1
|
Chai D, Wang J, Lim JM, Xie X, Yu X, Zhao D, Maza PAM, Wang Y, Cyril-Remirez D, Young KH, Li Y. Lipid nanoparticles deliver DNA-encoded biologics and induce potent protective immunity. Mol Cancer 2025; 24:12. [PMID: 39806486 PMCID: PMC11727718 DOI: 10.1186/s12943-024-02211-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025] Open
Abstract
Lipid nanoparticles (LNPs) for mRNA delivery have advanced significantly, but LNP-mediated DNA delivery still faces clinical challenges. This study compared various LNP formulations for delivering DNA-encoded biologics, assessing their expression efficacy and the protective immunity generated by LNP-encapsulated DNA in different models. The LNP formulation used in Moderna's Spikevax mRNA vaccine (LNP-M) demonstrated a stable nanoparticle structure, high expression efficiency, and low toxicity. Notably, a DNA vaccine encoding the spike protein, delivered via LNP-M, induced stronger antigen-specific antibody and T cell immune responses compared to electroporation. Single-cell RNA sequencing (scRNA-seq) analysis revealed that the LNP-M/pSpike vaccine enhanced CD80 activation signaling in CD8+ T cells, NK cells, macrophages, and DCs, while reducing the immunosuppressive signals. The enrichment of TCR and BCR by LNP-M/pSpike suggested an increase in immune response specificity and diversity. Additionally, LNP-M effectively delivered DNA-encoded antigens, such as mouse PD-L1 and p53R172H, or monoclonal antibodies targeting mouse PD1 and human p53R282W. This approach inhibited tumor growth or metastasis in several mouse models. The long-term anti-tumor effects of LNP-M-delivered anti-p53R282W antibody relied on memory CD8+ T cell responses and enhanced MHC-I signaling from APCs to CD8+ T cells. These results highlight LNP-M as a promising and effective platform for delivering DNA-based vaccines and cancer immunotherapies.
Collapse
Affiliation(s)
- Dafei Chai
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Junhao Wang
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jing Ming Lim
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiaohui Xie
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xinfang Yu
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dan Zhao
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Perry Ayn Mayson Maza
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yifei Wang
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dana Cyril-Remirez
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ken H Young
- Department of Pathology, Division of Hematopathology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Yong Li
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
He X, Payne TJ, Takanashi A, Fang Y, Kerai SD, Morrow JP, Al-Wassiti H, Pouton CW, Kempe K. Tailored Monoacyl Poly(2-oxazoline)- and Poly(2-oxazine)-Lipids as PEG-Lipid Alternatives for Stabilization and Delivery of mRNA-Lipid Nanoparticles. Biomacromolecules 2024; 25:4591-4603. [PMID: 38918933 DOI: 10.1021/acs.biomac.4c00651] [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: 06/27/2024]
Abstract
The successful use of lipid nanoparticles (LNPs) for clinical development of the COVID-19 mRNA vaccines marked a breakthrough in mRNA-LNP therapeutics. As one of the vital components of LNPs, poly(ethylene glycol)-lipid conjugates (PEG-lipids) influence particle biophysical properties and stability, as well as interactions within biological environments. Reports suggesting that anti-PEG antibodies can be detected quite commonly within the human population raise concerns that PEG content in commercial LNP products could further stimulate immune responses to PEG. The presence of anti-PEG antibodies has been linked to accelerated clearance of LNPs, potentially a source of variability in the biological response to mRNA-LNP products. This motivated us to explore potential PEG alternatives. Herein, we report physicochemical and biological properties of mRNA-LNPs assembled using poly(2-oxazoline) (POx)- and poly(2-oxazine) (POz)-based polymer-lipid conjugates. Notably, we investigated monoacyl lipids as alternatives to diacyl lipids. mRNA-LNPs produced using monoacyl POx/POz-lipids displayed comparable biophysical characteristics and cytocompatibility. Delivery of reporter mRNA resulted in similar transfection efficiencies, in both adherent and suspension cells, and in mice, compared to PEG-lipid equivalents. Our results suggest that monoacyl POx/POz-lipid-containing LNPs are promising candidates for the development of PEG-free LNP-based therapeutic products.
