1
|
Zubair A, Ahmad H, Arif MM, Ali M. mRNA vaccines against HIV: Hopes and challenges. HIV Med 2025. [PMID: 40195015 DOI: 10.1111/hiv.70024] [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: 10/16/2024] [Accepted: 03/23/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Since the introduction of the first licensed mRNA-based vaccines against COVID-19, there has been significant interest in leveraging this technology for other vaccines. An unprecedented surge of mRNA vaccines has emerged in preclinical, clinical, and various research phases since 2020. The rapid development of mRNA formulations, delivery methods, and manufacturing processes has made this trend foreseeable. There is an urgent demand for effective and easily transportable vaccines in regions where the virus is prevalent, and mRNA technology shows promise in addressing this need. METHODOLOGY The data was retrieved from various databases, including Google Scholar, PubMed, Science Direct, ClinicalTrials.gov, and government websites. The following terms were used in the search strategies: HIV, vaccines, mRNA vaccines, clinical trials, and preclinical trials. A total of 35 articles were identified and subsequently screened for data regarding mRNA vaccines for HIV. RESULTS mRNA vaccines are an effective solution for HIV treatment, as demonstrated by various research studies referenced in the article. CONCLUSION This review evaluates the current state of HIV-1 mRNA vaccine development, clarifies various targeting strategies, highlights recent research findings, and provides insights into the challenges and potential solutions associated with these issues. In this review, we have explored mRNA vaccines, focusing on their functional structure, design, manufacturing, and distribution methodologies.
Collapse
Affiliation(s)
- Akmal Zubair
- Department of Biotechnology, Quaid-i-Azam University Islamabad, Islamabad, Pakistan
| | - Hanbal Ahmad
- Department of Biotechnology, Quaid-i-Azam University Islamabad, Islamabad, Pakistan
| | - Muhammad Muaz Arif
- Department of Biotechnology, Quaid-i-Azam University Islamabad, Islamabad, Pakistan
| | - Muhammad Ali
- Department of Biotechnology, Quaid-i-Azam University Islamabad, Islamabad, Pakistan
| |
Collapse
|
2
|
Gambari R, Papi C, Gasparello J, Agostinelli E, Finotti A. Preliminary results and a theoretical perspective of co‑treatment using a miR‑93‑5p mimic and aged garlic extract to inhibit the expression of the pro‑inflammatory interleukin‑8 gene. Exp Ther Med 2025; 29:85. [PMID: 40084194 PMCID: PMC11904878 DOI: 10.3892/etm.2025.12835] [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: 10/08/2024] [Accepted: 01/07/2025] [Indexed: 03/16/2025] Open
Abstract
The coronavirus disease-19 (COVID-19) pandemic has been a very significant health issue in the period between 2020 and 2023, forcing research to characterize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) sequences and to develop novel therapeutic approaches. Interleukin-6 (IL-6) and IL-8 are considered significant therapeutic targets for COVID-19 and emerging evidence has suggested that microRNAs (miRNAs/miRs) serve a key role in regulating these genes. MiRNAs are short, 19-25 nucleotides in length, non-coding RNAs that regulate gene expression at the post-transcriptional level through the sequence-selective recognition of the 3'-untranslated region (3'-UTR) of the regulated mRNAs, eventually repressing translation, commonly, via mRNA degradation. For example, among several miRNAs involved in the regulation of the COVID-19 'cytokine storm', miR-93-5p can inhibit IL-8 gene expression by directly targeting the 3'-UTR of IL-8 mRNA. In addition, miR-93-5p can regulate Toll-like receptor-4 (TLR4) and interleukin-1 receptor-associated kinase 4 (IRAK4) expression, thus affecting the nuclear factor-κB (NF-κB) pathway and the expression of NF-κB-regulated genes, such as IL-6, IL-1β and other hyper-expressed genes during the COVID-19 'cytokine storm'. In the present study, the results provided preliminary evidence suggesting that the miR-93-5p-based miRNA therapeutics could be combined with the anti-inflammatory aged garlic extract (AGE) to more effectively inhibit IL-8 gene expression. The human bronchial epithelial IB3-1 cell line was employed as experimental model system. IB3-1 cells were stimulated with the BNT162b2 COVID-19 vaccine and transfected with pre-miR-93-5p in the absence or in the presence of AGE, to verify the inhibitory effects on the BNT162b2-induced expression of the IL-8 gene. The accumulation of IL-8 mRNA was assessed by RT-qPCR; the release of IL-8 protein was determined by Bio-Plex assay. In addition, the possible applications of TLR4/NF-κB inhibitory agents (such as miR-93-5p and AGE) for treating human pathologies at a hyperinflammatory state, such as COVID-19, cystic fibrosis and other respiratory diseases, were summarized.
Collapse
Affiliation(s)
- Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation ‘ETS-ONLUS’, I-00159 Rome, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| |
Collapse
|
3
|
Ngalle Loth A, Maroquenne M, Medjmedj A, Coste F, Bizien T, Pichon C, Logeart-Avramoglou D, Perche F. Structural and functional characterization of a histidylated liposome for mRNA delivery. J Control Release 2025; 379:164-176. [PMID: 39788374 DOI: 10.1016/j.jconrel.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
The development of lipid-based mRNA delivery systems has significantly facilitated recent advances in mRNA-based therapeutics. Liposomes, as the pioneering class of mRNA vectors, continue to lead in clinical trials. We previously developed a histidylated liposome that demonstrated efficient nucleic acid delivery. In this study, the liposome preparation process was optimized by freeze-drying followed by extrusion to homogenize size distribution and improve storage stability. A comprehensive characterization of these LYX liposomes was performed, including evaluation in cellular and murine animal models. LYX liposomes can be stored for up to one year at 4 °C, maintaining a stable size (150 ± 10 nm) and polydispersity index (0.10 ± 0.02), while preserving their transfection efficacy. They exhibit high encapsulation efficacy (∼95 %) and protect mRNA from RNase degradation. Lamellar organization was confirmed by Small Angle X-ray Scattering and CryoTEM, and intracellular trafficking was examined using confocal microscopy. LYX-mRNA lipoplexes can transfect both cell lines and primary cells, albeit with a lower transfection efficacy compared to the commercial Lipofectamine MessengerMAX™ vector. Our data suggest that this could be attributed to slower cell uptake and reduced endosomal escape of LYX. LYX liposomes effectively delivered mRNA encoding therapeutic BMP2 and BMP9 molecules, producing significant amounts of functional proteins that successfully induced BMP signaling. In addition, in vivo studies demonstrated the potential of LYX lipoplexes when incorporated into hydrogels and implanted subcutaneously in mice. These findings provided evidence that LYX liposomes are a promising platform for mRNA delivery, offering versatility for multiple applications.
Collapse
Affiliation(s)
| | - Manon Maroquenne
- Université Paris Cité, CNRS, INSERM, ENVA, B3OA, 75010, Paris, France
| | - Ayoub Medjmedj
- Centre de Biophysique Moléculaire, CBM, CNRS UPR4301, Orléans, France
| | - Franck Coste
- Centre de Biophysique Moléculaire, CBM, CNRS UPR4301, Orléans, France
| | - Thomas Bizien
- Université Paris-Saclay, Synchrotron Soleil, 91190 Saint-Aubin, France
| | - Chantal Pichon
- Inserm UMS 55 ART ARNm and LI2RSO, Université d'Orléans, F-45100 Orléans, France; Institut Universitaire de France, 1 rue Descartes, F-75035 Paris, France
| | | | - Federico Perche
- Centre de Biophysique Moléculaire, CBM, CNRS UPR4301, Orléans, France.
| |
Collapse
|
4
|
Hosseini-Kharat M, Bremmell KE, Grubor-Bauk B, Prestidge CA. Enhancing non-viral DNA delivery systems: Recent advances in improving efficiency and target specificity. J Control Release 2025; 378:170-194. [PMID: 39647508 DOI: 10.1016/j.jconrel.2024.12.002] [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: 06/07/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
DNA-based therapies are often limited by challenges such as stability, long-term integration, low transfection efficiency, and insufficient targeted DNA delivery. This review focuses on recent progress in the design of non-viral delivery systems for enhancing targeted DNA delivery and modulation of therapeutic efficiency. Cellular uptake and intracellular trafficking mechanisms play a crucial role in optimizing gene delivery efficiency. There are two main strategies employed to improve the efficiency of gene delivery vectors: (i) explore different administration routes (e.g., mucosal, intravenous, intramuscular, subcutaneous, intradermal, intratumoural, and intraocular) that best facilitates optimal uptake into the targeted cells and organs and (ii) modify the delivery vectors with cell-specific ligands (e.g., natural ligands, antibodies, peptides, carbohydrates, or aptamers) that enable targeted uptake to specific cells with higher specificity and improved biodistribution. We describe how recent progress in employing these DNA delivery strategies is advancing the field and increasing the clinical translation and ultimate clinical application of DNA therapies.
