1016/j.ijpharm.2021.121020] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/03/2021] [Accepted: 08/14/2021] [Indexed: 12/11/2022]
Abstract
As an emerging new class of nucleic acid drugs, messenger RNA (mRNA) has huge potential in immunotherapy, regenerative medicine, vaccine, and gene editing. Comparing with siRNA and pDNA, mRNA is more vulnerable to nucleases in vivo. However, the lack of effective and safe delivery methods impedes the broad application of mRNA-based therapeutics. Up to now, the delivery of mRNA remains largely unexplored, and therefore, is a hot topic in the field of gene therapy. In this review, we will summarize the ongoing challenges in mRNA-based therapeutics and unmet requirements for delivery vehicles in terms of the unique structure of mRNA. We then highlight the advancement in mRNA delivery in both fundamental research and clinical applications. Finally, a prospective will be proposed upon reviewing the current progress in mRNA delivery.
Collapse
|
12
|
Li M, Li S, Huang Y, Chen H, Zhang S, Zhang Z, Wu W, Zeng X, Zhou B, Li B. Secreted Expression of mRNA-Encoded Truncated ACE2 Variants for SARS-CoV-2 via Lipid-Like Nanoassemblies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2101707. [PMID: 34278613 PMCID: PMC8420471 DOI: 10.1002/adma.202101707] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/23/2021] [Indexed: 05/24/2023]
Abstract
The transfer of foreign synthetic messenger RNA (mRNA) into cells is essential for mRNA-based protein-replacement therapies. Prophylactic mRNA COVID-19 vaccines commonly utilize nanotechnology to deliver mRNA encoding SARS-CoV-2 vaccine antigens, thereby triggering the body's immune response and preventing infections. In this study, a new combinatorial library of symmetric lipid-like compounds is constructed, and among which a lead compound is selected to prepare lipid-like nanoassemblies (LLNs) for intracellular delivery of mRNA. After multiround optimization, the mRNA formulated into core-shell-structured LLNs exhibits more than three orders of magnitude higher resistance to serum than the unprotected mRNA, and leads to sustained and high-level protein expression in mammalian cells. A single intravenous injection of LLNs into mice achieves over 95% mRNA translation in the spleen, without causing significant hematological and histological changes. Delivery of in-vitro-transcribed mRNA that encodes high-affinity truncated ACE2 variants (tACE2v mRNA) through LLNs induces elevated expression and secretion of tACE2v decoys, which is able to effectively block the binding of the receptor-binding domain of the SARS-CoV-2 to the human ACE2 receptor. The robust neutralization activity in vitro suggests that intracellular delivery of mRNA encoding ACE2 receptor mimics via LLNs may represent a potential intervention strategy for COVID-19.
Collapse
Affiliation(s)
- Min Li
- Department of Infectious DiseaseShenzhen People's HospitalThe First Affiliated Hospital of Southern University of Science and TechnologyShenzhen518020China
| | - Sanpeng Li
- Department of Infectious DiseaseShenzhen People's HospitalThe First Affiliated Hospital of Southern University of Science and TechnologyShenzhen518020China
| | - Yixuan Huang
- Department of Infectious DiseaseShenzhen People's HospitalThe First Affiliated Hospital of Southern University of Science and TechnologyShenzhen518020China
| | - Haixia Chen
- Department of Clinical LaboratoryThe Second Clinical Medical College of Jinan UniversityShenzhen518020China
| | - Songya Zhang
- Shenzhen Institute of Synthetic BiologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055China
| | - Zhicheng Zhang
- Department of Infectious DiseaseShenzhen People's HospitalThe First Affiliated Hospital of Southern University of Science and TechnologyShenzhen518020China
| | - Weigang Wu
- Department of Infectious DiseaseShenzhen People's HospitalThe First Affiliated Hospital of Southern University of Science and TechnologyShenzhen518020China
| | - Xiaobin Zeng
- Department of Infectious DiseaseShenzhen People's HospitalThe First Affiliated Hospital of Southern University of Science and TechnologyShenzhen518020China
| | - Boping Zhou
- Department of Infectious DiseaseShenzhen People's HospitalThe First Affiliated Hospital of Southern University of Science and TechnologyShenzhen518020China
| | - Bin Li
- Department of Infectious DiseaseShenzhen People's HospitalThe First Affiliated Hospital of Southern University of Science and TechnologyShenzhen518020China
- Department of Infectious DiseaseThe Second Clinical Medical College of Jinan UniversityShenzhen518020China
| |
Collapse
|
13
|
Meyer T, Rabeah J, Brückner A, Wu XF. Visible-Light-Induced Palladium-Catalyzed Dehydrogenative Carbonylation of Amines to Oxalamides. Chemistry 2021; 27:5642-5647. [PMID: 33565685 DOI: 10.1002/chem.202100009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/08/2021] [Indexed: 12/23/2022]
Abstract
The palladium-catalyzed oxidative carbonylation of amines toward the synthesis of oxalamides has been established around 30 years ago and it usually needs the presence of (over)stoichiometric amounts of oxidant. In this work, the first transformation of this type in which the oxidant was replaced by visible light is described. The new approach uses a simple robust Pd complex, which can even be partially recycled. A mechanistic reason is provided and supported by control experiments and EPR studies, showing that PdI was formed and Pd0 was the active species. Both nitrogen- and the intermediate acyl radical can be detected. Moreover, the formation of hydrogen was confirmed by gas GC.