Collapse
Affiliation(s)
- Xie He
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Thomas J Payne
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Asuka Takanashi
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Yunji Fang
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Simran D Kerai
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Joshua P Morrow
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Hareth Al-Wassiti
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Colin W Pouton
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
- Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
3
|
De Peña AC, Zimmer D, Gutterman-Johns E, Chen NM, Tripathi A, Bailey-Hytholt CM. Electrophoretic Microfluidic Characterization of mRNA- and pDNA-Loaded Lipid Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2024; 16:26984-26997. [PMID: 38753459 DOI: 10.1021/acsami.4c00208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Lipid nanoparticles (LNPs) are clinically advanced nonviral gene delivery vehicles with a demonstrated ability to address viral, oncological, and genetic diseases. However, the further development of LNP therapies requires rapid analytical techniques to support their development and manufacturing. The method developed and described in this paper presents an approach to rapidly and accurately analyze LNPs for optimized therapeutic loading by utilizing an electrophoresis microfluidic platform to analyze the composition of LNPs with different clinical lipid compositions (Onpattro, Comirnaty, and Spikevax) and nucleic acid (plasmid DNA (pDNA) and messenger RNA (mRNA)) formulations. This method enables the high-throughput screening of LNPs using a 96- or 384-well plate with approximate times of 2-4 min per sample using a total volume of 11 μL. The lipid analysis requires concentrations approximately between 109 and 1010 particles/mL and has an average precision error of 10.4% and a prediction error of 19.1% when compared to using a NanoSight, while the nucleic acid analysis requires low concentrations of 1.17 ng/μL for pDNA and 0.17 ng/μL for mRNA and has an average precision error of 4.8% and a prediction error of 9.4% when compared to using a PicoGreen and RiboGreen assay. In addition, our method quantifies the relative concentration of nucleic acid per LNP. Utilizing this approach, we observed an average of 263 ± 62.2 mRNA per LNP and 126.3 ± 21.2 pDNA per LNP for the LNP formulations used in this study, where the accuracy of these estimations is dependent on reference standards. We foresee the utility of this technique in the high-throughput characterization of LNPs during manufacturing and formulation research and development.
Collapse
Affiliation(s)
- Adriana Coll De Peña
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Daniel Zimmer
- Department of Chemical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, United States
| | - Everett Gutterman-Johns
- Department of Molecular Biology, Cell Biology, and Biochemistry, Division of Biology and Medicine, Brown University, Providence, Rhode Island 02912, United States
| | - Nicole M Chen
- Department of Molecular Biology, Cell Biology, and Biochemistry, Division of Biology and Medicine, Brown University, Providence, Rhode Island 02912, United States
| | - Anubhav Tripathi
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Christina M Bailey-Hytholt
- Department of Chemical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, United States
| |
Collapse
|
4
|
Jaradat E, Weaver E, Meziane A, Lamprou DA. Synthesis and Characterization of Paclitaxel-Loaded PEGylated Liposomes by the Microfluidics Method. Mol Pharm 2023; 20:6184-6196. [PMID: 37931072 DOI: 10.1021/acs.molpharmaceut.3c00596] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
For cancer therapy, paclitaxel (PX) possesses several limitations, including limited solubility and untargeted effects. Loading PX into nanoliposomes to enhance PX solubility and target their delivery as a drug delivery system has the potential to overcome these limitations. Over the other conventional method to prepare liposomes, a microfluidic system is used to formulate PX-loaded PEGylated liposomes. The impact of changing the flow rate ratio (FRR) between the aqueous and lipid phases on the particle size and polydispersity index (PDI) is investigated. Moreover, the effect of changing the polyethylene glycol (PEG) lipid ratio on the particle size, PDI, stability, encapsulation efficiency % (EE %), and release profile is studied. The physicochemical characteristics of the obtained formulation were analyzed by dynamic light scattering, FTIR spectroscopy, and AFM. This work aims to use microfluidic technology to produce PEGylated PX-loaded liposomes with a diameter of <200 nm, low PDI < 0.25 high homogeneity, and viable 28 day stability. The results show a significant impact of FRR and PEG lipid ratio on the empty liposomes' physicochemical characteristics. Among the prepared formulations, two formulations produce size-controlled, low PDI, and stable liposomes, which make them preferable for PX encapsulation. The average EE % was >90% for both formulations, and the variation in the PEG lipid ratio affected the EE % slightly; a high packing for PX was reported at different drug concentrations. A variation in the release profiles was notified for the different PEG lipid ratios.