Collapse
Affiliation(s)
- Mahboubeh Hosseini-Kharat
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia
| | - Kristen E Bremmell
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia
| | - Branka Grubor-Bauk
- Viral Immunology Group, Adelaide Medical School, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA, Australia
| | - Clive A Prestidge
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
5
|
Deng S, Shao H, Shang H, Pang L, Chen X, Cao J, Wang Y, Zhao Z. Development of a Cationic Polymeric Micellar Structure with Endosomal Escape Capability Enables Enhanced Intramuscular Transfection of mRNA-LNPs. Vaccines (Basel) 2024; 13:25. [PMID: 39852804 PMCID: PMC11768556 DOI: 10.3390/vaccines13010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: The endosomal escape of lipid nanoparticles (LNPs) is crucial for efficient mRNA-based therapeutics. Here, we present a cationic polymeric micelle (cPM) as a safe and potent co-delivery system with enhanced endosomal escape capabilities. Methods: We synthesized a cationic and ampholytic di-block copolymer, poly (poly (ethylene glycol)4-5 methacrylatea-co-hexyl methacrylateb)X-b-poly(butyl methacrylatec-co-dimethylaminoethyl methacrylated-co-propyl acrylatee)Y (p(PEG4-5MAa-co-HMAb)X-b-p(BMAc-co-DMAEMAd-co-PAAe)Y), via reversible addition-fragmentation chain transfer polymerization. The cPMs were then formulated using the synthesized polymer by the dispersion-diffusion method and characterized by dynamic light scattering (DLS) and cryo-transmission electron microscopy (CryoTEM). The membrane-destabilization activity of the cPMs was evaluated by a hemolysis assay. We performed an in vivo functional assay of firefly luciferase (Fluc) mRNA using two of the most commonly studied LNPs, SM102 LNP and Dlin-MC3-DMA LNPs. Results: With a particle size of 61.31 ± 0.68 nm and a zeta potential of 37.76 ± 2.18 mV, the cPMs exhibited a 2-3 times higher firefly luciferase signal at the injection site compared to the control groups without cPMs following intramuscular injection in mice, indicating the high potential of cPMs to enhance the endosomal escape efficiency of mRNA-LNPs. Conclusions: The developed cPM, with enhanced endosomal escape capabilities, presents a promising strategy to improve the expression efficiency of delivered mRNAs. This approach offers a novel alternative strategy with no modifications to the inherent properties of mRNA-LNPs, preventing any unforeseeable changes in formulation characteristics. Consequently, this polymer-based nanomaterial holds immense potential for clinical applications in mRNA-based vaccines.
Collapse
Affiliation(s)
- Siyuan Deng
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| | - Han Shao
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| | - Hongtao Shang
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| | - Lingjin Pang
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| | - Xiaomeng Chen
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| | - Jingyi Cao
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
- NeoCura Bio-Medical Technology Co., Ltd., 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China
| | - Yi Wang
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
- NeoCura Bio-Medical Technology Co., Ltd., 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China
| | - Zhao Zhao
- Shenzhen Neocurna Biotechnology Corporation, 12/F, Block B, Building 1, Yinxingzhijie Phase II, Longhua District, Shenzhen 518100, China; (S.D.); (H.S.); (H.S.); (L.P.); (X.C.); (J.C.)
| |
Collapse
|
6
|
Peng S, Zhang Y, Zhao X, Wang Y, Zhang Z, Zhang X, Li J, Zheng H, Zhang Y, Shi H, Li H, Liu L. Pathologic Tissue Injury and Inflammation in Mice Immunized with Plasmid DNA-Encapsulated DOTAP-Based Lipid Nanoparticles. Bioconjug Chem 2024; 35:2015-2026. [PMID: 39656061 DOI: 10.1021/acs.bioconjchem.4c00536] [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: 12/19/2024]
Abstract
Ionizable cationic lipids have been developed to mitigate the toxicity of quaternary ammonium lipids, such as DOTAP. Despite its toxicity, DOTAP can promote localization of lipid nanoparticles (LNPs) in target tissues, serving as one of the ionizable cationic helper lipids. Notably, DOTAP-based nanoadjuvants prepared via microfluidic methods showed a better T-cell response. Previous studies showed that DOTAP-based LNPs prepared by the lipid-film method resulted in obvious adverse events. Therefore, our research focused on evaluating the tissue localization and adverse toxicity of a DOTAP-based delivery system prepared through microfluidic techniques. We assessed the delivery efficacy, biodistribution, inflammatory response, and pathological injury in various tissues. In our study, the plasmid DNA encoding the receptor-binding domain (RBD) of SARS-CoV-2 was encapsulated using a mixture of lipids that included DOTAP, DOPE, cholesterol, and DMG-PEG2000 via microfluidic mixing. The LNP-RBDs were smaller than those prepared via the traditional lipid membrane system. We found that LNP-DNA complexes can be effectively delivered and expressed in muscle tissue, with specific antibodies in serum induced postimmunization. Initial distribution of the liposomes was observed in the muscle and liver. Interestingly, both LNPs and DNA showed sustained presence in the lungs and spleen in the group immunized with DNA-encapsulated DOTAP-based LNPs, whereas lower amounts of DNA were detected in the group immunized with dissociative DNA. We detected obvious inflammatory responses and pathological injuries in the muscle, heart, and liver, and the side effects decreased when the immunization dose decreased. These findings suggest that DOTAP-based LNPs have obvious advantages for targeting the lungs and spleen. Additionally, inflammatory responses and pathological injuries occur in a dose-dependent manner in the muscles, heart, and liver. In conclusion, these findings contribute to the development of an LNP delivery system with DOTAP, highlighting its potential to enhance tissue localization and promote high levels of expression when coordinated with ionizable lipids.
Collapse
Affiliation(s)
- Shasha Peng
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Yifan Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, Yunnan 650118, China
| | - Xin Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Yibin Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Zihan Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Xin Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Jiali Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Huiwen Zheng
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Ying Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Haijing Shi
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Heng Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, Yunnan 650118, China
| | - Longding Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, Yunnan 650118, China
| |
Collapse
|
7
|
Gasparello J, Papi C, Marzaro G, Macone A, Zurlo M, Finotti A, Agostinelli E, Gambari R. Aged Garlic Extract (AGE) and Its Constituent S-Allyl-Cysteine (SAC) Inhibit the Expression of Pro-Inflammatory Genes Induced in Bronchial Epithelial IB3-1 Cells by Exposure to the SARS-CoV-2 Spike Protein and the BNT162b2 Vaccine. Molecules 2024; 29:5938. [PMID: 39770027 PMCID: PMC11677098 DOI: 10.3390/molecules29245938] [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: 10/31/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 01/05/2025] Open
Abstract
Garlic (Allium sativum L.) is a species of the onion family (Alliaceae) widely used as a food and a folk medicine. The objective of this study was to determine the effects of AGE (aged garlic extract) on pro-inflammatory genes relevant to COVID-19. To this aim, we treated bronchial epithelial IB3-1 cells with SARS-CoV-2 spike protein (S-protein) or with the COVID-19 BNT162b2 vaccine in the absence or in the presence of AGE. The results obtained demonstrated that AGE is a potent inhibitor of the S-protein-induced expression of the IL-1β, IL-6 and IL-8 genes. Bio-Plex analysis demonstrated that AGE reduced release of IL-6 and IL-8, which were highly induced by S-protein. No inhibition of cells' growth, toxicity and pro-apoptotic effects were found in AGE-treated cells. The effects of one of the major AGE constituents (S-allyl cysteine, SAC) were studied on the same experimental model systems. SAC was able to inhibit the S-protein-induced expression of IL-1β, IL-6 and IL-8 genes and extracellular release of IL-6 and IL-8, confirming that S-allyl-cysteine is one of the constituents of AGE that is responsible for inhibiting S-protein-induced pro-inflammatory genes. Docking experiments suggest that a possible mechanism of action of SAC is an interference with the activity of Toll-like receptors (TLRs), particularly TLR4, thereby inhibiting NF-κB- and NF-κB-regulated genes, such as IL-1β, IL-6 and IL-8 genes. These results suggest that both AGE and SAC deserve further experimental efforts to verify their effects on pro-inflammatory genes in SARS-CoV-2-infected cells.
Collapse
Affiliation(s)
- Jessica Gasparello
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Giovanni Marzaro
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy;
| | - Alberto Macone
- Department of Biochemical Sciences ‘A. Rossi Fanelli’, Sapienza University of Rome, 00185 Rome, Italy;
| | - Matteo Zurlo
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Enzo Agostinelli
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico 155, 00161 Rome, Italy
- International Polyamines Foundation ‘ETS-ONLUS’, Via del Forte Tiburtino 98, 00159 Rome, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| |
Collapse
|
8
|
Ramadan E, Ahmed A, Naguib YW. Advances in mRNA LNP-Based Cancer Vaccines: Mechanisms, Formulation Aspects, Challenges, and Future Directions. J Pers Med 2024; 14:1092. [PMID: 39590584 PMCID: PMC11595619 DOI: 10.3390/jpm14111092] [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: 09/21/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
After the COVID-19 pandemic, mRNA-based vaccines have emerged as a revolutionary technology in immunization and vaccination. These vaccines have shown remarkable efficacy against the virus and opened up avenues for their possible application in other diseases. This has renewed interest and investment in mRNA vaccine research and development, attracting the scientific community to explore all its other applications beyond infectious diseases. Recently, researchers have focused on the possibility of adapting this vaccination approach to cancer immunotherapy. While there is a huge potential, challenges still remain in the design and optimization of the synthetic mRNA molecules and the lipid nanoparticle delivery system required to ensure the adequate elicitation of the immune response and the successful eradication of tumors. This review points out the basic mechanisms of mRNA-LNP vaccines in cancer immunotherapy and recent approaches in mRNA vaccine design. This review displays the current mRNA modifications and lipid nanoparticle components and how these factors affect vaccine efficacy. Furthermore, this review discusses the future directions and clinical applications of mRNA-LNP vaccines in cancer treatment.
