Transfection Potency of Lipid Nanoparticles Containing mRNA Depends on Relative Loading LevelsClick to copy article linkArticle link copied!
- Suiyang LiaoSuiyang LiaoDepartment of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMichael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, CanadaSchool of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 2B9, CanadaMore by Suiyang Liao
- Shuangyu WangShuangyu WangDepartment of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMore by Shuangyu Wang
- Abishek WadhwaAbishek WadhwaDepartment of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaLife Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMore by Abishek Wadhwa
- Alexandra BirkenshawAlexandra BirkenshawFaculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMore by Alexandra Birkenshaw
- Kevin FoxKevin FoxDepartment of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaFaculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMore by Kevin Fox
- Miffy Hok Yan ChengMiffy Hok Yan ChengDepartment of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMore by Miffy Hok Yan Cheng
- Irafasha C. CasmilIrafasha C. CasmilMichael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, CanadaSchool of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 2B9, CanadaMore by Irafasha C. Casmil
- Armando Alcazar MaganaArmando Alcazar MaganaMichael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, CanadaLife Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMore by Armando Alcazar Magana
- Nuthan Vikas BathulaNuthan Vikas BathulaMichael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, CanadaSchool of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 2B9, CanadaMore by Nuthan Vikas Bathula
- Chia Hao HoChia Hao HoMichael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, CanadaMore by Chia Hao Ho
- Jin-Yu ChengJin-Yu ChengDepartment of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMore by Jin-Yu Cheng
- Leonard J. FosterLeonard J. FosterDepartment of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMichael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, CanadaLife Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMore by Leonard J. Foster
- Kenneth W. HarderKenneth W. HarderDepartment of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaLife Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMore by Kenneth W. Harder
- Colin J. D. RossColin J. D. RossFaculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMore by Colin J. D. Ross
- Pieter R. Cullis*Pieter R. Cullis*Email: [email protected]Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, CanadaMore by Pieter R. Cullis
- Anna K. Blakney*Anna K. Blakney*Email: [email protected]Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, CanadaSchool of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 2B9, CanadaMore by Anna K. Blakney
Abstract
When formulating mRNA into lipid nanoparticles (LNP), various copy numbers of mRNA are encapsulated, leading to a distribution of mRNA loading levels within the LNPs. It is unclear whether the mRNA loading level affects the functional delivery of the message. Here we show that depending on the mRNA loading level, LNPs exhibit distinct mass densities and can be fractionated via ultracentrifugation. Upon fractionation, we investigated if mRNA loading levels influence LNP sizing, lipid composition, and morphology. We further conducted in vitro and in vivo functional delivery of mRNA and found that the LNP fraction with the highest mRNA loading levels was the least transfection competent.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Results and Discussion
Centrifugal Fractionation of LNPs and the Integrity of LNP/mRNA
Figure 1
Figure 1. Centrifugal fractionation of LNPs based on their mRNA loading levels. (A) Schematic illustration of LNP fractionation via ultracentrifugation. Depending on the LNPs’ density contrast with the aqueous system, two fractions (LDLNP and HDLNP) are generated from the fractionation process. Together with the original batch, three samples are compared throughout this paper; (B) particle size distribution by intensity measured with DLS; payload mRNA integrity by Bioanalyzer shown as electropherograms in (C1) and bands in (C2).
Characterization of Lipid Composition and N/P Ratio
Figure 2
Figure 2. Validation of centrifugal fractionation, characterization of lipid composition, and calculation of N/P ratio. (A) Bar chart of RNA/total lipid ratio to represent the mRNA loading levels and validate the fractionation method; (B) pie charts showing lipid composition of OLNP, LDLNP, and HDLNP as characterized by ultrahigh-performance liquid chromatography–mass spectrometry (UHPLC-MS).
LNP Morphology Depends on mRNA Loading Level
Figure 3
Figure 3. mRNA loading levels can influence the distribution of LNP morphology. (A1–A3) Representative cryoEM images of OLNP, LDLNP, and HDLNP (scale bar: 100 nm); (B) pie charts showing the composition of LNPs with various number of blebs; (C) schematic illustration of measuring LNPs and blebs with FIJI; (D1–D3) scatter plots mapping the dimension of LNPs and blebs (purple dots: LNPs without blebs; green dots: LNPs with blebs) (E) raincloud plot showing the size distribution of blebs.
OLNP | LDLNP | HDLNP | ||
---|---|---|---|---|
composition (LNP with vs without blebs) | 50:50 | 73:27 | 49:51 | |
particles without blebs | size/nm | 52.1 | 55.1 | 49.1 |
isotropy | 0.98 | 1.00 | 1.02 | |
particles with blebs | size/nm | 69.6 | 68.7 | 74.6 |
isotropy | 1.19 | 1.08 | 1.23 | |
bleb size/nm | 37.1 | 33.8 | 42.0 | |
average number of blebs per particle (with blebs) | 1.23 | 1.11 | 1.48 | |
N/P ratio | 2.9 | 7.5 | 0.5 |
In Vitro mRNA Delivery with OLNP, LDLNP, and HDLNP
Figure 4
Figure 4. Cellular uptake and functional delivery of mRNA with OLNP, LDLNP, and HDLNP. (A) CLSM images of OLNP, LDLNP, and HDLNP ((A1), (A2), and (A3), respectively) delivering Cy5-labeled mEGFP into HEK293 cells (scale bar: 50 μm); (B1–B3) CLSM study on the colocalization between LNP colocalization and Rab7a, representing late endosomes (scale bar: 50 μm).
In Vivo mRNA Delivery with OLNP, LDLNP, and HDLNP
Figure 5
Figure 5. In vivo functional delivery of mFluc. (A) Schematic illustration of the animal study; (B) in vivo bioluminescence images of mice at 3 h after IV injection at mFluc dosage of 0.3 mg/kg (n = 4) and bar chart showing whole-body bioluminescence quantification from IVIS imaging. Data are presented as the mean ± standard deviation (SD) (n = 4). (C) Representative ex vivo bioluminescence images of extracted livers and spleens from mice dosed with OLNP/LDLNP/HDLNP (top to bottom) and bar chart showing bioluminescence of extracted livers and spleens. Data are presented as the mean ± SD (n = 4). (D) Scatter plot by projecting ex vivo liver radiance readout against spleen readout to indicate tissue tropism.
Conclusions
Methods
Materials
LNP Formulation and Characterization
Bioanalyzer
Cryo-TEM
Liquid Chromatography–Mass Spectrometry (LC-MS) Analysis
Cell Culture and Treatments
Animal Studies
Statistical Analysis
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsami.4c20077.
Mathematical deduction of eq 1; photos of DiD-labeled LNPs before and after ultracentrifugation (Figure S1); RiboGreen assay results show encapsulation efficiency (Figure S2); LNP size distribution by cryoEM (Figure S3); other representative cryoEM images (Figure S4); cellular uptake and functional delivery of mRNA in Huh7 cells (Figure S5); quantification of CLSM images (Figure S6); and colocalization analysis shown as Pearson’s coefficient (Figure S7) (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
References
This article references 29 other publications.
- 1Akinc, A.; Maier, M. A.; Manoharan, M.; Fitzgerald, K.; Jayaraman, M.; Barros, S.; Ansell, S.; Du, X.; Hope, M. J.; Madden, T. D.; Mui, B. L.; Semple, S. C.; Tam, Y. K.; Ciufolini, M.; Witzigmann, D.; Kulkarni, J. A.; van der Meel, R.; Cullis, P. R. The Onpattro Story and the Clinical Translation of Nanomedicines Containing Nucleic Acid-Based Drugs. Nat. Nanotechnol. 2019, 14 (12), 1084– 1087, DOI: 10.1038/s41565-019-0591-yGoogle Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlWit7vO&md5=3184ac1c3c74e6924fb1bb14b5229e95The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugsAkinc, Akin; Maier, Martin A.; Manoharan, Muthiah; Fitzgerald, Kevin; Jayaraman, Muthusamy; Barros, Scott; Ansell, Steven; Du, Xinyao; Hope, Michael J.; Madden, Thomas D.; Mui, Barbara L.; Semple, Sean C.; Tam, Ying K.; Ciufolini, Marco; Witzigmann, Dominik; Kulkarni, Jayesh A.; van der Meel, Roy; Cullis, Pieter R.Nature Nanotechnology (2019), 14 (12), 1084-1087CODEN: NNAABX; ISSN:1748-3387. (Nature Research)The regulatory approval of Onpattro, a lipid nanoparticle-based short interfering RNA drug for the treatment of polyneuropathies induced by hereditary transthyretin amyloidosis, paves the way for clin. development of many nucleic acid-based therapies enabled by nanoparticle delivery.