Collapse
Affiliation(s)
- Eman Jaradat
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL Belfast, U.K
| | - Edward Weaver
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL Belfast, U.K
| | | | - Dimitrios A Lamprou
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL Belfast, U.K
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Li D, Liu Q, Yang M, Xu H, Zhu M, Zhang Y, Xu J, Tian C, Yao J, Wang L, Liang Y. Nanomaterials for mRNA-based therapeutics: Challenges and opportunities. Bioeng Transl Med 2023; 8:e10492. [PMID: 37206219 PMCID: PMC10189457 DOI: 10.1002/btm2.10492] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/31/2023] Open
Abstract
Messenger RNA (mRNA) holds great potential in developing immunotherapy, protein replacement, and genome editing. In general, mRNA does not have the risk of being incorporated into the host genome and does not need to enter the nucleus for transfection, and it can be expressed even in nondividing cells. Therefore, mRNA-based therapeutics provide a promising strategy for clinical treatment. However, the efficient and safe delivery of mRNA remains a crucial constraint for the clinical application of mRNA therapeutics. Although the stability and tolerability of mRNA can be enhanced by directly retouching the mRNA structure, there is still an urgent need to improve the delivery of mRNA. Recently, significant progress has been made in nanobiotechnology, providing tools for developing mRNA nanocarriers. Nano-drug delivery system is directly used for loading, protecting, and releasing mRNA in the biological microenvironment and can be used to stimulate the translation of mRNA to develop effective intervention strategies. In the present review, we summarized the concept of emerging nanomaterials for mRNA delivery and the latest progress in enhancing the function of mRNA, primarily focusing on the role of exosomes in mRNA delivery. Moreover, we outlined its clinical applications so far. Finally, the key obstacles of mRNA nanocarriers are emphasized, and promising strategies to overcome these obstacles are proposed. Collectively, nano-design materials exert functions for specific mRNA applications, provide new perception for next-generation nanomaterials, and thus revolution of mRNA technology.