Collapse
Affiliation(s)
- Eslam Ramadan
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, H-6720 Szeged, Hungary;
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Ali Ahmed
- Department of Clinical Pharmacy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt;
| | - Youssef Wahib Naguib
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
9
|
Eftekhari Z, Zohrabi H, Oghalaie A, Ebrahimi T, Shariati FS, Behdani M, Kazemi-Lomedasht F. Advancements and challenges in mRNA and ribonucleoprotein-based therapies: From delivery systems to clinical applications. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102313. [PMID: 39281702 PMCID: PMC11402252 DOI: 10.1016/j.omtn.2024.102313] [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] [Indexed: 09/18/2024]
Abstract
The use of mRNA and ribonucleoproteins (RNPs) as therapeutic agents is a promising strategy for treating diseases such as cancer and infectious diseases. This review provides recent advancements and challenges in mRNA- and RNP-based therapies, focusing on delivery systems such as lipid nanoparticles (LNPs), which ensure efficient delivery to target cells. Strategies such as microfluidic devices are employed to prepare LNPs loaded with mRNA and RNPs, demonstrating effective genome editing and protein expression in vitro and in vivo. These applications extend to cancer treatment and infectious disease management, with promising results in genome editing for cancer therapy using LNPs encapsulating Cas9 mRNA and single-guide RNA. In addition, tissue-specific targeting strategies offer potential for improved therapeutic outcomes and reduced off-target effects. Despite progress, challenges such as optimizing delivery efficiency and targeting remain. Future research should enhance delivery efficiency, explore tissue-specific targeting, investigate combination therapies, and advance clinical translation. In conclusion, mRNA- and RNP-based therapies offer a promising avenue for treating various diseases and have the potential to revolutionize medicine, providing new hope for patients worldwide.
Collapse
Affiliation(s)
- Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Horieh Zohrabi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Tahereh Ebrahimi
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Fatemeh Sadat Shariati
- Department of Influenza and other Respiratory Viruses, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| |
Collapse
|
10
|
Korzun T, Moses AS, Jozic A, Grigoriev V, Newton S, Kim J, Diba P, Sattler A, Levasseur PR, Le N, Singh P, Sharma KS, Goo YT, Mamnoon B, Raitmayr C, Souza APM, Taratula OR, Sahay G, Taratula O, Marks DL. Lipid Nanoparticles Elicit Reactogenicity and Sickness Behavior in Mice Via Toll-Like Receptor 4 and Myeloid Differentiation Protein 88 Axis. ACS NANO 2024; 18:24842-24859. [PMID: 39186628 PMCID: PMC11916992 DOI: 10.1021/acsnano.4c05088] [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] [Indexed: 08/28/2024]
Abstract
mRNA therapeutics encapsulated in lipid nanoparticles (LNPs) offer promising avenues for treating various diseases. While mRNA vaccines anticipate immunogenicity, the associated reactogenicity of mRNA-loaded LNPs poses significant challenges, especially in protein replacement therapies requiring multiple administrations, leading to adverse effects and suboptimal therapeutic outcomes. Historically, research has primarily focused on the reactogenicity of mRNA cargo, leaving the role of LNPs understudied in this context. Adjuvanticity and pro-inflammatory characteristics of LNPs, originating at least in part from ionizable lipids, may induce inflammation, activate toll-like receptors (TLRs), and impact mRNA translation. Knowledge gaps remain in understanding LNP-induced TLR activation and its impact on induction of animal sickness behavior. We hypothesized that ionizable lipids in LNPs, structurally resembling lipid A from lipopolysaccharide, could activate TLR4 signaling via MyD88 and TRIF adaptors, thereby propagating LNP-associated reactogenicity. Our comprehensive investigation utilizing gene ablation studies and pharmacological receptor manipulation proves that TLR4 activation by LNPs triggers distinct physiologically meaningful responses in mice. We show that TLR4 and MyD88 are essential for reactogenic signal initiation, pro-inflammatory gene expression, and physiological outcomes like food intake and body weight─robust metrics of sickness behavior in mice. The application of the TLR4 inhibitor TAK-242 effectively reduces the reactogenicity associated with LNPs by mitigating TLR4-driven inflammatory responses. Our findings elucidate the critical role of the TLR4-MyD88 axis in LNP-induced reactogenicity, providing a mechanistic framework for developing safer mRNA therapeutics and offering a strategy to mitigate adverse effects through targeted inhibition of this pathway.
Collapse
Affiliation(s)
- Tetiana Korzun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon, 97239, USA
- Medical Scientist Training Program, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239, USA
| | - Abraham S. Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Antony Jozic
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Vladislav Grigoriev
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Samuel Newton
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Mail Code L481 Portland, Oregon, 97239, USA
| | - Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Parham Diba
- Medical Scientist Training Program, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239, USA
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Mail Code L481 Portland, Oregon, 97239, USA
| | - Ariana Sattler
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Ave, Portland, OR 97201
| | - Peter R. Levasseur
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Mail Code L481 Portland, Oregon, 97239, USA
| | - Ngoc Le
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Prem Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Kongbrailatpam Shitaljit Sharma
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Yoon Tae Goo
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Babak Mamnoon
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Constanze Raitmayr
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Ana Paula Mesquita Souza
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Olena R. Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon, 97201, USA
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon, 97239, USA
| | - Daniel L. Marks
- Endevica Bio, 1935 Techny Rd, Northbrook, Illinois, 60062, USA
| |
Collapse
|
11
|
Delehedde C, Ciganek I, Bernard PL, Laroui N, Da Silva CC, Gonçalves C, Nunes J, Bennaceur-Griscelli AL, Imeri J, Huyghe M, Even L, Midoux P, Rameix N, Guittard G, Pichon C. Enhancing natural killer cells proliferation and cytotoxicity using imidazole-based lipid nanoparticles encapsulating interleukin-2 mRNA. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102263. [PMID: 39104868 PMCID: PMC11298638 DOI: 10.1016/j.omtn.2024.102263] [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: 03/09/2024] [Accepted: 06/24/2024] [Indexed: 08/07/2024]
Abstract
mRNA applications have undergone unprecedented applications-from vaccination to cell therapy. Natural killer (NK) cells are recognized to have a significant potential in immunotherapy. NK-based cell therapy has drawn attention as allogenic graft with a minimal graft-versus-host risk leading to easier off-the-shelf production. NK cells can be engineered with either viral vectors or electroporation, involving high costs, risks, and toxicity, emphasizing the need for alternative way as mRNA technology. We successfully developed, screened, and optimized novel lipid-based platforms based on imidazole lipids. Formulations are produced by microfluidic mixing and exhibit a size of approximately 100 nm with a polydispersity index of less than 0.2. They are able to transfect NK-92 cells, KHYG-1 cells, and primary NK cells with high efficiency without cytotoxicity, while Lipofectamine Messenger Max and D-Lin-MC3 lipid nanoparticle-based formulations do not. Moreover, the translation of non-modified mRNA was higher and more stable in time compared with a modified one. Remarkably, the delivery of therapeutically relevant interleukin 2 mRNA resulted in extended viability together with preserved activation markers and cytotoxic ability of both NK cell lines and primary NK cells. Altogether, our platforms feature all prerequisites needed for the successful deployment of NK-based therapeutic strategies.