Collapse
Affiliation(s)
- Tim Meyer
- Leibniz-Institut für Katalyse e.V., Universität Rostock, Albert-Einstein-Straße 29a, 18059, Rostock, Germany
| | - Jabor Rabeah
- Leibniz-Institut für Katalyse e.V., Universität Rostock, Albert-Einstein-Straße 29a, 18059, Rostock, Germany
| | - Angelika Brückner
- Leibniz-Institut für Katalyse e.V., Universität Rostock, Albert-Einstein-Straße 29a, 18059, Rostock, Germany
| | - Xiao-Feng Wu
- Leibniz-Institut für Katalyse e.V., Universität Rostock, Albert-Einstein-Straße 29a, 18059, Rostock, Germany.,Dalian National Laboratory for Clean Energy, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, Liaoning, P. R. China
| |
Collapse
|
14
|
Zhao X, Glass Z, Chen J, Yang L, Kaplan DL, Xu Q. mRNA Delivery Using Bioreducible Lipidoid Nanoparticles Facilitates Neural Differentiation of Human Mesenchymal Stem Cells. Adv Healthc Mater 2021; 10:e2000938. [PMID: 32815325 DOI: 10.1002/adhm.202000938] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/12/2020] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cells (MSCs) are widely used in regenerative medicine and tissue engineering and delivering biological molecules into MSCs has been used to control stem cell behavior. However, the efficient delivery of large biomolecules such as DNA, RNA, and proteins into MSCs using nonviral delivery strategies remains an ongoing challenge. Herein, nanoparticles composed of cationic bioreducible lipid-like materials (lipidoids) are developed to intracellularly deliver mRNA into human mesenchymal stem cells (hMSCs). The delivery efficacy to hMSCs is improved by adding three excipients including cholesterol, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-polyethylene glycol (DSPE-PEG) during lipidoid nanoparticle formulation. Using an optimized lipidoid formulation, Cas9 mRNA and single guide RNA (sgRNA) targeting neuron restrictive silencing factor (NRSF) are delivered to hMSCs, leading to successful neural-like differentiation as demonstrated by the expression of synaptophysin (SYP), brain-derived neurotrophic factor (BDNF), neuron-specific enolase (NSE), and neuron-specific growth-associated protein (SCG10). Overall, a synthetic lipid formulation that can efficiently deliver mRNA to hMSCs is identified, leading to CRISPR-based gene knockdown to facilitate hMSCs transdifferentiation into neural-like lineage.
Collapse
Affiliation(s)
- Xuewei Zhao
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Zachary Glass
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Jinjin Chen
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Liu Yang
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - David L. Kaplan
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Qiaobing Xu
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| |
Collapse
|
15
|
Weng Y, Li C, Yang T, Hu B, Zhang M, Guo S, Xiao H, Liang XJ, Huang Y. The challenge and prospect of mRNA therapeutics landscape. Biotechnol Adv 2020; 40:107534. [PMID: 32088327 DOI: 10.1016/j.biotechadv.2020.107534] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 02/05/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022]
Abstract
Messenger RNA (mRNA)-based therapeutics hold the potential to cause a major revolution in the pharmaceutical industry because they can be used for precise and individualized therapy, and enable patients to produce therapeutic proteins in their own bodies without struggling with the comprehensive manufacturing issues associated with recombinant proteins. Compared with the current therapeutics, the production of mRNA is much cost-effective, faster and more flexible because it can be easily produced by in vitro transcription, and the process is independent of mRNA sequence. Moreover, mRNA vaccines allow people to develop personalized medications based on sequencing results and/or personalized conditions rapidly. Along with the great potential from bench to bedside, technical obstacles facing mRNA pharmaceuticals are also obvious. The stability, immunogenicity, translation efficiency, and delivery are all pivotal issues need to be addressed. In the recently published research results, these issues are gradually being overcome by state-of-the-art development technologies. In this review, we describe the structural properties and modification technologies of mRNA, summarize the latest advances in developing mRNA delivery systems, review the preclinical and clinical applications, and put forward our views on the prospect and challenges of developing mRNA into a new class of drug.