Collapse
Affiliation(s)
- De‐feng Li
- Department of GastroenterologyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Qi‐song Liu
- National Clinical Research Center for Infectious DiseasesShenzhen Third People's Hospital, Southern University of Science and TechnologyShenzhenChina
| | - Mei‐feng Yang
- Department of HematologyYantian District People's HospitalShenzhenGuangdongChina
| | - Hao‐ming Xu
- Department of Gastroenterology and HepatologyGuangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Min‐zheng Zhu
- Department of Gastroenterology and Hepatologythe Second Affiliated Hospital, School of Medicine, South China University of TechnologyGuangzhouGuangdongChina
| | - Yuan Zhang
- Department of Medical AdministrationHuizhou Institute of Occupational Diseases Control and PreventionHuizhouGuangdongChina
| | - Jing Xu
- Department of Gastroenterology and HepatologyGuangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Cheng‐mei Tian
- Department of EmergencyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Jun Yao
- Department of GastroenterologyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Li‐sheng Wang
- Department of GastroenterologyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Yu‐jie Liang
- Department of Child and Adolescent PsychiatryShenzhen Kangning Hospital, Shenzhen Mental Health CenterShenzhenChina
- Affiliated Hospital of Jining Medical University, Jining Medical UniversityJiningShandongChina
| |
Collapse
|
8
|
De A, Ko YT. A tale of nucleic acid-ionizable lipid nanoparticles: Design and manufacturing technology and advancement. Expert Opin Drug Deliv 2023; 20:75-91. [PMID: 36445261 DOI: 10.1080/17425247.2023.2153832] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
INTRODUCTION Ionizable lipid nanoparticles (LNPs) have been proven to have high encapsulation, cellular uptake, and effective endosomal escape and are therefore promising for nucleic acid delivery. The combination of ionizable lipids, helper lipids, cholesterol, and PEG lipids advances nucleic acid-ionizable LNPs and distinguishes them from liposomes, SLNs, NLCs, and other lipid particles. Solvent injection and microfluidics technology are the primary manufacturing techniques for commercialized ionizable LNPs. Microfluidics technology limitations restrict the rapid industrial scale-up and therapeutic effectiveness of ionized LNPs. Alternative manufacturing technologies and target-specific lipids are urgently needed. AREA COVERED This article provides an in-depth update on the lipid compositions, clinical trials, and manufacturing technologies for nucleic acid-ionizable LNPs. For the first time, we updated the distinction between ionizable LNPs and other lipid particles. We also proposed an alternate thermocycling technology for high industrial scale-up and the stability of nucleic acid-ionizing LNPs. EXPERT OPINION Nucleic acid-ionizable LNPs have a promising future for delivering nucleic acids in a target-specific manner. Though ionizing LNPs are in their early stages, they face several challenges, including only hepatic delivery, a short shelf life, and ultra-cold storage. In our opinion, ligand-based, target-specific synthesized novel lipids and advanced manufacturing technologies can easily overcome the restrictions and open up a new approach for improved therapeutic efficacy for chronic disorders.
Collapse
Affiliation(s)
- Anindita De
- College of Pharmacy, Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, South Korea
| | - Young Tag Ko
- College of Pharmacy, Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, South Korea
| |
Collapse
|
9
|
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
|
10
|
Gu Y, Duan J, Yang N, Yang Y, Zhao X. mRNA vaccines in the prevention and treatment of diseases. MedComm (Beijing) 2022; 3:e167. [PMID: 36033422 PMCID: PMC9409637 DOI: 10.1002/mco2.167] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/11/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Messenger ribonucleic acid (mRNA) vaccines made their successful public debut in the effort against the COVID-19 outbreak starting in late 2019, although the history of mRNA vaccines can be traced back decades. This review provides an overview to discuss the historical course and present situation of mRNA vaccine development in addition to some basic concepts that underly mRNA vaccines. We discuss the general preparation and manufacturing of mRNA vaccines and also discuss the scientific advances in the in vivo delivery system and evaluate popular approaches (i.e., lipid nanoparticle and protamine) in detail. Next, we highlight the clinical value of mRNA vaccines as potent candidates for therapeutic treatment and discuss clinical progress in the treatment of cancer and coronavirus disease 2019. Data suggest that mRNA vaccines, with several prominent advantages, have achieved encouraging results and increasing attention due to tremendous potential in disease management. Finally, we suggest some potential directions worthy of further investigation and optimization. In addition to basic research, studies that help to facilitate storage and transportation will be indispensable for practical applications.