Collapse
Affiliation(s)
- Christophe Delehedde
- Centre de Biophysique Moléculaire, CNRS UPR4301, 45071 Orléans Cedex 02, France
- Sanofi R&D, Integrated Drug Discovery, 94400 Vitry-sur-Seine, France
| | - Ivan Ciganek
- Centre de Biophysique Moléculaire, CNRS UPR4301, 45071 Orléans Cedex 02, France
- Inserm UMS 55 ART ARNm and University of Orléans, 45100 Orléans, France
- Sanofi R&D, Integrated Drug Discovery, 94400 Vitry-sur-Seine, France
| | - Pierre Louis Bernard
- Immunity and Cancer Team, Onco-Hemato Immuno-Onco Department, OHIO, Cancer Research Centre of Marseille, CRCM, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, 13273 Marseille, France
| | - Nabila Laroui
- Centre de Biophysique Moléculaire, CNRS UPR4301, 45071 Orléans Cedex 02, France
- Inserm UMS 55 ART ARNm and University of Orléans, 45100 Orléans, France
| | - Cathy Costa Da Silva
- Immunity and Cancer Team, Onco-Hemato Immuno-Onco Department, OHIO, Cancer Research Centre of Marseille, CRCM, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, 13273 Marseille, France
| | - Cristine Gonçalves
- Centre de Biophysique Moléculaire, CNRS UPR4301, 45071 Orléans Cedex 02, France
- Inserm UMS 55 ART ARNm and University of Orléans, 45100 Orléans, France
| | - Jacques Nunes
- Immunity and Cancer Team, Onco-Hemato Immuno-Onco Department, OHIO, Cancer Research Centre of Marseille, CRCM, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, 13273 Marseille, France
| | - Anne-Lise Bennaceur-Griscelli
- Inserm U 1310 F-94800 Villejuif and CITHERA/ UMS45 Infrastructure INGESTEM, 91100 Evry, France
- University Paris Saclay, APHP Paul Brousse Hospital, School of Medicine, 94270 Le Kremlin Bicêtre, France
| | - Jusuf Imeri
- Inserm U 1310 F-94800 Villejuif and CITHERA/ UMS45 Infrastructure INGESTEM, 91100 Evry, France
- University Paris Saclay, APHP Paul Brousse Hospital, School of Medicine, 94270 Le Kremlin Bicêtre, France
| | - Matthias Huyghe
- Inserm U 1310 F-94800 Villejuif and CITHERA/ UMS45 Infrastructure INGESTEM, 91100 Evry, France
- University Paris Saclay, APHP Paul Brousse Hospital, School of Medicine, 94270 Le Kremlin Bicêtre, France
| | - Luc Even
- Sanofi R&D, Integrated Drug Discovery, 94400 Vitry-sur-Seine, France
| | - Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR4301, 45071 Orléans Cedex 02, France
- Inserm UMS 55 ART ARNm and University of Orléans, 45100 Orléans, France
| | - Nathalie Rameix
- Sanofi R&D, Integrated Drug Discovery, 94400 Vitry-sur-Seine, France
| | - Geoffrey Guittard
- Immunity and Cancer Team, Onco-Hemato Immuno-Onco Department, OHIO, Cancer Research Centre of Marseille, CRCM, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, 13273 Marseille, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, 45071 Orléans Cedex 02, France
- Inserm UMS 55 ART ARNm and University of Orléans, 45100 Orléans, France
- Institut Universitaire de France, 1 rue Descartes, 75035 Paris, France
| |
Collapse
|
12
|
Qassem S, Breier D, Naidu GS, Hazan-Halevy I, Peer D. Unlocking the therapeutic potential of locked nucleic acids through lipid nanoparticle delivery. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102224. [PMID: 38933259 PMCID: PMC11201112 DOI: 10.1016/j.omtn.2024.102224] [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] [Indexed: 06/28/2024]
Abstract
Locked nucleic acids (LNAs) are a subtype of antisense oligonucleotides (ASOs) that are characterized by a bridge within the sugar moiety. LNAs owe their robustness to this chemical modification, which as the name suggests, locks it in one conformation. This perspective includes two components: a general overview on ASOs from one side and on delivery issues focusing on lipid nanoparticles (LNPs) on the other side. Throughout, a screening of the ongoing clinical trials involving ASOs is given, as well as a take on the versatility and challenges of using LNAs. Finally, we highlight the potential of LNPs as carriers for the successful delivery of LNAs.
Collapse
Affiliation(s)
- Shahd Qassem
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dor Breier
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Gonna Somu Naidu
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Inbal Hazan-Halevy
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
13
|
Fedorovskiy AG, Antropov DN, Dome AS, Puchkov PA, Makarova DM, Konopleva MV, Matveeva AM, Panova EA, Shmendel EV, Maslov MA, Dmitriev SE, Stepanov GA, Markov OV. Novel Efficient Lipid-Based Delivery Systems Enable a Delayed Uptake and Sustained Expression of mRNA in Human Cells and Mouse Tissues. Pharmaceutics 2024; 16:684. [PMID: 38794346 PMCID: PMC11125954 DOI: 10.3390/pharmaceutics16050684] [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: 04/18/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Over the past decade, mRNA-based therapy has displayed significant promise in a wide range of clinical applications. The most striking example of the leap in the development of mRNA technologies was the mass vaccination against COVID-19 during the pandemic. The emergence of large-scale technology and positive experience of mRNA immunization sparked the development of antiviral and anti-cancer mRNA vaccines as well as therapeutic mRNA agents for genetic and other diseases. To facilitate mRNA delivery, lipid nanoparticles (LNPs) have been successfully employed. However, the diverse use of mRNA therapeutic approaches requires the development of adaptable LNP delivery systems that can control the kinetics of mRNA uptake and expression in target cells. Here, we report effective mRNA delivery into cultured mammalian cells (HEK293T, HeLa, DC2.4) and living mouse muscle tissues by liposomes containing either 1,26-bis(cholest-5-en-3β-yloxycarbonylamino)-7,11,16,20-tetraazahexacosane tetrahydrochloride (2X3) or the newly applied 1,30-bis(cholest-5-en-3β-yloxycarbonylamino)-9,13,18,22-tetraaza-3,6,25,28-tetraoxatriacontane tetrahydrochloride (2X7) cationic lipids. Using end-point and real-time monitoring of Fluc mRNA expression, we showed that these LNPs exhibited an unusually delayed (of over 10 h in the case of the 2X7-based system) but had highly efficient and prolonged reporter activity in cells. Accordingly, both LNP formulations decorated with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] (DSPE-PEG2000) provided efficient luciferase production in mice, peaking on day 3 after intramuscular injection. Notably, the bioluminescence was observed only at the site of injection in caudal thigh muscles, thereby demonstrating local expression of the model gene of interest. The developed mRNA delivery systems hold promise for prophylactic applications, where sustained synthesis of defensive proteins is required, and open doors to new possibilities in mRNA-based therapies.
Collapse
Affiliation(s)
- Artem G. Fedorovskiy
- Belozersky Institute of Physico-Chemical Biology, Department of Materials Science, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.G.F.); (M.V.K.); (E.A.P.)
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
| | - Denis N. Antropov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (D.N.A.); (A.S.D.); (A.M.M.); (G.A.S.)
| | - Anton S. Dome
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (D.N.A.); (A.S.D.); (A.M.M.); (G.A.S.)
| | - Pavel A. Puchkov
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
| | - Daria M. Makarova
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
| | - Maria V. Konopleva
- Belozersky Institute of Physico-Chemical Biology, Department of Materials Science, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.G.F.); (M.V.K.); (E.A.P.)
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Anastasiya M. Matveeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (D.N.A.); (A.S.D.); (A.M.M.); (G.A.S.)
| | - Eugenia A. Panova
- Belozersky Institute of Physico-Chemical Biology, Department of Materials Science, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.G.F.); (M.V.K.); (E.A.P.)
| | - Elena V. Shmendel
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
| | - Mikhail A. Maslov
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
| | - Sergey E. Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Department of Materials Science, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.G.F.); (M.V.K.); (E.A.P.)
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Grigory A. Stepanov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (D.N.A.); (A.S.D.); (A.M.M.); (G.A.S.)
| | - Oleg V. Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (D.N.A.); (A.S.D.); (A.M.M.); (G.A.S.)
| |
Collapse
|
14
|
Kimura S, Harashima H. Nano-Bio Interactions: Exploring the Biological Behavior and the Fate of Lipid-Based Gene Delivery Systems. BioDrugs 2024; 38:259-273. [PMID: 38345754 DOI: 10.1007/s40259-024-00647-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2024] [Indexed: 03/06/2024]
Abstract
Gene therapy for many diseases is rapidly becoming a reality, as demonstrated by the recent approval of various nucleic acid-based therapeutics. Non-viral systems such as lipid-based carriers, lipid nanoparticles (LNPs), for delivering different payloads including small interfering RNA, plasmid DNA, and messenger RNA have been particularly extensively explored and developed for clinical uses. One of the most important issues in LNP development is delivery to extrahepatic tissues. To achieve this, various lipids and lipid-like materials are being examined and screened. Several LNP formulations that target extrahepatic tissues, such as the spleen and the lungs have been developed by adjusting the lipid compositions of LNPs. However, mechanistic details of how the characteristics of LNPs affect delivery efficiency remains unclear. The purpose of this review is to provide an overview of LNP-based nucleic acid delivery focusing on LNP components and their structures, as well as discussing biological factors, such as biomolecular corona and cellular responses related to the delivery efficiency.