Collapse
Affiliation(s)
- Yuhua Weng
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Chunhui Li
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Tongren Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Bo Hu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Shuai Guo
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, PR China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, PR China.
| |
Collapse
|
16
|
Peng Y, Bariwal J, Kumar V, Tan C, Mahato RI. Organic Nanocarriers for Delivery and Targeting of Therapeutic Agents for Cancer Treatment. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900136] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yang Peng
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Jitender Bariwal
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Virender Kumar
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug DeliveryUniversity of Mississippi University MS 38677 USA
| | - Ram I. Mahato
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| |
Collapse
|
17
|
Guimaraes PPG, Zhang R, Spektor R, Tan M, Chung A, Billingsley MM, El-Mayta R, Riley RS, Wang L, Wilson JM, Mitchell MJ. Ionizable lipid nanoparticles encapsulating barcoded mRNA for accelerated in vivo delivery screening. J Control Release 2019; 316:404-417. [PMID: 31678653 PMCID: PMC7032071 DOI: 10.1016/j.jconrel.2019.10.028] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/08/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022]
Abstract
Messenger RNA (mRNA) has recently emerged as a promising class of nucleic acid therapy, with the potential to induce protein production to treat and prevent a range of diseases. However, the widespread use of mRNA as a therapeutic requires safe and effective in vivo delivery technologies. Libraries of ionizable lipid nanoparticles (LNPs) have been designed to encapsulate mRNA, prevent its degradation, and mediate intracellular delivery. However, these LNPs are typically characterized and screened in an in vitro setting, which may not fully replicate the biological barriers that they encounter in vivo. Here, we designed and evaluated a library of engineered LNPs containing barcoded mRNA (b-mRNA) to accelerate the screening of mRNA delivery platforms in vivo. These b-mRNA are similar in structure and function to regular mRNA, and contain barcodes that enable their delivery to be quantified via deep sequencing. Using a mini-library of b-mRNA LNPs formulated via microfluidic mixing, we show that these different formulations can be pooled together, administered intravenously into mice as a single pool, and their delivery to multiple organs (liver, spleen, brain, lung, heart, kidney, pancreas, and muscle) can be quantified simultaneously using deep sequencing. In the context of liver and spleen delivery, LNPs that exhibited high b-mRNA delivery also yielded high luciferase expression, indicating that this platform can identify lead LNP candidates as well as optimal formulation parameters for in vivo mRNA delivery. Interestingly, LNPs with identical formulation parameters that encapsulated different types of nucleic acid barcodes (b-mRNA versus a DNA barcode) altered in vivo delivery, suggesting that the structure of the barcoded nucleic acid affects LNP in vivo delivery. This platform, which enables direct barcoding and subsequent quantification of a functional mRNA, can accelerate the in vivo screening and design of LNPs for mRNA therapeutic applications such as CRISPR-Cas9 gene editing, mRNA vaccination, and other mRNA-based regenerative medicine and protein replacement therapies.
Collapse
Affiliation(s)
- Pedro P G Guimaraes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States; Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rui Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Roman Spektor
- Graduate Field of Genetics, Genomics and Development, Cornell University, Ithaca, NY, United States
| | - Mingchee Tan
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Amanda Chung
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Margaret M Billingsley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Rachel S Riley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Lili Wang
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
18
|
Zhang C, Zhang X, Zhao W, Zeng C, Li W, Li B, Luo X, Li J, Jiang J, Deng B, McComb DW, Dong Y. Chemotherapy drugs derived nanoparticles encapsulating mRNA encoding tumor suppressor proteins to treat triple-negative breast cancer. NANO RESEARCH 2019; 12:855-861. [PMID: 31737223 PMCID: PMC6858063 DOI: 10.1007/s12274-019-2308-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/17/2019] [Accepted: 01/20/2019] [Indexed: 05/19/2023]
Abstract
Triple-negative breast cancer (TNBC) is one type of the most aggressive breast cancers with poor prognosis. It is of great urgency to develop new therapeutics for treating TNBC. Based on current treatment guideline and genetic information of TNBC, a combinational therapy platform integrating chemotherapy drugs and mRNA encoding tumor suppressor proteins may become an efficacious strategy. In this study, we developed paclitaxel amino lipid (PAL) derived nanoparticles (NPs) to incorporate both chemotherapy drugs and P53 mRNA. The PAL P53 mRNA NPs showed superior properties compared to Abraxane® and Lipusu® used in the clinic including high paclitaxel loading capacity (24 wt.%, calculated by paclitaxel in PAL), PAL encapsulation efficiency (94.7% ± 6.8%) and mRNA encapsulation efficiency (88.7% ± 0.7%). Meanwhile, these NPs displayed synergetic cytotoxicity of paclitaxel and P53 mRNA in cultured TNBC cells. More importantly, we demonstrated in vivo anti-tumor efficacy of PAL P53 mRNA NPs in an orthotopic TNBC mouse model. Overall, these chemotherapy drugs derived mRNA NPs provide a new platform to integrate chemotherapy and personalized medicine using tumor genetic information, and therefore represent a promising approach for TNBC treatment.