- 2Polack, F. P.; Thomas, S. J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J. L.; Pérez Marc, G.; Moreira, E. D.; Zerbini, C.; Bailey, R.; Swanson, K. A.; Roychoudhury, S.; Koury, K.; Li, P.; Kalina, W. V.; Cooper, D.; Frenck, R. W.; Hammitt, L. L.; Türeci, Ö.; Nell, H.; Schaefer, A.; Ünal, S.; Tresnan, D. B.; Mather, S.; Dormitzer, P. R.; Şahin, U.; Jansen, K. U.; Gruber, W. C. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N. Engl. J. Med. 2020, 383 (27), 2603– 2615, DOI: 10.1056/NEJMoa2034577Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXotFSjuw%253D%253D&md5=08cb7204334ed37c7949dfcdfe5af75cSafety and efficacy of the BNT162b2 mRNA Covid-19 vaccinePolack, Fernando P.; Thomas, Stephen J.; Kitchin, Nicholas; Absalon, Judith; Gurtman, Alejandra; Lockhart, Stephen; Perez, John L.; Marc, Gonzalo Perez; Moreira, Edson D.; Zerbini, Cristiano; Bailey, Ruth; Swanson, Kena A.; Roychoudhury, Satrajit; Koury, Kenneth; Li, Ping; Kalina, Warren V.; Cooper, David; Frenck, Robert W., Jr.; Hammitt, Laura L.; Tureci, Ozlem; Nell, Haylene; Schaefer, Axel; Unal, Serhat; Tresnan, Dina B.; Mather, Susan; Dormitzer, Philip R.; Sahin, Ugur; Jansen, Kathrin U.; Gruber, William C.New England Journal of Medicine (2020), 383 (27), 2603-2615CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)A review. Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting coronavirus disease 2019 (Covid-19) have afflicted tens of millions of people in a worldwide pandemic. Safe and effective vaccines are needed urgently. methods In an ongoing multinational, placebo-controlled, observer-blinded, pivotal efficacy trial, we randomly assigned persons 16 years of age or older in a 1:1 ratio to receive two doses, 21 days apart, of either placebo or the BNT162b2 vaccine candidate (30μg per dose). BNT162b2 is a lipid nanoparticle-formulated, nucleoside-modified RNA vaccine that encodes a prefusion stabilized, membrane-anchored SARS-CoV-2 full-length spike protein. The primary end points were efficacy of the vaccine against lab.-confirmed Covid-19 and safety. results A total of 43,548 participants underwent randomization, of whom 43,448 received injections: 21,720 with BNT162b2 and 21,728 with placebo. There were 8 cases of Covid-19 with onset at least 7 days after the second dose among participants assigned to receive BNT162b2 and 162 cases among those assigned to placebo; BNT162b2 was 95% effective in preventing Covid-19 (95% credible interval, 90.3 to 97.6). Similar vaccine efficacy (generally 90 to 100%) was obsd. across subgroups defined by age, sex, race, ethnicity, baseline body-mass index, and the presence of coexisting conditions. Among 10 cases of severe Covid-19 with onset after the first dose, 9 occurred in placebo recipients and 1 in a BNT162b2 recipient. The safety profile of BNT162b2 was characterized by short-term, mild-to-moderate pain at the injection site, fatigue, and headache. The incidence of serious adverse events was low and was similar in the vaccine and placebo groups. conclusions A two-dose regimen of BNT162b2 conferred 95% protection against Covid-19 in persons 16 years of age or older. Safety over a median of 2 mo was similar to that of other viral vaccines.
- 3Baden, L. R.; El Sahly, H. M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S. A.; Rouphael, N.; Creech, C. B.; McGettigan, J.; Khetan, S.; Segall, N.; Solis, J.; Brosz, A.; Fierro, C.; Schwartz, H.; Neuzil, K.; Corey, L.; Gilbert, P.; Janes, H.; Follmann, D.; Marovich, M.; Mascola, J.; Polakowski, L.; Ledgerwood, J.; Graham, B. S.; Bennett, H.; Pajon, R.; Knightly, C.; Leav, B.; Deng, W.; Zhou, H.; Han, S.; Ivarsson, M.; Miller, J.; Zaks, T. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N. Engl. J. Med. 2021, 384 (5), 403– 416, DOI: 10.1056/NEJMoa2035389Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjvFSmsrc%253D&md5=9b0c79f3e3fd2ea452a3fbf57281720fEfficacy and safety of the mRNA-1273 SARS-CoV-2 vaccineBaden, L. R.; El Sahly, H. M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S. A.; Rouphael, N.; Creech, C. B.; McGettigan, J.; Khetan, S.; Segall, N.; Solis, J.; Brosz, A.; Fierro, C.; Schwartz, H.; Neuzil, K.; Corey, L.; Gilbert, P.; Janes, H.; Follmann, D.; Marovich, M.; Mascola, J.; Polakowski, L.; Ledgerwood, J.; Graham, B. S.; Bennett, H.; Pajon, R.; Knightly, C.; Leav, B.; Deng, W.; Zhou, H.; Han, S.; Ivarsson, M.; Miller, J.; Zaks, T.New England Journal of Medicine (2021), 384 (5), 403-416CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)Vaccines are needed to prevent coronavirus disease 2019 (Covid-19) and to protect persons who are at high risk for complications. The mRNA-1273 vaccine is a lipid nanoparticle-encapsulated mRNA-based vaccine that encodes the prefusion stabilized full-length spike protein of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes Covid-19. This phase 3 randomized, observer-blinded, placebo-controlled trial was conducted at 99 centers across the United States. Persons at high risk for SARS-CoV-2 infection or its complications were randomly assigned in a 1:1 ratio to receive two i.m. injections of mRNA-1273 (100μg) or placebo 28 days apart. The primary end point was prevention of Covid-19 illness with onset at least 14 days after the second injection in participants who had not previously been infected with SARS-CoV-2. The trial enrolled 30,420 volunteers who were randomly assigned in a 1:1 ratio to receive either vaccine or placebo (15,210 participants in each group). More than 96% of participants received both injections, and 2.2% had evidence (serol., virol., or both) of SARS-CoV-2 infection at baseline. Symptomatic Covid-19 illness was confirmed in 185 participants in the placebo group (56.5 per 1000 person-years; 95% confidence interval [CI], 48.7 to 65.3) and in 11 participants in the mRNA-1273 group (3.3 per 1000 person-years; 95% CI, 1.7 to 6.0); vaccine efficacy was 94.1% (95% CI, 89.3 to 96.8%; P<0.001). Efficacy was similar across key secondary analyses, including assessment 14 days after the first dose, analyses that included participants who had evidence of SARS-CoV-2 infection at baseline, and analyses in participants 65 years of age or older. Severe Covid-19 occurred in 30 participants, with one fatality; all 30 were in the placebo group. Moderate, transient reactogenicity after vaccination occurred more frequently in the mRNA-1273 group. Serious adverse events were rare, and the incidence was similar in the two groups. The mRNA-1273 vaccine showed 94.1% efficacy at preventing Covid-19 illness, including severe disease. Aside from transient local and systemic reactions, no safety concerns were identified.
- 4Wilson, E.; Goswami, J.; Baqui, A. H.; Doreski, P. A.; Perez-Marc, G.; Zaman, K.; Monroy, J.; Duncan, C. J. A.; Ujiie, M.; Rämet, M.; Pérez-Breva, L.; Falsey, A. R.; Walsh, E. E.; Dhar, R.; Wilson, L.; Du, J.; Ghaswalla, P.; Kapoor, A.; Lan, L.; Mehta, S.; Mithani, R.; Panozzo, C. A.; Simorellis, A. K.; Kuter, B. J.; Schödel, F.; Huang, W.; Reuter, C.; Slobod, K.; Stoszek, S. K.; Shaw, C. A.; Miller, J. M.; Das, R.; Chen, G. L. Efficacy and Safety of an mRNA-Based RSV PreF Vaccine in Older Adults. N. Engl. J. Med. 2023, 389 (24), 2233– 2244, DOI: 10.1056/NEJMoa2307079Google ScholarThere is no corresponding record for this reference.