Collapse
Affiliation(s)
- Seigo Kimura
- Integrated Research Consortium on Chemical Sciences, Graduate School of Science, Nagoya University, Nagoya, 464-8602, Japan.
| | - Hideyoshi Harashima
- Laboratory for Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
| |
Collapse
|
15
|
Imani S, Tagit O, Pichon C. Neoantigen vaccine nanoformulations based on Chemically synthesized minimal mRNA (CmRNA): small molecules, big impact. NPJ Vaccines 2024; 9:14. [PMID: 38238340 PMCID: PMC10796345 DOI: 10.1038/s41541-024-00807-1] [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: 09/03/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
Recently, chemically synthesized minimal mRNA (CmRNA) has emerged as a promising alternative to in vitro transcribed mRNA (IVT-mRNA) for cancer therapy and immunotherapy. CmRNA lacking the untranslated regions and polyadenylation exhibits enhanced stability and efficiency. Encapsulation of CmRNA within lipid-polymer hybrid nanoparticles (LPPs) offers an effective approach for personalized neoantigen mRNA vaccines with improved control over tumor growth. LPP-based delivery systems provide superior pharmacokinetics, stability, and lower toxicity compared to viral vectors, naked mRNA, or lipid nanoparticles that are commonly used for mRNA delivery. Precise customization of LPPs in terms of size, surface charge, and composition allows for optimized cellular uptake, target specificity, and immune stimulation. CmRNA-encoded neo-antigens demonstrate high translational efficiency, enabling immune recognition by CD8+ T cells upon processing and presentation. This perspective highlights the potential benefits, challenges, and future directions of CmRNA neoantigen vaccines in cancer therapy compared to Circular RNAs and IVT-mRNA. Further research is needed to optimize vaccine design, delivery, and safety assessment in clinical trials. Nevertheless, personalized LPP-CmRNA vaccines hold great potential for advancing cancer immunotherapy, paving the way for personalized medicine.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China.
| | - Oya Tagit
- Institute of Chemistry and Bioanalytics, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland
| | - Chantal Pichon
- Center of Molecular Biophysics, CNRS, Orléans, France.
- ART-ARNm, National Institute of Health and Medical Research (Inserm) and University of Orléans, Orléans, France.
- Institut Universitaire de France, Paris, France.
| |
Collapse
|
16
|
Delehedde C, Ciganek I, Laroui N, Rameix N, Perche F, Pichon C. Messenger RNA Lipid-Based Nanoparticles: Optimization of Formulations in the Lab. Methods Mol Biol 2024; 2786:255-287. [PMID: 38814399 DOI: 10.1007/978-1-0716-3770-8_12] [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: 05/31/2024]
Abstract
Among the large variety of messenger RNA (mRNA) delivery systems, those developed with lipid-based formulations were the most widely used and efficient. In our lab, we produced different mRNA formulations made with liposomes, hybrid lipid polymer, and lipid nanoparticles. Our formulations were made with lipids bearing imidazole groups that trigger the endosomal escape of nanoparticles once protonated inside the mild acidic milieu of endosomes upon their cell uptake. Herein, we describe protocols that we used to produce, optimize, and characterize those formulations. The transfection efficiency is influenced by various factors including the physicochemical parameters of the nanoparticles, their efficiency to be internalized in cells, and their intracellular routing as well as their capacity to induce immune system sensors. We provide details on how to quantify the amount of mRNA nanoparticles uptake by cells and evaluate the acidity of the intracellular compartments where they are located, to investigate the endosomal escape, and to assess the activation of innate immune sensors as phosphorylation of PKR hampering mRNA translation.
Collapse
Affiliation(s)
- Christophe Delehedde
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Ivan Ciganek
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
- Inserm, ART-ARNm Inserm US55, Orléans, France
| | - Nabila Laroui
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
- Inserm, ART-ARNm Inserm US55, Orléans, France
| | - Nathalie Rameix
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Federico Perche
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
| | - Chantal Pichon
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France.
- Orléans University, Château de la Source, Orleans, France.
- Institut Universitaire de France, Paris, France.
- Inserm, ART-ARNm Inserm US55, Orléans, France.
| |
Collapse
|
17
|
Delehedde C, Ciganek I, Rameix N, Laroui N, Gonçalves C, Even L, Midoux P, Pichon C. Impact of net charge, targeting ligand amount and mRNA modification on the uptake, intracellular routing and the transfection efficiency of mRNA lipopolyplexes in dendritic cells. Int J Pharm 2023; 647:123531. [PMID: 37863445 DOI: 10.1016/j.ijpharm.2023.123531] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/08/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
Targeting mRNA formulations to achieve cell specificity is one of the challenges that must be tackled to mettle their therapeutic potential. Here, lipopolyplexes (LPR) bearing tri-mannose-lipid (TM) are used to target mannose receptor on dendritic cells. We investigated the impact of the net charge and percentage of TM units on the binding, uptake, transfection efficiency (TE) and RNA sensors activation. Binding and uptake capacities of naked and targeted LPR increase with the percent of cationic lipid, but the latter are 2-fold more up taken by the cells. Cationic LPR bearing 5 % and 10 % TM were localized in acidic compartments in contrast to naked LPR and 2.5 % TM-LPR. The drawback is the dramatic decrease of TE as the number of TM-units increases. Cationic LPR bearing 5 % and 10 % TM strongly induced NF-κB and PKR phosphorylation at 6 h. Conversely, mTOR is less activated in line with their low TE. Those side effects are overcome by using 5-methoxyuridine mRNA resulting in an improved TE due to non-phosphorylation of NF-κB and PKR and mTOR activation. Our results point out that targeting DC via mannose receptor triggers a higher uptake of cationic LPRs and fast routing to acidic compartments, and that efficient TE requires low number of TM units use or modified mRNA to escape RNA sensors activation to enhance the translation.
Collapse
Affiliation(s)
- Christophe Delehedde
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France; Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Ivan Ciganek
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Nathalie Rameix
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Nabila Laroui
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Cristine Gonçalves
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Luc Even
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France; Inserm UMS 55 ART ARNm and University of Orléans, F-45100 Orléans; Institut Universitaire de France, 1 rue Descartes, F-75035 Paris, France.
| |
Collapse
|
18
|
Miliotou AN, Georgiou-Siafis SK, Ntenti C, Pappas IS, Papadopoulou LC. Recruiting In Vitro Transcribed mRNA against Cancer Immunotherapy: A Contemporary Appraisal of the Current Landscape. Curr Issues Mol Biol 2023; 45:9181-9214. [PMID: 37998753 PMCID: PMC10670245 DOI: 10.3390/cimb45110576] [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: 10/15/2023] [Revised: 11/05/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Over 100 innovative in vitro transcribed (IVT)-mRNAs are presently undergoing clinical trials, with a projected substantial impact on the pharmaceutical market in the near future. Τhe idea behind this is that after the successful cellular internalization of IVT-mRNAs, they are subsequently translated into proteins with therapeutic or prophylactic relevance. Simultaneously, cancer immunotherapy employs diverse strategies to mobilize the immune system in the battle against cancer. Therefore, in this review, the fundamental principles of IVT-mRNA to its recruitment in cancer immunotherapy, are discussed and analyzed. More specifically, this review paper focuses on the development of mRNA vaccines, the exploitation of neoantigens, as well as Chimeric Antigen Receptor (CAR) T-Cells, showcasing their clinical applications and the ongoing trials for the development of next-generation immunotherapeutics. Furthermore, this study investigates the synergistic potential of combining the CAR immunotherapy and the IVT-mRNAs by introducing our research group novel, patented delivery method that utilizes the Protein Transduction Domain (PTD) technology to transduce the IVT-mRNAs encoding the CAR of interest into the Natural Killer (NK)-92 cells, highlighting the potential for enhancing the CAR NK cell potency, efficiency, and bioenergetics. While IVT-mRNA technology brings exciting progress to cancer immunotherapy, several challenges and limitations must be acknowledged, such as safety, toxicity, and delivery issues. This comprehensive exploration of IVT-mRNA technology, in line with its applications in cancer therapeutics, offers valuable insights into the opportunities and challenges in the evolving landscape of cancer immunotherapy, setting the stage for future advancements in the field.
Collapse
Affiliation(s)
- Androulla N. Miliotou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
- Department of Health Sciences, KES College, 1055 Nicosia, Cyprus
- Faculty of Pharmacy, Department of Health Sciences, University of Nicosia, 1700 Nicosia, Cyprus
| | - Sofia K. Georgiou-Siafis
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
- Laboratory of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Thessaly, 43100 Karditsa, Thessaly, Greece;
| | - Charikleia Ntenti
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
- 1st Laboratory of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece
| | - Ioannis S. Pappas
- Laboratory of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Thessaly, 43100 Karditsa, Thessaly, Greece;
| | - Lefkothea C. Papadopoulou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
| |
Collapse
|
19
|
Kimura S, Harashima H. On the mechanism of tissue-selective gene delivery by lipid nanoparticles. J Control Release 2023; 362:797-811. [PMID: 37004796 DOI: 10.1016/j.jconrel.2023.03.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/04/2023]
Abstract
The era of nucleic acid nanomedicine has arrived, as evidenced by Patisiran, a small interfering RNA (siRNA) encapsulated lipid nanoparticle (LNP), and mRNA-loaded LNPs used in COVID-19 vaccines. The diversity of nano-designs for delivering nucleic acid molecules tested in Phase II/III clinical trials reflects the potential of these technologies. These breakthroughs in non-viral gene delivery, including the use of LNPs, have attracted substantial interest worldwide for developing more effective drugs. A next step in this field is to target tissues other than the liver, which requires significant research efforts and material development. However, mechanistic studies in this area are lacking. This study compares two types of LNPs with different tissue-selectivity for delivering plasmid DNA (pDNA), one being liver-selective and the other spleen-selective, in an effort to understand the mechanisms responsible for differences in gene expression of delivered genes. We observed little difference in the biodistribution of these two LNPs despite the 100-1000-fold differences in gene expression. We then quantified the amount of delivered pDNA and mRNA expression in each tissue by quantitative real-time PCR (qPCR) to evaluate various intracellular processes, such as nuclear delivery, transcription and translation. The results showed a >100-fold difference in the translation step but there were little differences in amount of pDNA delivered to the nucleus or the amount of mRNA expression for the two LNP deliveries. Our findings suggest that endogenous factors affect gene expression efficiency not the extent of biodistribution.