Collapse
Affiliation(s)
- Chengxiang Zhang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Xinfu Zhang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Weiyu Zhao
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Chunxi Zeng
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Wenqing Li
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Bin Li
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Xiao Luo
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Junan Li
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Justin Jiang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Binbin Deng
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH 43212, USA
| | - David W McComb
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH 43212, USA
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
- The Center for Clinical and Translational Science, The Ohio State University, Columbus, OH 43210, USA
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
19
|
Li Y, Li AC, Xu Q. Intracellular Delivery of His-Tagged Genome-Editing Proteins Enabled by Nitrilotriacetic Acid-Containing Lipidoid Nanoparticles. Adv Healthc Mater 2019; 8:e1800996. [PMID: 30565897 PMCID: PMC6474682 DOI: 10.1002/adhm.201800996] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/26/2018] [Indexed: 12/26/2022]
Abstract
Protein- and peptide-based therapeutics with high tolerance and specificity along with low off-target effects and genetic risks have attracted tremendous attention over the last three decades. Herein, a new type of noncationic lipidoid nanoparticle (LNP) is reported for His-tagged protein delivery. Active lipidoids are synthesized by conjugating a nitrilotriacetic acid group with hydrophobic tails (EC16, O16B, and O17O) and nanoparticles are formulated in the presence of nickel ions and helper lipids (cholesterol, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine, and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000]). It is demonstrated that the newly developed LNPs are capable of delivering various His-tagged proteins including green fluorescent protein (GFP), (-30)GFP-Cre recombinase, and CRISPR/Cas9 ribonucleoprotein into mammalian cells.
Collapse
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, US,
| | - Alice Chukun Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, US,
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, US,
| |
Collapse
|
20
|
Li B, Zhang X, Dong Y. Nanoscale platforms for messenger RNA delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1530. [PMID: 29726120 PMCID: PMC6443240 DOI: 10.1002/wnan.1530] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/27/2018] [Accepted: 04/01/2018] [Indexed: 12/27/2022]
Abstract
Messenger RNA (mRNA) has become a promising class of drugs for diverse therapeutic applications in the past few years. A series of clinical trials are ongoing or will be initiated in the near future for the treatment of a variety of diseases. Currently, mRNA-based therapeutics mainly focuses on ex vivo transfection and local administration in clinical studies. Efficient and safe delivery of therapeutically relevant mRNAs remains one of the major challenges for their broad applications in humans. Thus, effective delivery systems are urgently needed to overcome this limitation. In recent years, numerous nanoscale biomaterials have been constructed for mRNA delivery in order to protect mRNA from extracellular degradation and facilitate endosomal escape after cellular uptake. Nanoscale platforms have expanded the feasibility of mRNA-based therapeutics, and enabled its potential applications to protein replacement therapy, cancer immunotherapy, therapeutic vaccines, regenerative medicine, and genome editing. This review focuses on recent advances, challenges, and future directions in nanoscale platforms designed for mRNA delivery, including lipid and lipid-derived nanoparticles, polymer-based nanoparticles, protein derivatives mRNA complexes, and other types of nanomaterials. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Biology-Inspired Nanomaterials > Lipid-Based Structures Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures.
Collapse
Affiliation(s)
- Bin Li
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Xinfu Zhang
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Yizhou Dong
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The Center for Clinical and Translational Science, The Ohio State University, Columbus, Ohio
- James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
21
|
Li Y, Bolinger J, Yu Y, Glass Z, Shi N, Yang L, Wang M, Xu Q. Intracellular delivery and biodistribution study of CRISPR/Cas9 ribonucleoprotein loaded bioreducible lipidoid nanoparticles. Biomater Sci 2019; 7:596-606. [DOI: 10.1039/c8bm00637g] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A combinatorial library of cationic lipidoids were used as nanocarriers for intracellular delivery of the CRISPR/Cas9 ribonucleoprotein complex.