Collapse
Affiliation(s)
- Yangzhuo Gu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University; Collaborative Innovation Center for BiotherapyChengduChina
| | - Jiangyao Duan
- Department of Life SciencesImperial College LondonLondonUK
| | - Na Yang
- Stem Cell and Tissue Engineering Research Center/School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
| | - Yuxin Yang
- Stem Cell and Tissue Engineering Research Center/School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
| | - Xing Zhao
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University; Collaborative Innovation Center for BiotherapyChengduChina
- Stem Cell and Tissue Engineering Research Center/School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
11
|
Multiscale Dynamics of Lipid Vesicles in Polymeric Microenvironment. MEMBRANES 2022; 12:membranes12070640. [PMID: 35877843 PMCID: PMC9318666 DOI: 10.3390/membranes12070640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/15/2022] [Accepted: 06/18/2022] [Indexed: 02/06/2023]
Abstract
Understanding dynamic and complex interaction of biological membranes with extracellular matrices plays a crucial role in controlling a variety of cell behavior and functions, from cell adhesion and growth to signaling and differentiation. Tremendous interest in tissue engineering has made it possible to design polymeric scaffolds mimicking the topology and mechanical properties of the native extracellular microenvironment; however, a fundamental question remains unanswered: that is, how the viscoelastic extracellular environment modifies the hierarchical dynamics of lipid membranes. In this work, we used aqueous solutions of poly(ethylene glycol) (PEG) with different molecular weights to mimic the viscous medium of cells and nearly monodisperse unilamellar DMPC/DMPG liposomes as a membrane model. Using small-angle X-ray scattering (SAXS), dynamic light scattering, temperature-modulated differential scanning calorimetry, bulk rheology, and fluorescence lifetime spectroscopy, we investigated the structural phase map and multiscale dynamics of the liposome–polymer mixtures. The results suggest an unprecedented dynamic coupling between polymer chains and phospholipid bilayers at different length/time scales. The microviscosity of the lipid bilayers is directly influenced by the relaxation of the whole chain, resulting in accelerated dynamics of lipids within the bilayers in the case of short chains compared to the polymer-free liposome case. At the macroscopic level, the gel-to-fluid transition of the bilayers results in a remarkable thermal-stiffening behavior of polymer–liposome solutions that can be modified by the concentration of the liposomes and the polymer chain length.
Collapse
|
12
|
Li J, Kalyanram P, Rozati S, Monje-Galvan V, Gupta A. Interaction of Cyanine-D112 with Binary Lipid Mixtures: Molecular Dynamics Simulation and Differential Scanning Calorimetry Study. ACS OMEGA 2022; 7:9765-9774. [PMID: 35350357 PMCID: PMC8945139 DOI: 10.1021/acsomega.1c07378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
This comprehensive molecular dynamics (MD) simulation and experimental study investigates the lipid bilayer interactions of dye D112 for potential photodynamic therapy (PDT) applications. PDT involves formation of a reactive oxidant species in the presence of a light sensitive molecule and light, interrupting cellular functions. D112 was developed as a photographic emulsifier, and we hypothesized that its combined cationic and lipophilic nature can render a superior photosensitizing property-crucial in various light therapies. The focus of this study is to elucidate the binding and insertion mechanisms of D112 with mixed lipid bilayers of anionic dipalmitoyl-phosphatidylserine (DPPS) and zwitterionic dipalmitoyl-phosphatidylcholine (DPPC) lipids to resemble cancer cell membranes. Our studies confirm initial electrostatic binding between the positively charged moieties of D112 and negatively charged lipid headgroups. Additionally, MD simulations combined with differential scanning calorimetry (DSC) studies confirm that D112-lipid interactions are governed by enthalpy-driven nonclassical hydrophobic effects in the membrane interior. It was further noted that despite the electrostatic preference of D112 toward the anionic lipids, D112 molecules colocalized on DPPC-rich domains after insertion. Atomistic level MD studies point toward two possible insertion mechanisms for D112: harpoon and flip. Further insights from the simulation showcase the interactions of low and high aggregates of D112 with the bilayer as the concentration of D112 increases in solution. The size of aggregates modulates the orientation and degree of insertion, providing important information for future studies on membrane permeation mechanisms.