Collapse
Affiliation(s)
- Seigo Kimura
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan; Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan; Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
20
|
Ma H, Xing F, Zhou Y, Yu P, Luo R, Xu J, Xiang Z, Rommens PM, Duan X, Ritz U. Design and fabrication of intracellular therapeutic cargo delivery systems based on nanomaterials: current status and future perspectives. J Mater Chem B 2023; 11:7873-7912. [PMID: 37551112 DOI: 10.1039/d3tb01008b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Intracellular cargo delivery, the introduction of small molecules, proteins, and nucleic acids into a specific targeted site in a biological system, is an important strategy for deciphering cell function, directing cell fate, and reprogramming cell behavior. With the advancement of nanotechnology, many researchers use nanoparticles (NPs) to break through biological barriers to achieving efficient targeted delivery in biological systems, bringing a new way to realize efficient targeted drug delivery in biological systems. With a similar size to many biomolecules, NPs possess excellent physical and chemical properties and a certain targeting ability after functional modification on the surface of NPs. Currently, intracellular cargo delivery based on NPs has emerged as an important strategy for genome editing regimens and cell therapy. Although researchers can successfully deliver NPs into biological systems, many of them are delivered very inefficiently and are not specifically targeted. Hence, the development of efficient, target-capable, and safe nanoscale drug delivery systems to deliver therapeutic substances to cells or organs is a major challenge today. In this review, on the basis of describing the research overview and classification of NPs, we focused on the current research status of intracellular cargo delivery based on NPs in biological systems, and discuss the current problems and challenges in the delivery process of NPs in biological systems.
Collapse
Affiliation(s)
- Hong Ma
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Fei Xing
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Yuxi Zhou
- Department of Periodontology, Justus-Liebig-University of Giessen, Ludwigstraße 23, 35392 Giessen, Germany
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Rong Luo
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Jiawei Xu
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Zhou Xiang
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Xin Duan
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
- Department of Orthopedic Surgery, The Fifth People's Hospital of Sichuan Province, Chengdu, China
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
21
|
Yihunie W, Nibret G, Aschale Y. Recent Advances in Messenger Ribonucleic Acid (mRNA) Vaccines and Their Delivery Systems: A Review. Clin Pharmacol 2023; 15:77-98. [PMID: 37554660 PMCID: PMC10405914 DOI: 10.2147/cpaa.s418314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/28/2023] [Indexed: 08/10/2023] Open
Abstract
Messenger ribonucleic acid (mRNA) was found as the intermediary that transfers genetic information from DNA to ribosomes for protein synthesis in 1961. The emergency use authorization of the two covid-19 mRNA vaccines, BNT162b2 and mRNA-1273, is a significant achievement in the history of vaccine development. Because they are generated in a cell-free environment using the in vitro transcription (IVT) process, mRNA vaccines are risk-free. Moreover, chemical modifications to the mRNA molecule, such as cap structures and changed nucleosides, have proved critical in overcoming immunogenicity concerns, achieving sustained stability, and achieving effective, accurate protein production in vivo. Several vaccine delivery strategies (including protamine, lipid nanoparticles (LNPs), polymers, nanoemulsions, and cell-based administration) were also optimized to load and transport RNA into the cytosol. LNPs, which are composed of a cationic or a pH-dependent ionizable lipid layer, a polyethylene glycol (PEG) component, phospholipids, and cholesterol, are the most advanced systems for delivering mRNA vaccines. Moreover, modifications of the four components that make up the LNPs showed to increase vaccine effectiveness and reduce side effects. Furthermore, the introduction of biodegradable lipids improved LNP biocompatibility. Furthermore, mRNA-based therapies are expected to be effective treatments for a variety of refractory conditions, including infectious diseases, metabolic genetic diseases, cancer, cardiovascular and cerebrovascular diseases. Therefore, the present review aims to provide the scientific community with up-to-date information on mRNA vaccines and their delivery systems.
Collapse
Affiliation(s)
- Wubetu Yihunie
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Getinet Nibret
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Aschale
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
22
|
Korzun T, Moses AS, Diba P, Sattler AL, Taratula OR, Sahay G, Taratula O, Marks DL. From Bench to Bedside: Implications of Lipid Nanoparticle Carrier Reactogenicity for Advancing Nucleic Acid Therapeutics. Pharmaceuticals (Basel) 2023; 16:1088. [PMID: 37631003 PMCID: PMC10459564 DOI: 10.3390/ph16081088] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
In biomedical applications, nanomaterial-based delivery vehicles, such as lipid nanoparticles, have emerged as promising instruments for improving the solubility, stability, and encapsulation of various payloads. This article provides a formal review focusing on the reactogenicity of empty lipid nanoparticles used as delivery vehicles, specifically emphasizing their application in mRNA-based therapies. Reactogenicity refers to the adverse immune responses triggered by xenobiotics, including administered lipid nanoparticles, which can lead to undesirable therapeutic outcomes. The key components of lipid nanoparticles, which include ionizable lipids and PEG-lipids, have been identified as significant contributors to their reactogenicity. Therefore, understanding the relationship between lipid nanoparticles, their structural constituents, cytokine production, and resultant reactogenic outcomes is essential to ensure the safe and effective application of lipid nanoparticles in mRNA-based therapies. Although efforts have been made to minimize these adverse reactions, further research and standardization are imperative. By closely monitoring cytokine profiles and assessing reactogenic manifestations through preclinical and clinical studies, researchers can gain valuable insights into the reactogenic effects of lipid nanoparticles and develop strategies to mitigate undesirable reactions. This comprehensive review underscores the importance of investigating lipid nanoparticle reactogenicity and its implications for the development of mRNA-lipid nanoparticle therapeutics in various applications beyond vaccine development.
Collapse
Affiliation(s)
- Tetiana Korzun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR 97201, USA; (T.K.)
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Avenue, Portland, OR 97239, USA
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Abraham S. Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR 97201, USA; (T.K.)
| | - Parham Diba
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Ariana L. Sattler
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Avenue, Portland, OR 97201, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 2730 S Moody Avenue, Portland, OR 97201, USA
| | - Olena R. Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR 97201, USA; (T.K.)
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR 97201, USA; (T.K.)
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR 97201, USA; (T.K.)
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Avenue, Portland, OR 97239, USA
| | - Daniel L. Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Avenue, Portland, OR 97201, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 2730 S Moody Avenue, Portland, OR 97201, USA
| |
Collapse
|
23
|
Ci L, Hard M, Zhang H, Gandham S, Hua S, Wickwire J, Wehrman T, Slauter R, Auerbach A, Kenney M, Mercer G, Hendrick T, Almarsson Ö, Cheung E, Burdette D. Biodistribution of Lipid 5, mRNA, and Its Translated Protein Following Intravenous Administration of mRNA-Encapsulated Lipid Nanoparticles in Rats. Drug Metab Dispos 2023; 51:813-823. [PMID: 37208184 DOI: 10.1124/dmd.122.000980] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 03/17/2023] [Accepted: 04/05/2023] [Indexed: 05/21/2023] Open
Abstract
RNA-based therapeutics and vaccines represent a novel and expanding class of medicines, the success of which depends on the encapsulation and protection of mRNA molecules in lipid nanoparticle (LNP)-based carriers. With the development of mRNA-LNP modalities, which can incorporate xenobiotic constituents, extensive biodistribution analyses are necessary to better understand the factors that influence their in vivo exposure profiles. This study investigated the biodistribution of heptadecan-9-yl 8-((2-hydroxyethyl)(8-(nonyloxy)-8-oxooctyl)amino)octanoate (Lipid 5)-a xenobiotic amino lipid-and its metabolites in male and female pigmented (Long-Evans) and nonpigmented (Sprague Dawley) rats by using quantitative whole-body autoradiography (QWBA) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) techniques. After intravenous injection of Lipid 5-containing LNPs, 14C-containing Lipid 5 ([14C]Lipid 5) and radiolabeled metabolites ([14C]metabolites) were rapidly distributed, with peak concentrations reached within 1 hour in most tissues. After 10 hours, [14C]Lipid 5 and [14C]metabolites concentrated primarily in the urinary and digestive tracts. By 24 hours, [14C]Lipid 5 and [14C]metabolites were localized almost exclusively in the liver and intestines, with few or no concentrations detected in non-excretory systems, which is suggestive of hepatobiliary and renal clearance. [14C]Lipid 5 and [14C]metabolites were completely cleared within 168 hours (7 days). Biodistribution profiles were similar between QWBA and LC-MS/MS techniques, pigmented and nonpigmented rats, and male and female rats, excluding the reproductive organs. In conclusion, the rapid clearance through known excretory systems, with no evidence of redistribution for Lipid 5 or accumulation of [14C]metabolites, provides confidence for the safe and effective use of Lipid 5-containing LNPs. SIGNIFICANCE STATEMENT: This study demonstrates the rapid, systemic distribution of intact and radiolabeled metabolites of Lipid 5, a xenobiotic amino lipid component of novel mRNA-LNP medicines, and its effective clearance without substantial redistribution after intravenous administration; additionally, findings were consistent between different mRNAs encapsulated within LNPs of similar composition. This study confirms the applicability of current analytical methods for lipid biodistribution analyses, and taken together with appropriate safety studies, supports the continued use of Lipid 5 in mRNA-medicines.