Collapse
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Justin Bolinger
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Yingjie Yu
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Zachary Glass
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Nicola Shi
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Liu Yang
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Ming Wang
- Beijing National Laboratory for Molecular Sciences
- Key Laboratory of Analytical Chemistry for Living Biosystems
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing
| | - Qiaobing Xu
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| |
Collapse
|
22
|
Cheng Q, Wei T, Jia Y, Farbiak L, Zhou K, Zhang S, Wei Y, Zhu H, Siegwart DJ. Dendrimer-Based Lipid Nanoparticles Deliver Therapeutic FAH mRNA to Normalize Liver Function and Extend Survival in a Mouse Model of Hepatorenal Tyrosinemia Type I. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1805308. [PMID: 30368954 DOI: 10.1002/adma.201805308] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/14/2018] [Indexed: 06/08/2023]
Abstract
mRNA-mediated protein replacement represents a promising concept for the treatment of liver disorders. Children born with fumarylacetoacetate hydrolase (FAH) mutations suffer from Hepatorenal Tyrosinemia Type 1 (HT-1) resulting in renal dysfunction, liver failure, neurological impairments, and cancer. Protein replacement therapy using FAH mRNA offers tremendous potential to cure HT-1, but is currently hindered by the development of effective mRNA carriers that can function in diseased livers. Structure-guided, rational optimization of 5A2-SC8 mRNA-loaded dendrimer lipid nanoparticles (mDLNPs) increases delivery potency of FAH mRNA, resulting in functional FAH protein and sustained normalization of body weight and liver function in FAH-/- knockout mice. Optimization using luciferase mRNA produces DLNP carriers that are efficacious at mRNA doses as low as 0.05 mg kg-1 in vivo. mDLNPs transfect > 44% of all hepatocytes in the liver, yield high FAH protein levels (0.5 mg kg-1 mRNA), and are well tolerated in a knockout mouse model with compromised liver function. Genetically engineered FAH-/- mice treated with FAH mRNA mDLNPs have statistically equivalent levels of TBIL, ALT, and AST compared to wild type C57BL/6 mice and maintain normal weight throughout the month-long course of treatment. This study provides a framework for the rational optimization of LNPs to improve delivery of mRNA broadly and introduces a specific and viable DLNP carrier with translational potential to treat genetic diseases of the liver.
Collapse
Affiliation(s)
- Qiang Cheng
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tuo Wei
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yuemeng Jia
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lukas Farbiak
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kejin Zhou
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Shuyuan Zhang
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yonglong Wei
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hao Zhu
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Daniel J Siegwart
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
23
|
Givens BE, Naguib YW, Geary SM, Devor EJ, Salem AK. Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing Therapeutics. AAPS J 2018; 20:108. [PMID: 30306365 PMCID: PMC6398936 DOI: 10.1208/s12248-018-0267-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/18/2018] [Indexed: 12/17/2022] Open
Abstract
The recent progress in harnessing the efficient and precise method of DNA editing provided by CRISPR/Cas9 is one of the most promising major advances in the field of gene therapy. However, the development of safe and optimally efficient delivery systems for CRISPR/Cas9 elements capable of achieving specific targeting of gene therapy to the location of interest without off-target effects is a primary challenge for clinical therapeutics. Nanoparticles (NPs) provide a promising means to meet such challenges. In this review, we present the most recent advances in developing innovative NP-based delivery systems that efficiently deliver CRISPR/Cas9 constructs and maximize their effectiveness.
Collapse
Affiliation(s)
- Brittany E Givens
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
- Department of Chemical and Biochemical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Youssef W Naguib
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Sean M Geary
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Eric J Devor
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA.
- Department of Chemical and Biochemical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
24
|
Luo X, Zhao W, Li B, Zhang X, Zhang C, Bratasz A, Deng B, McComb DW, Dong Y. Co-delivery of mRNA and SPIONs through amino-ester nanomaterials. NANO RESEARCH 2018; 11:5596-5603. [PMID: 31737222 PMCID: PMC6858065 DOI: 10.1007/s12274-018-2082-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/21/2018] [Accepted: 04/26/2018] [Indexed: 05/27/2023]
Abstract
Nanoparticles have been widely explored for combined therapeutic and diagnostic applications. For example, lipid-based nanoparticles have been used to encapsulate multiple types of agents and achieve multi-functions. Herein, we enabled a co-delivery of mRNA molecules and superparamagnetic iron oxide nanoparticles (SPIONs) by using an amino-ester lipid-like nanomaterial. An orthogonal experimental design was used to identify the optimal formulation. The optimal formulation, MPA-Ab-8 LLNs, not only showed high encapsulation of both mRNA and SPIONs, but also increased the r 2 relaxivity of SPIONs by more than 1.5-fold in vitro. MPA-Ab-8 LLNs effectively delivered mRNA and SPIONs into cells, and consequently induced high protein expression as well as strong MRI contrast. Consistent herewith, we observed both mRNA-mediated protein expression and an evident negative contrast enhancement of MRI signal in mice. In conclusion, amino-ester nanomaterials demonstrate great potential as delivery vehicles for theranostic applications.