- 5Kulkarni, J. A.; Witzigmann, D.; Leung, J.; van der Meel, R.; Zaifman, J.; Darjuan, M. M.; Grisch-Chan, H. M.; Thöny, B.; Tam, Y. Y. C.; Cullis, P. R. Fusion-Dependent Formation of Lipid Nanoparticles Containing Macromolecular Payloads. Nanoscale 2019, 11 (18), 9023– 9031, DOI: 10.1039/C9NR02004GGoogle Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXnvFymtLk%253D&md5=f588b6991dd4e67e61e1f286fa3c2b4fFusion-dependent formation of lipid nanoparticles containing macromolecular payloadsKulkarni, Jayesh A.; Witzigmann, Dominik; Leung, Jerry; van der Meel, Roy; Zaifman, Josh; Darjuan, Maria M.; Grisch-Chan, Hiu Man; Thony, Beat; Tam, Yuen Yi C.; Cullis, Pieter R.Nanoscale (2019), 11 (18), 9023-9031CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)The success of Onpattro (patisiran) clearly demonstrates the utility of lipid nanoparticle (LNP) systems for enabling gene therapies. These systems are composed of ionizable cationic lipids, phospholipid, cholesterol, and polyethylene glycol (PEG)-lipids, and are produced through rapid-mixing of an ethanolic-lipid soln. with an acidic aq. soln. followed by dialysis into neutralizing buffer. A detailed understanding of the mechanism of LNP formation is crucial to improving LNP design. Here we use cryogenic transmission electron microscopy and fluorescence techniques to further demonstrate that LNP are formed through the fusion of precursor, pH-sensitive liposomes into large electron-dense core structures as the pH is neutralized. Next, we show that the fusion process is limited by the accumulation of PEG-lipid on the emerging particle. Finally, we show that the fusion-dependent mechanism of formation also applies to LNP contg. macromol. payloads including mRNA, DNA vectors, and gold nanoparticles.
- 6Li, S.; Hu, Y.; Lin, J.; Schneiderman, Z.; Shao, F.; Wei, L.; Li, A.; Hsieh, K.; Kokkoli, E.; Curk, T.; Mao, H.-Q.; Wang, T.-H. Single-Particle Spectroscopic Chromatography Reveals Heterogeneous RNA Loading and Size Correlations in Lipid Nanoparticles. ACS Nano 2024, 18, 15729, DOI: 10.1021/acsnano.4c02341Google ScholarThere is no corresponding record for this reference.
- 7Li, S.; Hu, Y.; Li, A.; Lin, J.; Hsieh, K.; Schneiderman, Z.; Zhang, P.; Zhu, Y.; Qiu, C.; Kokkoli, E.; Wang, T.-H.; Mao, H.-Q. Payload Distribution and Capacity of mRNA Lipid Nanoparticles. Nat. Commun. 2022, 13 (1), 5561 DOI: 10.1038/s41467-022-33157-4Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XisVyrsL7J&md5=df0c413e8724a661b328523b99a7d4b1Payload distribution and capacity of mRNA lipid nanoparticlesLi, Sixuan; Hu, Yizong; Li, Andrew; Lin, Jinghan; Hsieh, Kuangwen; Schneiderman, Zachary; Zhang, Pengfei; Zhu, Yining; Qiu, Chenhu; Kokkoli, Efrosini; Wang, Tza-Huei; Mao, Hai-QuanNature Communications (2022), 13 (1), 5561CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)Lipid nanoparticles (LNPs) are effective vehicles to deliver mRNA vaccines and therapeutics. It has been challenging to assess mRNA packaging characteristics in LNPs, including payload distribution and capacity, which are crit. to understanding structure-property-function relationships for further carrier development. Here, we report a method based on the multi-laser cylindrical illumination confocal spectroscopy (CICS) technique to examine mRNA and lipid contents in LNP formulations at the single-nanoparticle level. By differentiating unencapsulated mRNAs, empty LNPs and mRNA-loaded LNPs via coincidence anal. of fluorescent tags on different LNP components, and quant. resolving single-mRNA fluorescence, we reveal that a commonly referenced benchmark formulation using DLin-MC3 as the ionizable lipid contains mostly 2 mRNAs per loaded LNP with a presence of 40%-80% empty LNPs depending on the assembly conditions. Systematic anal. of different formulations with control variables reveals a kinetically controlled assembly mechanism that governs the payload distribution and capacity in LNPs. These results form the foundation for a holistic understanding of the mol. assembly of mRNA LNPs.
- 8De Kloet, S. R.; Andrean, B. A. G. Buoyant Density Gradient Centrifugation of RNA and DNA in Alkali Iodide Solutions. Biochim. Biophys. Acta, Nucleic Acids Protein Synth. 1971, 247 (4), 519– 527, DOI: 10.1016/0005-2787(71)90688-5Google ScholarThere is no corresponding record for this reference.
- 9Guerrini, G.; Mehn, D.; Scaccabarozzi, D.; Gioria, S.; Calzolai, L. Analytical Ultracentrifugation to Assess the Quality of LNP-mRNA Therapeutics. Int. J. Mol. Sci. 2024, 25 (11), 5718 DOI: 10.3390/ijms25115718Google ScholarThere is no corresponding record for this reference.
- 10Schiel, J. E.; Hage, D. S. Density Measurements of Potassium Phosphate Buffer from 4 to 45 °C. Talanta 2005, 65 (2), 495– 500, DOI: 10.1016/j.talanta.2004.06.029Google ScholarThere is no corresponding record for this reference.
- 11Henrickson, A.; Kulkarni, J. A.; Zaifman, J.; Gorbet, G. E.; Cullis, P. R.; Demeler, B. Density Matching Multi-Wavelength Analytical Ultracentrifugation to Measure Drug Loading of Lipid Nanoparticle Formulations. ACS Nano 2021, 15, 5068, DOI: 10.1021/acsnano.0c10069Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXks1yjtLg%253D&md5=2b8a668202896863661f0971b05d51e3Density Matching Multi-wavelength Analytical Ultracentrifugation to Measure Drug Loading of Lipid Nanoparticle FormulationsHenrickson, Amy; Kulkarni, Jayesh A.; Zaifman, Josh; Gorbet, Gary E.; Cullis, Pieter R.; Demeler, BorriesACS Nano (2021), 15 (3), 5068-5076CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Previous work suggested that lipid nanoparticle (LNP) formulations, encapsulating nucleic acids, display electron-dense morphol. when examd. by cryogenic-transmission electron microscopy (cryo-TEM). Critically, the employed cryo-TEM method cannot differentiate between loaded and empty LNP formulations. Clin. relevant formulations contain high lipid-to-nucleic acid ratios (10-25 (wt./wt.)), and for systems that contain mRNA or DNA, it is anticipated that a substantial fraction of the LNP population does not contain a payload. Here, we present a method based on the global anal. of multi-wavelength sedimentation velocity anal. ultracentrifugation, using d. matching with heavy water, that not only measures the std. sedimentation and diffusion coeff. distributions of LNP mixts., but also reports the corresponding partial sp. vol. distributions and optically separates signal contributions from nucleic acid cargo and lipid shell. This makes it possible to reliably predict molar mass and anisotropy distributions, in particular, for systems that are heterogeneous in partial sp. vol. and have low to intermediate densities. Our method makes it possible to unambiguously measure the d. of nanoparticles and is motivated by the need to characterize the extent to which lipid nanoparticles are loaded with nucleic acid cargoes. Since the densities of nucleic acids and lipids substantially differ, the measured d. is directly proportional to the loading of nanoparticles. Hence, different loading levels will produce particles with variable d. and partial sp. vol. An UltraScan software module was developed to implement this approach for routine anal.
- 12Bepperling, A.; Richter, G. Determination of mRNA Copy Number in Degradable Lipid Nanoparticles via Density Contrast Analytical Ultracentrifugation. Eur. Biophys J. 2023, 52, 393, DOI: 10.1007/s00249-023-01663-yGoogle Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXhtF2iurnO&md5=ae9b4805103592dc6f27bfd07cef7c5bDetermination of mRNA copy number in degradable lipid nanoparticles via density contrast analytical ultracentrifugationBepperling, Alexander; Richter, GesaEuropean Biophysics Journal (2023), 52 (4-5), 393-400CODEN: EBJOE8; ISSN:0175-7571. (Springer International Publishing AG)Lipid nanoparticles as delivery system for mRNA have recently attracted attention to a broader audience as COVID-19 mRNA vaccines. Their low immunogenicity and capability to deliver a variety of nucleic acids renders them an interesting and complementary alternative to gene therapy vectors like AAVs. An important quality attribute of LNPs is the copy no. of the encapsulated cargo mol. This work describes how d. and mol. wt. distributions obtained by d. contrast sedimentation velocity can be used to calc. the mRNA copy no. of a degradable lipid nanoparticle formulation. The detd. av. copy no. of 5 mRNA mols. per LNP is consistent with the previous studies using other biophys. techniques, such as single particle imaging microscopy and multi-laser cylindrical illumination confocal spectroscopy (CICS).