Collapse
Affiliation(s)
- Lei Ci
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Marjie Hard
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Hannah Zhang
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Srujan Gandham
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Serenus Hua
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - John Wickwire
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Tod Wehrman
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Richard Slauter
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Andrew Auerbach
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Matthew Kenney
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Greg Mercer
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Tracy Hendrick
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Örn Almarsson
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Eugene Cheung
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| | - Douglas Burdette
- Moderna, Inc., Cambridge, Massachusetts (L.C., H.Z., S.G., S.H., J.W., A.A., M.K., G.M., E.C., D.B.); Praxis Precision Medicines, Boston, Massachusetts (M.H.); Charles River Laboratories, Mattawan, Michigan (T.W., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (O.A.)
| |
Collapse
|
24
|
Castruita JAS, Schneider UV, Mollerup S, Leineweber TD, Weis N, Bukh J, Pedersen MS, Westh H. SARS-CoV-2 spike mRNA vaccine sequences circulate in blood up to 28 days after COVID-19 vaccination. APMIS 2023; 131:128-132. [PMID: 36647776 PMCID: PMC10107710 DOI: 10.1111/apm.13294] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023]
Abstract
In Denmark, vaccination against Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) has been with the Pfizer-BioNTech (BTN162b2) or the Moderna (mRNA-1273) mRNA vaccines. Patients with chronic hepatitis C virus (HCV) infection followed in our clinic received mRNA vaccinations according to the Danish roll-out vaccination plan. To monitor HCV infection, RNA was extracted from patient plasma and RNA sequencing was performed on the Illumina platform. In 10 of 108 HCV patient samples, full-length or traces of SARS-CoV-2 spike mRNA vaccine sequences were found in blood up to 28 days after COVID-19 vaccination. Detection of mRNA vaccine sequences in blood after vaccination adds important knowledge regarding this technology and should lead to further research into the design of lipid-nanoparticles and the half-life of these and mRNA vaccines in humans.
Collapse
Affiliation(s)
| | - Uffe Vest Schneider
- Department of Clinical Microbiology, Copenhagen University Hospital Amager-Hvidovre, Hvidovre, Denmark
| | - Sarah Mollerup
- Department of Clinical Microbiology, Copenhagen University Hospital Amager-Hvidovre, Hvidovre, Denmark
| | - Thomas Daell Leineweber
- Department of Clinical Microbiology, Copenhagen University Hospital Amager-Hvidovre, Hvidovre, Denmark
| | - Nina Weis
- Department of Infectious Diseases, Copenhagen University Hospital Amager-Hvidovre, Hvidovre, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bukh
- Department of Infectious Diseases, Copenhagen University Hospital Amager-Hvidovre, Hvidovre, Denmark.,Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin Schou Pedersen
- Department of Clinical Microbiology, Copenhagen University Hospital Amager-Hvidovre, Hvidovre, Denmark.,Department of Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Henrik Westh
- Department of Clinical Microbiology, Copenhagen University Hospital Amager-Hvidovre, Hvidovre, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
von der Haar T, Mulroney TE, Hedayioglu F, Kurusamy S, Rust M, Lilley KS, Thaventhiran JE, Willis AE, Smales CM. Translation of in vitro-transcribed RNA therapeutics. Front Mol Biosci 2023; 10:1128067. [PMID: 36845540 PMCID: PMC9943971 DOI: 10.3389/fmolb.2023.1128067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
In vitro transcribed, modified messenger RNAs (IVTmRNAs) have been used to vaccinate billions of individuals against the SARS-CoV-2 virus, and are currently being developed for many additional therapeutic applications. IVTmRNAs must be translated into proteins with therapeutic activity by the same cellular machinery that also translates native endogenous transcripts. However, different genesis pathways and routes of entry into target cells as well as the presence of modified nucleotides mean that the way in which IVTmRNAs engage with the translational machinery, and the efficiency with which they are being translated, differs from native mRNAs. This review summarises our current knowledge of commonalities and differences in translation between IVTmRNAs and cellular mRNAs, which is key for the development of future design strategies that can generate IVTmRNAs with improved activity in therapeutic applications.
Collapse
Affiliation(s)
- Tobias von der Haar
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, United Kingdom
| | - Thomas E. Mulroney
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, United Kingdom
| | - Fabio Hedayioglu
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, United Kingdom
| | - Sathishkumar Kurusamy
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, United Kingdom
| | - Maria Rust
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, United Kingdom
| | - Kathryn S. Lilley
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - James E. Thaventhiran
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, United Kingdom
| | - Anne E. Willis
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, United Kingdom
| | - C. Mark Smales
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
26
|
Chavda VP, Soni S, Vora LK, Soni S, Khadela A, Ajabiya J. mRNA-Based Vaccines and Therapeutics for COVID-19 and Future Pandemics. Vaccines (Basel) 2022; 10:2150. [PMID: 36560560 PMCID: PMC9785933 DOI: 10.3390/vaccines10122150] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
An unheard mobilization of resources to find SARS-CoV-2 vaccines and therapies has been sparked by the COVID-19 pandemic. Two years ago, COVID-19's launch propelled mRNA-based technologies into the public eye. Knowledge gained from mRNA technology used to combat COVID-19 is assisting in the creation of treatments and vaccines to treat existing illnesses and may avert pandemics in the future. Exploiting the capacity of mRNA to create therapeutic proteins to impede or treat a variety of illnesses, including cancer, is the main goal of the quickly developing, highly multidisciplinary field of biomedicine. In this review, we explore the potential of mRNA as a vaccine and therapeutic using current research findings.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, LM College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Shailvi Soni
- Massachussets College of Pharmacy and Health Science, 19 Foster Street, Worcester, MA 01608, USA
| | - Lalitkumar K. Vora
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Shruti Soni
- PharmD Section, LM College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Avinash Khadela
- Department of Pharmacology, LM College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Jinal Ajabiya
- Department of Pharmaceutics Analysis and Quality Assurance, LM College of Pharmacy, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
27
|
Zadory M, Lopez E, Babity S, Gravel SP, Brambilla D. Current knowledge on the tissue distribution of mRNA nanocarriers for therapeutic protein expression. Biomater Sci 2022; 10:6077-6115. [PMID: 36097955 DOI: 10.1039/d2bm00859a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Exogenously delivered mRNA-based drugs are emerging as a new class of therapeutics with the potential to treat several diseases. Over the last decade, advancements in the design of non-viral delivery tools have enabled mRNA to be evaluated for several therapeutic purposes including protein replacement therapies, gene editing, and vaccines. However, in vivo delivery of mRNA to targeted organs and cells remains a critical challenge. Evaluation of the biodistribution of mRNA vehicles is of utmost importance for the development of effective pharmaceutical candidates. In this review, we discuss the recent advances in the design of nanoparticles loaded with mRNA and extrapolate the key factors influencing their biodistribution following administration. Finally, we highlight the latest developments in the preclinical and clinical translation of mRNA therapeutics for protein supplementation therapy.
Collapse
Affiliation(s)
- Matthias Zadory
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Elliot Lopez
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Samuel Babity
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Simon-Pierre Gravel
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Davide Brambilla
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| |
Collapse
|
28
|
The Delivery of mRNA Vaccines for Therapeutics. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081254. [PMID: 36013433 PMCID: PMC9410089 DOI: 10.3390/life12081254] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 12/12/2022]
Abstract
mRNA vaccines have been revolutionary in combating the COVID-19 pandemic in the past two years. They have also become a versatile tool for the prevention of infectious diseases and treatment of cancers. For effective vaccination, mRNA formulation, delivery method and composition of the mRNA carrier play an important role. mRNA vaccines can be delivered using lipid nanoparticles, polymers, peptides or naked mRNA. The vaccine efficacy is influenced by the appropriate delivery materials, formulation methods and selection of a proper administration route. In addition, co-delivery of several mRNAs could also be beneficial and enhance immunity against various variants of an infectious pathogen or several pathogens altogether. Here, we review the recent progress in the delivery methods, modes of delivery and patentable mRNA vaccine technologies.