Collapse
Affiliation(s)
- Xiao Luo
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA
| | - Weiyu Zhao
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA
| | - Bin Li
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA
| | - Xinfu Zhang
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA
| | - Chengxiang Zhang
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA
| | - Anna Bratasz
- Small Animal Imaging Core, The Ohio State University, Columbus, Ohio 43210, USA
| | - Binbin Deng
- Center for Electron Microscopy and Analysis, Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - David W McComb
- Center for Electron Microscopy and Analysis, Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Yizhou Dong
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
- The Center for Clinical and Translational Science, The Ohio State University, Columbus, Ohio 43210, USA
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, USA
| |
Collapse
|
25
|
Jiang Y, Gaudin A, Zhang J, Agarwal T, Song E, Kauffman AC, Tietjen GT, Wang Y, Jiang Z, Cheng CJ, Saltzman WM. A "top-down" approach to actuate poly(amine-co-ester) terpolymers for potent and safe mRNA delivery. Biomaterials 2018; 176:122-130. [PMID: 29879653 PMCID: PMC6038928 DOI: 10.1016/j.biomaterials.2018.05.043] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/26/2018] [Accepted: 05/25/2018] [Indexed: 01/05/2023]
Abstract
Gene delivery is known to be a complicated multi-step biological process. It has been observed that subtle differences in the structure and properties of polymeric materials used for gene delivery can lead to dramatic differences in transfection efficiency. Therefore, screening of properties is pivotal to optimizing the polymer. So far, most polymeric materials are built in a "bottom-up" manner, i.e. synthesized from monomers that allow modification of polymer composition or structural factors. With this method, we previously synthesized and screened a library of biodegradable poly(amine-co-ester) (PACE) terpolymers for optimized DNA delivery. However, it can be tedious and time consuming to synthesize a polymer library for screening, particularly when small changes of a factor need to be tested, when multiple factors are involved, and when the effects of different factors are synergistic. In the present work, we evaluate the potential of PACE to deliver mRNA. After observing that mRNA transfection efficiency was highly dependent on both end group composition and molecular weight (MW) of PACE in a synergistic manner, we developed a "top-down" process we called actuation, to simultaneously vary these two factors. Some of the actuated PACE (aPACE) materials presented superior mRNA delivery properties compared to regular PACE, with up to a 106-fold-increase in mRNA transfection efficiency in vitro. Moreover, when aPACE was used to deliver mRNA coding for erythropoietin (EPO) in vivo, it produced high levels of EPO in the blood for up to 48 h without inducing systemic toxicity. This polymer constitutes a new delivery vehicle for mRNA-based treatments that provides safe yet potent protein production.
Collapse
Affiliation(s)
- Yuhang Jiang
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Alice Gaudin
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Junwei Zhang
- Department of Chemical and Environmental Engineering, Yale University, New Haven, CT, 06511, USA
| | - Tushar Agarwal
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Eric Song
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Amy C Kauffman
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Gregory T Tietjen
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Yongheng Wang
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Zhaozhong Jiang
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | | | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA; Department of Chemical and Environmental Engineering, Yale University, New Haven, CT, 06511, USA.
| |
Collapse
|
26
|
McKinlay CJ, Benner NL, Haabeth OA, Waymouth RM, Wender PA. Enhanced mRNA delivery into lymphocytes enabled by lipid-varied libraries of charge-altering releasable transporters. Proc Natl Acad Sci U S A 2018; 115:E5859-E5866. [PMID: 29891683 PMCID: PMC6042134 DOI: 10.1073/pnas.1805358115] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
We report a strategy for generating a combinatorial library of oligonucleotide transporters with varied lipid domains and their use in the efficient transfection of lymphocytes with mRNA in vitro and in vivo. This library is based on amphiphilic charge-altering releasable transporters (CARTs) that contain a lipophilic block functionalized with various side-chain lipids and a polycationic α-amino ester mRNA-binding block that undergoes rearrangement to neutral small molecules, resulting in mRNA release. We show that certain binary mixtures of these lipid-varied CARTs provide up to a ninefold enhancement in mRNA translation in lymphocytes in vitro relative to either a single-lipid CART component alone or the commercial reagent Lipofectamine 2000, corresponding to a striking increase in percent transfection from 9-12% to 80%. Informed by the results with binary mixtures, we further show that CARTs consisting of optimized ratios of the two lead lipids incorporated into a single hybrid-lipid transporter molecule maintain the same delivery efficacy as the noncovalent mixture of two CARTs. The lead lipid CART mixtures and hybrid-lipid CARTs show enhanced lymphocyte transfection in primary T cells and in vivo in mice. This combinatorial approach for rapidly screening mRNA delivery vectors has provided lipid-varied CART mixtures and hybrid-lipid CARTs that exhibit significant improvement in mRNA delivery to lymphocytes, a finding of potentially broad value in research and clinical applications.