- 13Leung, A. K. K.; Hafez, I. M.; Baoukina, S.; Belliveau, N. M.; Zhigaltsev, I. V.; Afshinmanesh, E.; Tieleman, D. P.; Hansen, C. L.; Hope, M. J.; Cullis, P. R. Lipid Nanoparticles Containing siRNA Synthesized by Microfluidic Mixing Exhibit an Electron-Dense Nanostructured Core. J. Phys. Chem. C 2012, 116 (34), 18440– 18450, DOI: 10.1021/jp303267yGoogle Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVOjs7zL&md5=0f25938ac9ee92e7c8dc34ccc0d0f27aLipid Nanoparticles Containing siRNA Synthesized by Microfluidic Mixing Exhibit an Electron-Dense Nanostructured CoreLeung, Alex K. K.; Hafez, Ismail M.; Baoukina, Svetlana; Belliveau, Nathan M.; Zhigaltsev, Igor V.; Afshinmanesh, Elham; Tieleman, D. Peter; Hansen, Carl L.; Hope, Michael J.; Cullis, Pieter R.Journal of Physical Chemistry C (2012), 116 (34), 18440-18450CODEN: JPCCCK; ISSN:1932-7447. (American Chemical Society)Lipid nanoparticles (LNP) contg. ionizable cationic lipids are the leading systems for enabling therapeutic applications of siRNA; however, the structure of these systems has not been defined. Here we examine the structure of LNP siRNA systems contg. DLinKC2-DMA(an ionizable cationic lipid), phospholipid, cholesterol and a polyethylene glycol (PEG) lipid formed using a rapid microfluidic mixing process. Techniques employed include cryo-transmission electron microscopy, 31P NMR, membrane fusion assays, d. measurements, and mol. modeling. The exptl. results indicate that these LNP siRNA systems have an interior lipid core contg. siRNA duplexes complexed to cationic lipid and that the interior core also contains phospholipid and cholesterol. Consistent with exptl. observations, mol. modeling calcns. indicate that the interior of LNP siRNA systems exhibits a periodic structure of aq. compartments, where some compartments contain siRNA. It is concluded that LNP siRNA systems formulated by rapid mixing of an ethanol soln. of lipid with an aq. medium contg. siRNA exhibit a nanostructured core. The results give insight into the mechanism whereby LNP siRNA systems are formed, providing an understanding of the high encapsulation efficiencies that can be achieved and information on methods of constructing more sophisticated LNP systems.
- 14Vaidya, A.; Parande, D.; Khadse, N.; Vargas-Montoya, N.; Agarwal, V.; Ortiz, C.; Ellis, G.; Kaushal, N.; Sarode, A.; Karve, S.; DeRosa, F. Analytical Characterization of Heterogeneities in mRNA-Lipid Nanoparticles Using Sucrose Density Gradient Ultracentrifugation. Anal. Chem. 2024, 96, 5570, DOI: 10.1021/acs.analchem.4c00031Google ScholarThere is no corresponding record for this reference.
- 15Zhao, H.; Sousa, A. A.; Schuck, P. Flotation Coefficient Distributions of Lipid Nanoparticles by Sedimentation Velocity Analytical Ultracentrifugation. ACS Nano 2024, 18, 18663, DOI: 10.1021/acsnano.4c05322Google ScholarThere is no corresponding record for this reference.
- 16Semple, S. C.; Akinc, A.; Chen, J.; Sandhu, A. P.; Mui, B. L.; Cho, C. K.; Sah, D. W. Y.; Stebbing, D.; Crosley, E. J.; Yaworski, E.; Hafez, I. M.; Dorkin, J. R.; Qin, J.; Lam, K.; Rajeev, K. G.; Wong, K. F.; Jeffs, L. B.; Nechev, L.; Eisenhardt, M. L.; Jayaraman, M.; Kazem, M.; Maier, M. A.; Srinivasulu, M.; Weinstein, M. J.; Chen, Q.; Alvarez, R.; Barros, S. A.; De, S.; Klimuk, S. K.; Borland, T.; Kosovrasti, V.; Cantley, W. L.; Tam, Y. K.; Manoharan, M.; Ciufolini, M. A.; Tracy, M. A.; de Fougerolles, A.; MacLachlan, I.; Cullis, P. R.; Madden, T. D.; Hope, M. J. Rational Design of Cationic Lipids for siRNA Delivery. Nat. Biotechnol. 2010, 28 (2), 172– 176, DOI: 10.1038/nbt.1602Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXntVCqtg%253D%253D&md5=8ed6986f3ed898efe69f4576f762b3adRational design of cationic lipids for siRNA deliverySemple, Sean C.; Akinc, Akin; Chen, Jianxin; Sandhu, Ammen P.; Mui, Barbara L.; Cho, Connie K.; Sah, Dinah W. Y.; Stebbing, Derrick; Crosley, Erin J.; Yaworski, Ed; Hafez, Ismail M.; Dorkin, J. Robert; Qin, June; Lam, Kieu; Rajeev, Kallanthottathil G.; Wong, Kim F.; Jeffs, Lloyd B.; Nechev, Lubomir; Eisenhardt, Merete L.; Jayaraman, Muthusamy; Kazem, Mikameh; Maier, Martin A.; Srinivasulu, Masuna; Weinstein, Michael J.; Chen, Qingmin; Alvarez, Rene; Barros, Scott A.; De, Soma; Klimuk, Sandra K.; Borland, Todd; Kosovrasti, Verbena; Cantley, William L.; Tam, Ying K.; Manoharan, Muthiah; Ciufolini, Marco A.; Tracy, Mark A.; de Fougerolles, Antonin; MacLachlan, Ian; Cullis, Pieter R.; Madden, Thomas D.; Hope, Michael J.Nature Biotechnology (2010), 28 (2), 172-176CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)We adopted a rational approach to design cationic lipids for use in formulations to deliver small interfering RNA (siRNA). Starting with the ionizable cationic lipid 1,2-dilinoleyloxy-3-dimethylaminopropane (DLinDMA), a key lipid component of stable nucleic acid lipid particles (SNALP) as a benchmark, we used the proposed in vivo mechanism of action of ionizable cationic lipids to guide the design of DLinDMA-based lipids with superior delivery capacity. The best-performing lipid recovered after screening (DLin-KC2-DMA) was formulated and characterized in SNALP and demonstrated to have in vivo activity at siRNA doses as low as 0.01 mg/kg in rodents and 0.1 mg/kg in nonhuman primates. To our knowledge, this represents a substantial improvement over previous reports of in vivo endogenous hepatic gene silencing.
- 17Svedberg, T.; Rinde, H. The Ultra-centrifuge, a New Instrument for the Determination of Size and Distribution of Size of Particle In Amicroscopic Colloids. J. Am. Chem. Soc. 1924, 46 (12), 2677– 2693, DOI: 10.1021/ja01677a011Google ScholarThere is no corresponding record for this reference.
- 18Kim, J.; Eygeris, Y.; Gupta, M.; Sahay, G. Self-Assembled mRNA Vaccines. Adv. Drug Delivery Rev. 2021, 170, 83, DOI: 10.1016/j.addr.2020.12.014Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslWgtr0%253D&md5=d213939af17262d61f009547d94627feSelf-assembled mRNA vaccinesKim, Jeonghwan; Eygeris, Yulia; Gupta, Mohit; Sahay, GauravAdvanced Drug Delivery Reviews (2021), 170 (), 83-112CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. MRNA vaccines have evolved from being a mere curiosity to emerging as COVID-19 vaccine front-runners. Recent advancements in the field of RNA technol., vaccinol., and nanotechnol. have generated interest in delivering safe and effective mRNA therapeutics. In this review, we discuss design and self-assembly of mRNA vaccines. Self-assembly, a spontaneous organization of individual mols., allows for design of nanoparticles with customizable properties. We highlight the materials commonly utilized to deliver mRNA, their physicochem. characteristics, and other relevant considerations, such as mRNA optimization, routes of administration, cellular fate, and immune activation, that are important for successful mRNA vaccination. We also examine the COVID-19 mRNA vaccines currently in clin. trials. mRNA vaccines are ready for the clinic, showing tremendous promise in the COVID-19 vaccine race, and have pushed the boundaries of gene therapy.