Collapse
|
29
|
Medjmedj A, Ngalle-Loth A, Clemençon R, Hamacek J, Pichon C, Perche F. In Cellulo and In Vivo Comparison of Cholesterol, Beta-Sitosterol and Dioleylphosphatidylethanolamine for Lipid Nanoparticle Formulation of mRNA. NANOMATERIALS 2022; 12:nano12142446. [PMID: 35889670 PMCID: PMC9317807 DOI: 10.3390/nano12142446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 12/10/2022]
Abstract
Lipid Nanoparticles (LNPs) are a leading class of mRNA delivery systems. LNPs are made of an ionizable lipid, a polyethyleneglycol (PEG)-lipid conjugate and helper lipids. The success of LNPs is due to proprietary ionizable lipids and appropriate helper lipids. Using a benchmark lipid (D-Lin-MC3) we compared the ability of three helper lipids to transfect dendritic cells in cellulo and in vivo. Studies revealed that the choice of helper lipid does not influence the transfection efficiency of immortalized cells but, LNPs prepared with DOPE (dioleylphosphatidylethanolamine) and β-sitosterol were more efficient for mRNA transfection in murine dendritic cells than LNPs containing DSPC (distearoylphosphatidylcholine). This higher potency of DOPE and β-sitosterol LNPs for mRNA expression was also evident in vivo but only at low mRNA doses. Overall, these data provide valuable insight for the design of novel mRNA LNP vaccines.
Collapse
Affiliation(s)
- Ayoub Medjmedj
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
| | - Albert Ngalle-Loth
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
| | - Rudy Clemençon
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
| | - Josef Hamacek
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
- Centre de Biophysique Moléculaire, University of Orléans, 45100 Orléans, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
- Centre de Biophysique Moléculaire, University of Orléans, 45100 Orléans, France
| | - Federico Perche
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans, France; (A.M.); (A.N.-L.); (R.C.); (J.H.); (C.P.)
- Correspondence: ; Tel.: +33-2-38-25-55-44
| |
Collapse
|
30
|
Kloczewiak M, Banks JM, Jin L, Brader ML. A Biopharmaceutical Perspective on Higher-Order Structure and Thermal Stability of mRNA Vaccines. Mol Pharm 2022; 19:2022-2031. [PMID: 35715255 PMCID: PMC9257798 DOI: 10.1021/acs.molpharmaceut.2c00092] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/27/2022]
Abstract
Preservation of the integrity of macromolecular higher-order structure is a tenet central to achieving biologic drug and vaccine product stability toward manufacturing, distribution, storage, handling, and administration. Given that mRNA lipid nanoparticles (mRNA-LNPs) are held together by an intricate ensemble of weak forces, there are some intriguing parallels to biologic drugs, at least at first glance. However, mRNA vaccines are not without unique formulation and stabilization challenges derived from the instability of unmodified mRNA and its limited history as a drug or vaccine. Since certain learning gained from biologic drug development may be applicable for the improvement of mRNA vaccines, we present a perspective on parallels and contrasts between the emerging role of higher-order structure pertaining to mRNA-LNPs compared to pharmaceutical proteins. In a recent publication, the location of mRNA encapsulated within lipid nanoparticles was identified, revealing new insights into the LNP structure, nanoheterogeneity, and microenvironment of the encapsulated mRNA molecules [Brader et al. Biophys. J. 2021, 120, 2766]. We extend those findings by considering the effect of encapsulation on mRNA thermal unfolding with the observation that encapsulation in LNPs increases mRNA unfolding temperatures.
Collapse
Affiliation(s)
- Marek Kloczewiak
- Moderna, Inc., 200 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Jessica M. Banks
- Moderna, Inc., 200 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Lin Jin
- Moderna, Inc., 200 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Mark L. Brader
- Moderna, Inc., 200 Technology Square, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
31
|
Lee J, Kim D, Byun J, Wu Y, Park J, Oh YK. In vivo fate and intracellular trafficking of vaccine delivery systems. Adv Drug Deliv Rev 2022; 186:114325. [PMID: 35550392 PMCID: PMC9085465 DOI: 10.1016/j.addr.2022.114325] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 01/12/2023]
Abstract
With the pandemic of severe acute respiratory syndrome coronavirus 2, vaccine delivery systems emerged as a core technology for global public health. Given that antigen processing takes place inside the cell, the intracellular delivery and trafficking of a vaccine antigen will contribute to vaccine efficiency. Investigations focusing on the in vivo behavior and intracellular transport of vaccines have improved our understanding of the mechanisms relevant to vaccine delivery systems and facilitated the design of novel potent vaccine platforms. In this review, we cover the intracellular trafficking and in vivo fate of vaccines administered via various routes and delivery systems. To improve immune responses, researchers have used various strategies to modulate vaccine platforms and intracellular trafficking. In addition to progress in vaccine trafficking studies, the challenges and future perspectives for designing next-generation vaccines are discussed.
Collapse
Affiliation(s)
- Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Dongyoon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Junho Byun
- 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
| | - Jinwon Park
- 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
|
32
|
Ezra Manicum AL, Sargazi S, Razzaq S, Kumar GV, Rahdar A, Er S, Ain QU, Bilal M, Aboudzadeh MA. Nano-immunotherapeutic strategies for targeted RNA delivery: Emphasizing the role of monocyte/macrophages as nanovehicles to treat glioblastoma multiforme. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
33
|
Uchida S. Delivery Systems of Plasmid DNA and Messenger RNA for Advanced Therapies. Pharmaceutics 2022; 14:pharmaceutics14040810. [PMID: 35456642 PMCID: PMC9029576 DOI: 10.3390/pharmaceutics14040810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/05/2022] [Indexed: 12/10/2022] Open
Abstract
The vast potential of non-viral delivery systems of messenger RNA (mRNA) and plasmid DNA (pDNA) has been demonstrated in the vaccines against coronavirus disease 2019 (COVID-19) [...]
Collapse
Affiliation(s)
- Satoshi Uchida
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 606-0823, Japan;
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki 210-0821, Japan
| |
Collapse
|
34
|
Daniel S, Kis Z, Kontoravdi C, Shah N. Quality by Design for enabling RNA platform production processes. Trends Biotechnol 2022; 40:1213-1228. [DOI: 10.1016/j.tibtech.2022.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/18/2022] [Accepted: 03/28/2022] [Indexed: 12/26/2022]
|
35
|
Nanoparticle-based delivery strategies of multifaceted immunomodulatory RNA for cancer immunotherapy. J Control Release 2022; 343:564-583. [DOI: 10.1016/j.jconrel.2022.01.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 12/18/2022]
|
36
|
Morán L, Woitok MM, Bartneck M, Cubero FJ. Hepatocyte-Directed Delivery of Lipid-Encapsulated Small Interfering RNA. Methods Mol Biol 2022; 2544:95-106. [PMID: 36125712 DOI: 10.1007/978-1-0716-2557-6_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Lipid formulations for cell transfection are among the most efficient systems for nucleic acid delivery. During the COVID-19 pandemic, lipid-encapsulated RNA (lipid nanoparticles, LNP) has succeeded as a superior vaccine. Moreover, other similar lipid nanocarriers for siRNA are approved and many are on the pipelines. While lipid encapsulation required several devices for the mixing of components, lipoplex technology allows to rapidly mix nucleic acids and positively charged lipids for cell transfection. In vivo, hepatocytes are important target cells of lipid formulated RNAi. This chapter describes the state-of-the-art lipoplex and LPN manufacturing for treating primary hepatocytes with lipid formulations. Furthermore, protocols for isolating murine hepatocytes and for transfecting these cells with pharmaceutically relevant lipid formulations are provided and discussed.
Collapse
Affiliation(s)
- Laura Morán
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | | | - Matthias Bartneck
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
37
|
Baptista B, Carapito R, Laroui N, Pichon C, Sousa F. mRNA, a Revolution in Biomedicine. Pharmaceutics 2021; 13:2090. [PMID: 34959371 PMCID: PMC8707022 DOI: 10.3390/pharmaceutics13122090] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 12/23/2022] Open
Abstract
The perspective of using messenger RNA (mRNA) as a therapeutic molecule first faced some uncertainties due to concerns about its instability and the feasibility of large-scale production. Today, given technological advances and deeper biomolecular knowledge, these issues have started to be addressed and some strategies are being exploited to overcome the limitations. Thus, the potential of mRNA has become increasingly recognized for the development of new innovative therapeutics, envisioning its application in immunotherapy, regenerative medicine, vaccination, and gene editing. Nonetheless, to fully potentiate mRNA therapeutic application, its efficient production, stabilization and delivery into the target cells are required. In recent years, intensive research has been carried out in this field in order to bring new and effective solutions towards the stabilization and delivery of mRNA. Presently, the therapeutic potential of mRNA is undoubtedly recognized, which was greatly reinforced by the results achieved in the battle against the COVID-19 pandemic, but there are still some issues that need to be improved, which are critically discussed in this review.
Collapse
Affiliation(s)
- Bruno Baptista
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (B.B.); (R.C.)
| | - Rita Carapito
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (B.B.); (R.C.)
| | - Nabila Laroui
- Centre de Biophysique Moléculaire (CBM), UPR 4301 CNRS, University of Orléans, 45071 Orléans, France;
| | - Chantal Pichon
- Centre de Biophysique Moléculaire (CBM), UPR 4301 CNRS, University of Orléans, 45071 Orléans, France;
| | - Fani Sousa
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (B.B.); (R.C.)
| |
Collapse
|