Collapse
Affiliation(s)
- Colin J McKinlay
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Nancy L Benner
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Ole A Haabeth
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA 94305
| | | | - Paul A Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305;
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305
| |
Collapse
|
27
|
Abstract
Messenger RNA is emerging as a highly versatile biological construct for creation of impactful medicines. mRNA vaccines directed toward infectious disease and cancer are in clinical development with encouraging early reads on tolerability and efficacy. The use of mRNA to direct intense but transient expression of paracrine factors is finding utility in reprogramming progenitor cells for wound healing and cardiac regeneration and for stimulation of antitumor immune responses, at least preclinically as we await clinical results. The use of mRNA for prolonged and repeated expression of proteins and enzymes to treat rare, typically monogenic disease is nearing clinical entry. These uses of mRNA require delivery solutions, and the application of and improvement to existing nanoparticle nucleic acid delivery systems have jump started the pace of development and reenergized the field of particle based nucleic acid delivery. The current status of mRNA delivery is reviewed in this article with an eye toward clinical tractability.
Collapse
|
28
|
Yan Y, Xiong H, Zhang X, Cheng Q, Siegwart DJ. Systemic mRNA Delivery to the Lungs by Functional Polyester-based Carriers. Biomacromolecules 2017; 18:4307-4315. [DOI: 10.1021/acs.biomac.7b01356] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Yunfeng Yan
- College
of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
- Simmons
Comprehensive Cancer Center, Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Hu Xiong
- Simmons
Comprehensive Cancer Center, Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Xinyi Zhang
- Simmons
Comprehensive Cancer Center, Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Qiang Cheng
- Simmons
Comprehensive Cancer Center, Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Daniel J. Siegwart
- Simmons
Comprehensive Cancer Center, Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| |
Collapse
|
29
|
Li F, Li Y, Zhou Z, Lv S, Deng Q, Xu X, Yin L. Engineering the Aromaticity of Cationic Helical Polypeptides toward "Self-Activated" DNA/siRNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2017; 9:23586-23601. [PMID: 28657294 DOI: 10.1021/acsami.7b08534] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The development of potent yet nontoxic membrane-penetrating materials is in high demand for effective intracellular gene delivery. We have recently developed α-helical polypeptides which afford potent membrane activities to facilitate intracellular DNA delivery via both endocytosis and the nonendocytic "pore formation" mechanism. Endocytosis will cause endosomal entrapment of the DNA cargo, while excessive "pore formation" would cause appreciable cytotoxicity. Additionally, helical polypeptides with stiff, rodlike structure suffer from low siRNA binding affinity. To address such critical issues, we herein incorporated various aromatic domains (benzyl, naphthyl, biphenyl, anthryl, and pyrenyl) into the side-chain terminals of guanidine-rich, helical polypeptides, wherein the flat-rigid shape, π-electronic structures of aromatic motifs "self-activated" the membrane-penetrating capabilities of polypeptides to promote intracellular gene delivery. Benzyl (Bn)- and naphthyl (Naph)-modified polypeptides demonstrated the highest DNA uptake level that outperformed the unmodified polypeptide, P2, by ∼4 fold. More importantly, compared with P2, Bn- and Naph-modified polypeptides allowed more DNA cargos to be internalized via the nonendocytic pathway, which significantly bypassed the endosomal entrapment and accordingly enhanced the transfection efficiency by up to 42 fold, outperforming PEI 25k as the commercial reagent by 3-4 orders of magnitude. The aromatic modification also improved the siRNA condensation capability of polypeptides, achieving notably enhanced gene-silencing efficiency against tumor necrosis factor-α to treat acute hepatic inflammation. Furthermore, we revealed that aromaticity-augmented membrane activity was accompanied by comparable or even significantly reduced "pore formation" capability, thus leading to diminished cytotoxicity at high concentrations. This study therefore provides a promising approach to manipulate the membrane activities and penetration mechanisms of polycations, which overcomes the multiple critical barriers preventing effective and safe gene delivery.
Collapse
Affiliation(s)
- Fangfang Li
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University , Suzhou 215123, China
| | - Yongjuan Li
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University , Suzhou 215123, China
| | - Zhuchao Zhou
- Department of General Surgery, Huashan Hospital, Fudan University , Shanghai 200040, China
| | - Shixian Lv
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University , Suzhou 215123, China
| | - Qiurong Deng
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University , Suzhou 215123, China
| | - Xin Xu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University , Suzhou 215123, China
| | - Lichen Yin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University , Suzhou 215123, China
| |
Collapse
|
30
|
Luo X, Li B, Zhang X, Zhao W, Bratasz A, Deng B, McComb DW, Dong Y. Dual-functional lipid-like nanoparticles for delivery of mRNA and MRI contrast agents. NANOSCALE 2017; 9:1575-1579. [PMID: 28067926 PMCID: PMC5316423 DOI: 10.1039/c6nr08496f] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Multi-functional nanomaterials possess unique properties, facilitating both therapeutic and diagnostic applications among others. Herein, we developed dual-functional lipid-like nanoparticles for simultaneous delivery of mRNA and magnetic resonance imaging (MRI) contrast agents in order to express functional proteins and provide real-time visualization. TT3-Gd18 LLNs were identified as a lead formulation, which was able to encapsulate 91% of mRNA and 74% of Gd. This formulation showed a comparable or a slightly higher delivery efficiency of mRNA compared to the initial TT3 LLNs. Moreover, a strong MRI signal was observed in the cell pellets treated with TT3-Gd18 LLNs. More importantly, TT3-Gd18 LLNs demonstrated an efficient delivery of mRNA and Gd contrast agents in vivo.