Collapse
Affiliation(s)
- Jinhui Li
- Department
of Chemical and Biological Engineering, University at Buffalo, Buffalo, New York 14260, United States
| | - Poornima Kalyanram
- Department
of Mechanical, Industrial and Manufacturing Engineering, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Seyedalireza Rozati
- Department
of Mechanical, Industrial and Manufacturing Engineering, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Viviana Monje-Galvan
- Department
of Chemical and Biological Engineering, University at Buffalo, Buffalo, New York 14260, United States
| | - Anju Gupta
- Department
of Mechanical, Industrial and Manufacturing Engineering, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| |
Collapse
|
13
|
Biophysical Characterization of Viral and Lipid-Based Vectors for Vaccines and Therapeutics with Light Scattering and Calorimetric Techniques. Vaccines (Basel) 2021; 10:vaccines10010049. [PMID: 35062710 PMCID: PMC8780473 DOI: 10.3390/vaccines10010049] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 01/01/2023] Open
Abstract
Novel vaccine platforms for delivery of nucleic acids based on viral and non-viral vectors, such as recombinant adeno associated viruses (rAAV) and lipid-based nanoparticles (LNPs), hold great promise. However, they pose significant manufacturing and analytical challenges due to their intrinsic structural complexity. During product development and process control, their design, characterization, and quality control require the combination of fit-for-purpose complementary analytical tools. Moreover, an in-depth methodological expertise and holistic approach to data analysis are required for robust measurements and to enable an adequate interpretation of experimental findings. Here the combination of complementary label-free biophysical techniques, including dynamic light scattering (DLS), multiangle-DLS (MADLS), Electrophoretic Light Scattering (ELS), nanoparticle tracking analysis (NTA), multiple detection SEC and differential scanning calorimetry (DSC), have been successfully used for the characterization of physical and chemical attributes of rAAV and LNPs encapsulating mRNA. Methods' performance, applicability, dynamic range of detection and method optimization are discussed for the measurements of multiple critical physical-chemical quality attributes, including particle size distribution, aggregation propensity, polydispersity, particle concentration, particle structural properties and nucleic acid payload.
Collapse
|
14
|
Melting and Re-Freezing Leads to Irreversible Changes in the Morphology and Molecular-Level Dynamics of Pfizer-BioNTech COVID-19 Vaccine. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57121343. [PMID: 34946288 PMCID: PMC8706477 DOI: 10.3390/medicina57121343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/01/2022]
Abstract
Background and Objectives: As an mRNA-based vaccine, the Pfizer-BioNTech COVID-19 vaccine has stringent cold storage requirements to preserve functionality of the mRNA active ingredient. To this end, lipid components of the vaccine formulation play an important role in stabilizing and protecting the mRNA molecule for long-term storage. The purpose of the current study was to measure molecular-level dynamics as a function of temperature in the Pfizer-BioNTech COVID-19 vaccine to gain microscopic insight into its thermal stability. Materials and Methods: We used quasielastic and inelastic neutron scattering to probe (1) the vaccine extracted from the manufacturer-supplied vials and (2) unperturbed vaccine in the original manufacturer-supplied vials. The latter measurement was possible due to the high penetrative power of neutrons. Results: Upon warming from the low-temperature frozen state, the vaccine in its original form exhibits two-step melting, indicative of a two-phase morphology. Once the melting is completed (above 0 °C), vaccine re-freezing cannot restore its original two-phase state. This observation is corroborated by the changes in the molecular vibrational spectra. The molecular-level mobility measured in the resulting single-phase state of the re-frozen vaccine greatly exceeds the mobility measured in the original vaccine. Conclusions: Even a brief melting (above 0 °C) leads to an irreversible alteration of the two-phase morphology of the original vaccine formulation. Re-freezing of the vaccine results in a one-phase morphology with much increased molecular-level mobility compared to that in the original vaccine, suggesting irreversible deterioration of the vaccine’s in-storage stability. Neutron scattering can be used to distinguish between the vibrational spectra characteristic of the original and deteriorated vaccines contained in the unperturbed original manufacturer-supplied vials.
Collapse
|