- 19Gilbert, J.; Sebastiani, F.; Arteta, M. Y.; Terry, A.; Fornell, A.; Russell, R.; Mahmoudi, N.; Nylander, T. Evolution of the Structure of Lipid Nanoparticles for Nucleic Acid Delivery: From in Situ Studies of Formulation to Colloidal Stability. J. Colloid Interface Sci. 2024, 660, 66– 76, DOI: 10.1016/j.jcis.2023.12.165Google ScholarThere is no corresponding record for this reference.
- 20Szebeni, J.; Kiss, B.; Bozó, T.; Turjeman, K.; Levi-Kalisman, Y.; Barenholz, Y.; Kellermayer, M. Insights into the Structure of Comirnaty Covid-19 Vaccine: A Theory on Soft, Partially Bilayer-Covered Nanoparticles with Hydrogen Bond-Stabilized mRNA–Lipid Complexes. ACS Nano 2023, 17 (14), 13147– 13157, DOI: 10.1021/acsnano.2c11904Google ScholarThere is no corresponding record for this reference.
- 21Forchette, L.; Sebastian, W.; Liu, T. A Comprehensive Review of COVID-19 Virology, Vaccines, Variants, and Therapeutics. Curr. Med. Sci. 2021, 41 (6), 1037– 1051, DOI: 10.1007/s11596-021-2395-1Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsFGhurbE&md5=d90b78150fd6761fc4a02e09ff4d1254A Comprehensive Review of COVID-19 Virology, Vaccines, Variants, and TherapeuticsForchette, Lauren; Sebastian, William; Liu, TuoenCurrent Medical Science (2021), 41 (6), 1037-1051CODEN: CMSUCM; ISSN:2523-899X. (Huazhong University of Science and Technology)A review. Summary: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative pathogen of the coronavirus disease 2019 (COVID-19), has caused more than 179 million infections and 3.8 million deaths worldwide. Throughout the past year, multiple vaccines have already been developed and used, while some others are in the process of being developed. However, the emergence of new mutant strains of SARS-CoV-2 that have demonstrated immune-evading characteristics and an increase in infective capabilities leads to potential ineffectiveness of the vaccines against these variants. The purpose of this review article is to highlight the current understanding of the immunol. mechanisms of the virus and vaccines, as well as to investigate some key variants and mutations of the virus driving the current pandemic and their impacts on current management guidelines. We also discussed new technologies being developed for the prevention, treatment, and detection of SARS-CoV-2. In this paper, we thoroughly reviewed and provided crucial information on SARS-CoV-2 virol., vaccines and drugs being used and developed for its prevention and treatment, as well as important variant strains. Our review paper will be beneficial to health care professionals and researchers so they can have a better understanding of the basic sciences, prevention, and clin. treatment of COVID-19 during the pandemic. This paper consists of the most updated information that has been available as of June 21, 2021.
- 22Heinz, F. X.; Stiasny, K. Distinguishing Features of Current COVID-19 Vaccines: Knowns and Unknowns of Antigen Presentation and Modes of Action. npj Vaccines 2021, 6 (1), 104 DOI: 10.1038/s41541-021-00369-6Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVemurzL&md5=818b1fef015ae9bbffda5db8b50a7f09Distinguishing features of current COVID-19 vaccines: knowns and unknowns of antigen presentation and modes of actionHeinz, Franz X.; Stiasny, Karinnpj Vaccines (2021), 6 (1), 104CODEN: VACCBC; ISSN:2059-0105. (Nature Research)A review. COVID-19 vaccines were developed with an unprecedented pace since the beginning of the pandemic. Several of them have reached market authorization and mass prodn., leading to their global application on a large scale. This enormous progress was achieved with fundamentally different vaccine technologies used in parallel. mRNA, adenoviral vector as well as inactivated whole-virus vaccines are now in widespread use, and a subunit vaccine is in a final stage of authorization. They all rely on the native viral spike protein (S) of SARS-CoV-2 for inducing potently neutralizing antibodies, but the presentation of this key antigen to the immune system differs substantially between the different categories of vaccines. In this article, we review the relevance of structural modifications of S in different vaccines and the different modes of antigen expression after vaccination with genetic adenovirus-vector and mRNA vaccines. Distinguishing characteristics and unknown features are highlighted in the context of protective antibody responses and reactogenicity of vaccines.
- 23Buschmann, M. D.; Carrasco, M. J.; Alishetty, S.; Paige, M.; Alameh, M. G.; Weissman, D. Nanomaterial Delivery Systems for mRNA Vaccines. Vaccines 2021, 9 (1), 65 DOI: 10.3390/vaccines9010065Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhs1GgsrjP&md5=e09076471c269d29078c6b2445d542d8Nanomaterial delivery systems for mRNA vaccinesBuschmann, Michael D.; Carrasco, Manuel J.; Alishetty, Suman; Paige, Mikell; Alameh, Mohamad Gabriel; Weissman, DrewVaccines (Basel, Switzerland) (2021), 9 (1), 65CODEN: VBSABP; ISSN:2076-393X. (MDPI AG)The recent success of mRNA vaccines in SARS-CoV-2 clin. trials is in part due to the development of lipid nanoparticle delivery systems that not only efficiently express the mRNA-encoded immunogen after i.m. injection, but also play roles as adjuvants and in vaccine reactogenicity. We present an overview of mRNA delivery systems and then focus on the lipid nanoparticles used in the current SARS-CoV-2 vaccine clin. trials. The review concludes with an anal. of the determinants of the performance of lipid nanoparticles in mRNA vaccines.
- 24Tenchov, R.; Bird, R.; Curtze, A. E.; Zhou, Q. Lipid Nanoparticles─From Liposomes to mRNA Vaccine Delivery, a Landscape of Research Diversity and Advancement. ACS Nano 2021, 15 (11), 16982– 17015, DOI: 10.1021/acsnano.1c04996Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsVSqsr%252FM&md5=c824f1ae324b984f94d2e33741ceef8bLipid Nanoparticles-From Liposomes to mRNA Vaccine Delivery, a Landscape of Research Diversity and AdvancementTenchov, Rumiana; Bird, Robert; Curtze, Allison E.; Zhou, QiongqiongACS Nano (2021), 15 (11), 16982-17015CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)A review. Lipid nanoparticles (LNPs) have emerged across the pharmaceutical industry as promising vehicles to deliver a variety of therapeutics. Currently in the spotlight as vital components of the COVID-19 mRNA vaccines, LNPs play a key role in effectively protecting and transporting mRNA to cells. Liposomes, an early version of LNPs, are a versatile nanomedicine delivery platform. A no. of liposomal drugs have been approved and applied to medical practice. Subsequent generations of lipid nanocarriers, such as solid lipid nanoparticles, nanostructured lipid carriers, and cationic lipid-nucleic acid complexes, exhibit more complex architectures and enhanced phys. stabilities. With their ability to encapsulate and deliver therapeutics to specific locations within the body and to release their contents at a desired time, LNPs provide a valuable platform for treatment of a variety of diseases. We present a landscape of LNP-related scientific publications, including patents and journal articles, based on anal. of the CAS Content Collection, the largest human-curated collection of published scientific knowledge. Rising trends are identified, such as nanostructured lipid carriers and solid lipid nanoparticles becoming the preferred platforms for numerous formulations. Recent advancements in LNP formulations as drug delivery platforms, such as antitumor and nucleic acid therapeutics and vaccine delivery systems, are discussed. Challenges and growth opportunities are also evaluated in other areas, such as medical imaging, cosmetics, nutrition, and agrochems. This report is intended to serve as a useful resource for those interested in LNP nanotechnologies, their applications, and the global research effort for their development.
- 25Kloczewiak, M.; Banks, J. M.; Jin, L.; Brader, M. L. A Biopharmaceutical Perspective on Higher-Order Structure and Thermal Stability of mRNA Vaccines. Mol. Pharmaceutics 2022, 19 (7), 2022– 2031, DOI: 10.1021/acs.molpharmaceut.2c00092Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsFejt7vM&md5=ce77c9d38757cb5a47a66a583008e2d9A Biopharmaceutical Perspective on Higher-Order Structure and Thermal Stability of mRNA VaccinesKloczewiak, Marek; Banks, Jessica M.; Jin, Lin; Brader, Mark L.Molecular Pharmaceutics (2022), 19 (7), 2022-2031CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)A review. Preservation of the integrity of macromol. higher-order structure is a tenet central to achieving biol. drug and vaccine product stability toward manufg., 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 biol. drugs, at least at 1st 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 biol. 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 mols. We extend those findings by considering the effect of encapsulation on mRNA thermal unfolding with the observation that encapsulation in LNPs increases mRNA unfolding temps.