Collapse
Affiliation(s)
- X Luo
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA.
| | - B Li
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA.
| | - X Zhang
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA.
| | - W Zhao
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA.
| | - A Bratasz
- Small Animal Imaging Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - B Deng
- Center for Electron Microscopy and Analysis, Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - D W McComb
- Center for Electron Microscopy and Analysis, Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Y Dong
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA.
| |
Collapse
|
31
|
Nanotechnologies in delivery of mRNA therapeutics using nonviral vector-based delivery systems. Gene Ther 2017; 24:133-143. [DOI: 10.1038/gt.2017.5] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 10/28/2016] [Accepted: 01/03/2017] [Indexed: 12/13/2022]
|
32
|
Sergeeva OV, Koteliansky VE, Zatsepin TS. mRNA-Based Therapeutics - Advances and Perspectives. BIOCHEMISTRY (MOSCOW) 2017; 81:709-22. [PMID: 27449617 DOI: 10.1134/s0006297916070075] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In this review we discuss features of mRNA synthesis and modifications used to minimize immune response and prolong efficiency of the translation process in vivo. Considerable attention is given to the use of liposomes and nanoparticles containing lipids and polymers for the mRNA delivery. Finally we briefly discuss mRNAs which are currently in the clinical trials for cancer immunotherapy, vaccination against infectious diseases, and replacement therapy.
Collapse
Affiliation(s)
- O V Sergeeva
- Lomonosov Moscow State University, Department of Chemistry, Moscow, 119991, Russia.
| | | | | |
Collapse
|
33
|
Li B, Dong Y. Preparation and Optimization of Lipid-Like Nanoparticles for mRNA Delivery. Methods Mol Biol 2017; 1632:207-217. [PMID: 28730441 DOI: 10.1007/978-1-4939-7138-1_13] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lipid-like nanoparticles (LLNs) have shown great promise for nucleic acid delivery. Recently, we have developed N 1,N 3,N 5-tris(2-aminoethyl)benzene-1,3,5-tricarboxamide (TT) derived lipid-like compounds, formulated them into TT LLNs for mRNA delivery, and applied an orthogonal array design to facilitate formulation optimization. This chapter focuses on the following contents relevant to lipid-like nanoparticles: formulation method, particle characterization, orthogonal array design, and in vitro assays.
Collapse
Affiliation(s)
- Bin Li
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, 496 W. 12th Ave., Columbus, OH, 43210, USA
| | - Yizhou Dong
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, 496 W. 12th Ave., Columbus, OH, 43210, USA.
| |
Collapse
|
34
|
Stanton MG, Murphy-Benenato KE. Messenger RNA as a Novel Therapeutic Approach. TOPICS IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1007/7355_2016_30] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
35
|
Gustincich S, Zucchelli S, Mallamaci A. The Yin and Yang of nucleic acid-based therapy in the brain. Prog Neurobiol 2016; 155:194-211. [PMID: 27887908 DOI: 10.1016/j.pneurobio.2016.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 11/16/2016] [Accepted: 11/20/2016] [Indexed: 02/06/2023]
Abstract
The post-genomic era has unveiled the existence of a large repertory of non-coding RNAs and repetitive elements that play a fundamental role in cellular homeostasis and dysfunction. These may represent unprecedented opportunities to modify gene expression at the right time in the correct space in vivo, providing an almost unlimited reservoir of new potential pharmacological agents. Hijacking their mode of actions, the druggable genome can be extended to regulatory RNAs and DNA elements in a scalable fashion. Here, we discuss the state-of-the-art of nucleic acid-based drugs to treat neurodegenerative diseases. Beneficial effects can be obtained by inhibiting (Yin) and increasing (Yang) gene expression, depending on the disease and the drug target. Together with the description of the current use of inhibitory RNAs (small inhibitory RNAs and antisense oligonucleotides) in animal models and clinical trials, we discuss the molecular basis and applications of new classes of activatory RNAs at transcriptional (RNAa) and translational (SINEUP) levels.
Collapse
Affiliation(s)
- Stefano Gustincich
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT), Genova, Italy; Area of Neuroscience, SISSA, Trieste, Italy.
| | - Silvia Zucchelli
- Area of Neuroscience, SISSA, Trieste, Italy; Department of Health Sciences, Universita' del Piemonte Orientale, Novara, Italy
| | | |
Collapse
|