- 26Brader, M. L.; Williams, S. J.; Banks, J. M.; Hui, W. H.; Zhou, Z. H.; Jin, L. Encapsulation State of Messenger RNA inside Lipid Nanoparticles. Biophys. J. 2021, 120 (14), 2766– 2770, DOI: 10.1016/j.bpj.2021.03.012Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXns1Cjsbc%253D&md5=dca599c7e6b5129440e6ce1b9ff78b44Encapsulation state of messenger RNA inside lipid nanoparticlesBrader, Mark L.; Williams, Sean J.; Banks, Jessica M.; Hui, Wong H.; Zhou, Z. Hong; Jin, LinBiophysical Journal (2021), 120 (14), 2766-2770CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)Understanding the structure of mRNA (mRNA) lipid nanoparticles, and specifically the microenvironment of the mRNA mols. within these entities, is fundamental to advancing their biomedical potential. Here, we show that a permeating cationic dye, thionine, can serve as a cryogenic electron microscopy contrasting agent by binding selectively to encapsulated mRNA without disturbing lipid nanoparticle morphol. Cryo-electron microscopy images identify the mRNA location, revealing that mRNA may exist within solvent-filled cavities or may be substantially lipid assocd.
- 27Cheng, M. H. Y.; Leung, J.; Zhang, Y.; Strong, C.; Basha, G.; Momeni, A.; Chen, Y.; Jan, E.; Abdolahzadeh, A.; Wang, X.; Kulkarni, J. A.; Witzigmann, D.; Cullis, P. R. Induction of Bleb Structures in Lipid Nanoparticle Formulations of mRNA Leads to Improved Transfection Potency. Adv. Mater. 2023, 35 (31), 2303370 DOI: 10.1002/adma.202303370Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXhtlSqtrnE&md5=6fadec23ad601f3cc8f817c2c8709265Induction of Bleb Structures in Lipid Nanoparticle Formulations of mRNA Leads to Improved Transfection PotencyCheng, Miffy Hok Yan; Leung, Jerry; Zhang, Yao; Strong, Colton; Basha, Genc; Momeni, Arash; Chen, Yihang; Jan, Eric; Abdolahzadeh, Amir; Wang, Xinying; Kulkarni, Jayesh A.; Witzigmann, Dominik; Cullis, Pieter R.Advanced Materials (Weinheim, Germany) (2023), 35 (31), 2303370CODEN: ADVMEW; ISSN:0935-9648. (Wiley-VCH Verlag GmbH & Co. KGaA)The transfection potency of lipid nanoparticle (LNP) mRNA systems is critically dependent on the ionizable cationic lipid component. LNP mRNA systems composed of optimized ionizable lipids often display distinctive mRNA-rich "bleb" structures. Here, it is shown that such structures can also be induced for LNPs contg. nominally less active ionizable lipids by formulating them in the presence of high concns. of pH 4 buffers such as sodium citrate, leading to improved transfection potencies both in vitro and in vivo. Induction of bleb structure and improved potency is dependent on the type of pH 4 buffer employed, with LNP mRNA systems prepd. using 300 mM sodium citrate buffer displaying max. transfection. The improved transfection potencies of LNP mRNA systems displaying bleb structure can be attributed, at least in part, to enhanced integrity of the encapsulated mRNA. It is concluded that enhanced transfection can be achieved by optimizing formulation parameters to improve mRNA stability and that optimization of ionizable lipids to achieve enhanced potency may well lead to improvements in mRNA integrity through formation of the bleb structure rather than enhanced intracellular delivery.
- 28Whitehead, K. A.; Matthews, J.; Chang, P. H.; Niroui, F.; Dorkin, J. R.; Severgnini, M.; Anderson, D. G. In Vitro–In Vivo Translation of Lipid Nanoparticles for Hepatocellular siRNA Delivery. ACS Nano 2012, 6 (8), 6922– 6929, DOI: 10.1021/nn301922xGoogle Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XpvFegtL8%253D&md5=011ddae7baf862301e24ac829334f599In Vitro-In Vivo Translation of Lipid Nanoparticles for Hepatocellular siRNA DeliveryWhitehead, Kathryn A.; Matthews, Jonathan; Chang, Philip H.; Niroui, Farnaz; Dorkin, J. Robert; Severgnini, Mariano; Anderson, Daniel G.ACS Nano (2012), 6 (8), 6922-6929CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)A significant challenge in the development of clin. viable siRNA delivery systems is a lack of in vitro-in vivo translatability: many delivery vehicles that are initially promising in cell culture do not retain efficacy in animals. Despite its importance, little information exists on the predictive nature of in vitro methodologies, most likely due to the cost and time assocd. with generating in vitro-in vivo data sets. Recently, high-throughput techniques have been developed that have allowed the examn. of hundreds of lipid nanoparticle formulations for transfection efficiency in multiple exptl. systems. The large resulting data set has allowed the development of correlations between in vitro and characterization data and in vivo efficacy for hepatocellular delivery vehicles. Consistency of formulation technique and the type of cell used for in vitro expts. was found to significantly affect correlations, with primary hepatocytes and HeLa cells yielding the most predictive data. Interestingly, in vitro data acquired using HeLa cells were more predictive of in vivo performance than mouse hepatoma Hepa1-6 cells. Of the characterization parameters, only siRNA entrapment efficiency was partially predictive of in vivo silencing potential, while zeta-potential and, surprisingly, nanoparticle size (when <300 nm) as measured by dynamic light scattering were not. These data provide guiding principles in the development of clin. viable siRNA delivery materials and have the potential to reduce exptl. costs while improving the translation of materials into animals.
- 29Paunovska, K.; Sago, C. D.; Monaco, C. M.; Hudson, W. H.; Castro, M. G.; Rudoltz, T. G.; Kalathoor, S.; Vanover, D. A.; Santangelo, P. J.; Ahmed, R.; Bryksin, A. V.; Dahlman, J. E. A Direct Comparison of in Vitro and in Vivo Nucleic Acid Delivery Mediated by Hundreds of Nanoparticles Reveals a Weak Correlation. Nano Lett. 2018, 18 (3), 2148– 2157, DOI: 10.1021/acs.nanolett.8b00432Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjs1SqtLo%253D&md5=a951cf961c11aa4fb49ef2de448780edA direct comparison of in vitro and in vivo nucleic acid delivery mediated by hundreds of nanoparticles reveals a weak correlationPaunovska, Kalina; Sago, Cory D.; Monaco, Christopher M.; Hudson, William H.; Castro, Marielena Gamboa; Rudoltz, Tobi G.; Kalathoor, Sujay; Vanover, Daryll A.; Santangelo, Philip J.; Ahmed, Rafi; Bryksin, Anton V.; Dahlman, James E.Nano Letters (2018), 18 (3), 2148-2157CODEN: NALEFD; ISSN:1530-6984. (American Chemical Society)Endothelial cells and macrophages play active roles in disease and as a result are important targets for nucleic acid therapies. While thousands of chem. distinct lipid nanoparticles (LNPs) can be synthesized to deliver nucleic acids, studying more than a few LNPs in vivo is challenging. As a result, it is difficult to understand how nanoparticles target these cells in vivo. Using high throughput LNP barcoding, we quantified how well LNPs delivered DNA barcodes to endothelial cells and macrophages in vitro, as well as endothelial cells and macrophages isolated from the lung, heart, and bone marrow in vivo. We focused on two fundamental questions in drug delivery. First, does in vitro LNP delivery predict in vivo LNP delivery. By comparing how 281 LNPs delivered barcodes to endothelial cells and macrophages in vitro and in vivo, we found in vitro delivery did not predict in vivo delivery. Second, does LNP delivery change within the microenvironment of a tissue. We quantified how 85 LNPs delivered barcodes to eight splenic cell populations, and found that cell types derived from myeloid progenitors tended to be targeted by similar LNPs, relative to cell types derived from lymphoid progenitors. These data demonstrate that barcoded LNPs can elucidate fundamental questions about in vivo nanoparticle delivery.
Cited By
This article has not yet been cited by other publications.
Article Views
Altmetric
Citations
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. Centrifugal fractionation of LNPs based on their mRNA loading levels. (A) Schematic illustration of LNP fractionation via ultracentrifugation. Depending on the LNPs’ density contrast with the aqueous system, two fractions (LDLNP and HDLNP) are generated from the fractionation process. Together with the original batch, three samples are compared throughout this paper; (B) particle size distribution by intensity measured with DLS; payload mRNA integrity by Bioanalyzer shown as electropherograms in (C1) and bands in (C2).
Figure 2
Figure 2. Validation of centrifugal fractionation, characterization of lipid composition, and calculation of N/P ratio. (A) Bar chart of RNA/total lipid ratio to represent the mRNA loading levels and validate the fractionation method; (B) pie charts showing lipid composition of OLNP, LDLNP, and HDLNP as characterized by ultrahigh-performance liquid chromatography–mass spectrometry (UHPLC-MS).
Figure 3
Figure 3. mRNA loading levels can influence the distribution of LNP morphology. (A1–A3) Representative cryoEM images of OLNP, LDLNP, and HDLNP (scale bar: 100 nm); (B) pie charts showing the composition of LNPs with various number of blebs; (C) schematic illustration of measuring LNPs and blebs with FIJI; (D1–D3) scatter plots mapping the dimension of LNPs and blebs (purple dots: LNPs without blebs; green dots: LNPs with blebs) (E) raincloud plot showing the size distribution of blebs.
Figure 4
Figure 4. Cellular uptake and functional delivery of mRNA with OLNP, LDLNP, and HDLNP. (A) CLSM images of OLNP, LDLNP, and HDLNP ((A1), (A2), and (A3), respectively) delivering Cy5-labeled mEGFP into HEK293 cells (scale bar: 50 μm); (B1–B3) CLSM study on the colocalization between LNP colocalization and Rab7a, representing late endosomes (scale bar: 50 μm).
Figure 5
Figure 5. In vivo functional delivery of mFluc. (A) Schematic illustration of the animal study; (B) in vivo bioluminescence images of mice at 3 h after IV injection at mFluc dosage of 0.3 mg/kg (n = 4) and bar chart showing whole-body bioluminescence quantification from IVIS imaging. Data are presented as the mean ± standard deviation (SD) (n = 4). (C) Representative ex vivo bioluminescence images of extracted livers and spleens from mice dosed with OLNP/LDLNP/HDLNP (top to bottom) and bar chart showing bioluminescence of extracted livers and spleens. Data are presented as the mean ± SD (n = 4). (D) Scatter plot by projecting ex vivo liver radiance readout against spleen readout to indicate tissue tropism.
References
This article references 29 other publications.
- 1Akinc, A.; Maier, M. A.; Manoharan, M.; Fitzgerald, K.; Jayaraman, M.; Barros, S.; Ansell, S.; Du, X.; Hope, M. J.; Madden, T. D.; Mui, B. L.; Semple, S. C.; Tam, Y. K.; Ciufolini, M.; Witzigmann, D.; Kulkarni, J. A.; van der Meel, R.; Cullis, P. R. The Onpattro Story and the Clinical Translation of Nanomedicines Containing Nucleic Acid-Based Drugs. Nat. Nanotechnol. 2019, 14 (12), 1084– 1087, DOI: 10.1038/s41565-019-0591-y1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlWit7vO&md5=3184ac1c3c74e6924fb1bb14b5229e95The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugsAkinc, Akin; Maier, Martin A.; Manoharan, Muthiah; Fitzgerald, Kevin; Jayaraman, Muthusamy; Barros, Scott; Ansell, Steven; Du, Xinyao; Hope, Michael J.; Madden, Thomas D.; Mui, Barbara L.; Semple, Sean C.; Tam, Ying K.; Ciufolini, Marco; Witzigmann, Dominik; Kulkarni, Jayesh A.; van der Meel, Roy; Cullis, Pieter R.Nature Nanotechnology (2019), 14 (12), 1084-1087CODEN: NNAABX; ISSN:1748-3387. (Nature Research)The regulatory approval of Onpattro, a lipid nanoparticle-based short interfering RNA drug for the treatment of polyneuropathies induced by hereditary transthyretin amyloidosis, paves the way for clin. development of many nucleic acid-based therapies enabled by nanoparticle delivery.
- 2Polack, F. P.; Thomas, S. J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J. L.; Pérez Marc, G.; Moreira, E. D.; Zerbini, C.; Bailey, R.; Swanson, K. A.; Roychoudhury, S.; Koury, K.; Li, P.; Kalina, W. V.; Cooper, D.; Frenck, R. W.; Hammitt, L. L.; Türeci, Ö.; Nell, H.; Schaefer, A.; Ünal, S.; Tresnan, D. B.; Mather, S.; Dormitzer, P. R.; Şahin, U.; Jansen, K. U.; Gruber, W. C. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N. Engl. J. Med. 2020, 383 (27), 2603– 2615, DOI: 10.1056/NEJMoa20345772https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXotFSjuw%253D%253D&md5=08cb7204334ed37c7949dfcdfe5af75cSafety and efficacy of the BNT162b2 mRNA Covid-19 vaccinePolack, Fernando P.; Thomas, Stephen J.; Kitchin, Nicholas; Absalon, Judith; Gurtman, Alejandra; Lockhart, Stephen; Perez, John L.; Marc, Gonzalo Perez; Moreira, Edson D.; Zerbini, Cristiano; Bailey, Ruth; Swanson, Kena A.; Roychoudhury, Satrajit; Koury, Kenneth; Li, Ping; Kalina, Warren V.; Cooper, David; Frenck, Robert W., Jr.; Hammitt, Laura L.; Tureci, Ozlem; Nell, Haylene; Schaefer, Axel; Unal, Serhat; Tresnan, Dina B.; Mather, Susan; Dormitzer, Philip R.; Sahin, Ugur; Jansen, Kathrin U.; Gruber, William C.New England Journal of Medicine (2020), 383 (27), 2603-2615CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)A review. Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting coronavirus disease 2019 (Covid-19) have afflicted tens of millions of people in a worldwide pandemic. Safe and effective vaccines are needed urgently. methods In an ongoing multinational, placebo-controlled, observer-blinded, pivotal efficacy trial, we randomly assigned persons 16 years of age or older in a 1:1 ratio to receive two doses, 21 days apart, of either placebo or the BNT162b2 vaccine candidate (30μg per dose). BNT162b2 is a lipid nanoparticle-formulated, nucleoside-modified RNA vaccine that encodes a prefusion stabilized, membrane-anchored SARS-CoV-2 full-length spike protein. The primary end points were efficacy of the vaccine against lab.-confirmed Covid-19 and safety. results A total of 43,548 participants underwent randomization, of whom 43,448 received injections: 21,720 with BNT162b2 and 21,728 with placebo. There were 8 cases of Covid-19 with onset at least 7 days after the second dose among participants assigned to receive BNT162b2 and 162 cases among those assigned to placebo; BNT162b2 was 95% effective in preventing Covid-19 (95% credible interval, 90.3 to 97.6). Similar vaccine efficacy (generally 90 to 100%) was obsd. across subgroups defined by age, sex, race, ethnicity, baseline body-mass index, and the presence of coexisting conditions. Among 10 cases of severe Covid-19 with onset after the first dose, 9 occurred in placebo recipients and 1 in a BNT162b2 recipient. The safety profile of BNT162b2 was characterized by short-term, mild-to-moderate pain at the injection site, fatigue, and headache. The incidence of serious adverse events was low and was similar in the vaccine and placebo groups. conclusions A two-dose regimen of BNT162b2 conferred 95% protection against Covid-19 in persons 16 years of age or older. Safety over a median of 2 mo was similar to that of other viral vaccines.
- 3Baden, L. R.; El Sahly, H. M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S. A.; Rouphael, N.; Creech, C. B.; McGettigan, J.; Khetan, S.; Segall, N.; Solis, J.; Brosz, A.; Fierro, C.; Schwartz, H.; Neuzil, K.; Corey, L.; Gilbert, P.; Janes, H.; Follmann, D.; Marovich, M.; Mascola, J.; Polakowski, L.; Ledgerwood, J.; Graham, B. S.; Bennett, H.; Pajon, R.; Knightly, C.; Leav, B.; Deng, W.; Zhou, H.; Han, S.; Ivarsson, M.; Miller, J.; Zaks, T. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N. Engl. J. Med. 2021, 384 (5), 403– 416, DOI: 10.1056/NEJMoa20353893https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjvFSmsrc%253D&md5=9b0c79f3e3fd2ea452a3fbf57281720fEfficacy and safety of the mRNA-1273 SARS-CoV-2 vaccineBaden, L. R.; El Sahly, H. M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S. A.; Rouphael, N.; Creech, C. B.; McGettigan, J.; Khetan, S.; Segall, N.; Solis, J.; Brosz, A.; Fierro, C.; Schwartz, H.; Neuzil, K.; Corey, L.; Gilbert, P.; Janes, H.; Follmann, D.; Marovich, M.; Mascola, J.; Polakowski, L.; Ledgerwood, J.; Graham, B. S.; Bennett, H.; Pajon, R.; Knightly, C.; Leav, B.; Deng, W.; Zhou, H.; Han, S.; Ivarsson, M.; Miller, J.; Zaks, T.New England Journal of Medicine (2021), 384 (5), 403-416CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)Vaccines are needed to prevent coronavirus disease 2019 (Covid-19) and to protect persons who are at high risk for complications. The mRNA-1273 vaccine is a lipid nanoparticle-encapsulated mRNA-based vaccine that encodes the prefusion stabilized full-length spike protein of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes Covid-19. This phase 3 randomized, observer-blinded, placebo-controlled trial was conducted at 99 centers across the United States. Persons at high risk for SARS-CoV-2 infection or its complications were randomly assigned in a 1:1 ratio to receive two i.m. injections of mRNA-1273 (100μg) or placebo 28 days apart. The primary end point was prevention of Covid-19 illness with onset at least 14 days after the second injection in participants who had not previously been infected with SARS-CoV-2. The trial enrolled 30,420 volunteers who were randomly assigned in a 1:1 ratio to receive either vaccine or placebo (15,210 participants in each group). More than 96% of participants received both injections, and 2.2% had evidence (serol., virol., or both) of SARS-CoV-2 infection at baseline. Symptomatic Covid-19 illness was confirmed in 185 participants in the placebo group (56.5 per 1000 person-years; 95% confidence interval [CI], 48.7 to 65.3) and in 11 participants in the mRNA-1273 group (3.3 per 1000 person-years; 95% CI, 1.7 to 6.0); vaccine efficacy was 94.1% (95% CI, 89.3 to 96.8%; P<0.001). Efficacy was similar across key secondary analyses, including assessment 14 days after the first dose, analyses that included participants who had evidence of SARS-CoV-2 infection at baseline, and analyses in participants 65 years of age or older. Severe Covid-19 occurred in 30 participants, with one fatality; all 30 were in the placebo group. Moderate, transient reactogenicity after vaccination occurred more frequently in the mRNA-1273 group. Serious adverse events were rare, and the incidence was similar in the two groups. The mRNA-1273 vaccine showed 94.1% efficacy at preventing Covid-19 illness, including severe disease. Aside from transient local and systemic reactions, no safety concerns were identified.
- 4Wilson, E.; Goswami, J.; Baqui, A. H.; Doreski, P. A.; Perez-Marc, G.; Zaman, K.; Monroy, J.; Duncan, C. J. A.; Ujiie, M.; Rämet, M.; Pérez-Breva, L.; Falsey, A. R.; Walsh, E. E.; Dhar, R.; Wilson, L.; Du, J.; Ghaswalla, P.; Kapoor, A.; Lan, L.; Mehta, S.; Mithani, R.; Panozzo, C. A.; Simorellis, A. K.; Kuter, B. J.; Schödel, F.; Huang, W.; Reuter, C.; Slobod, K.; Stoszek, S. K.; Shaw, C. A.; Miller, J. M.; Das, R.; Chen, G. L. Efficacy and Safety of an mRNA-Based RSV PreF Vaccine in Older Adults. N. Engl. J. Med. 2023, 389 (24), 2233– 2244, DOI: 10.1056/NEJMoa2307079There is no corresponding record for this reference.
- 5Kulkarni, J. A.; Witzigmann, D.; Leung, J.; van der Meel, R.; Zaifman, J.; Darjuan, M. M.; Grisch-Chan, H. M.; Thöny, B.; Tam, Y. Y. C.; Cullis, P. R. Fusion-Dependent Formation of Lipid Nanoparticles Containing Macromolecular Payloads. Nanoscale 2019, 11 (18), 9023– 9031, DOI: 10.1039/C9NR02004G5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXnvFymtLk%253D&md5=f588b6991dd4e67e61e1f286fa3c2b4fFusion-dependent formation of lipid nanoparticles containing macromolecular payloadsKulkarni, Jayesh A.; Witzigmann, Dominik; Leung, Jerry; van der Meel, Roy; Zaifman, Josh; Darjuan, Maria M.; Grisch-Chan, Hiu Man; Thony, Beat; Tam, Yuen Yi C.; Cullis, Pieter R.Nanoscale (2019), 11 (18), 9023-9031CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)The success of Onpattro (patisiran) clearly demonstrates the utility of lipid nanoparticle (LNP) systems for enabling gene therapies. These systems are composed of ionizable cationic lipids, phospholipid, cholesterol, and polyethylene glycol (PEG)-lipids, and are produced through rapid-mixing of an ethanolic-lipid soln. with an acidic aq. soln. followed by dialysis into neutralizing buffer. A detailed understanding of the mechanism of LNP formation is crucial to improving LNP design. Here we use cryogenic transmission electron microscopy and fluorescence techniques to further demonstrate that LNP are formed through the fusion of precursor, pH-sensitive liposomes into large electron-dense core structures as the pH is neutralized. Next, we show that the fusion process is limited by the accumulation of PEG-lipid on the emerging particle. Finally, we show that the fusion-dependent mechanism of formation also applies to LNP contg. macromol. payloads including mRNA, DNA vectors, and gold nanoparticles.
- 6Li, S.; Hu, Y.; Lin, J.; Schneiderman, Z.; Shao, F.; Wei, L.; Li, A.; Hsieh, K.; Kokkoli, E.; Curk, T.; Mao, H.-Q.; Wang, T.-H. Single-Particle Spectroscopic Chromatography Reveals Heterogeneous RNA Loading and Size Correlations in Lipid Nanoparticles. ACS Nano 2024, 18, 15729, DOI: 10.1021/acsnano.4c02341There is no corresponding record for this reference.
- 7Li, S.; Hu, Y.; Li, A.; Lin, J.; Hsieh, K.; Schneiderman, Z.; Zhang, P.; Zhu, Y.; Qiu, C.; Kokkoli, E.; Wang, T.-H.; Mao, H.-Q. Payload Distribution and Capacity of mRNA Lipid Nanoparticles. Nat. Commun. 2022, 13 (1), 5561 DOI: 10.1038/s41467-022-33157-47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XisVyrsL7J&md5=df0c413e8724a661b328523b99a7d4b1Payload distribution and capacity of mRNA lipid nanoparticlesLi, Sixuan; Hu, Yizong; Li, Andrew; Lin, Jinghan; Hsieh, Kuangwen; Schneiderman, Zachary; Zhang, Pengfei; Zhu, Yining; Qiu, Chenhu; Kokkoli, Efrosini; Wang, Tza-Huei; Mao, Hai-QuanNature Communications (2022), 13 (1), 5561CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)Lipid nanoparticles (LNPs) are effective vehicles to deliver mRNA vaccines and therapeutics. It has been challenging to assess mRNA packaging characteristics in LNPs, including payload distribution and capacity, which are crit. to understanding structure-property-function relationships for further carrier development. Here, we report a method based on the multi-laser cylindrical illumination confocal spectroscopy (CICS) technique to examine mRNA and lipid contents in LNP formulations at the single-nanoparticle level. By differentiating unencapsulated mRNAs, empty LNPs and mRNA-loaded LNPs via coincidence anal. of fluorescent tags on different LNP components, and quant. resolving single-mRNA fluorescence, we reveal that a commonly referenced benchmark formulation using DLin-MC3 as the ionizable lipid contains mostly 2 mRNAs per loaded LNP with a presence of 40%-80% empty LNPs depending on the assembly conditions. Systematic anal. of different formulations with control variables reveals a kinetically controlled assembly mechanism that governs the payload distribution and capacity in LNPs. These results form the foundation for a holistic understanding of the mol. assembly of mRNA LNPs.
- 8De Kloet, S. R.; Andrean, B. A. G. Buoyant Density Gradient Centrifugation of RNA and DNA in Alkali Iodide Solutions. Biochim. Biophys. Acta, Nucleic Acids Protein Synth. 1971, 247 (4), 519– 527, DOI: 10.1016/0005-2787(71)90688-5There is no corresponding record for this reference.
- 9Guerrini, G.; Mehn, D.; Scaccabarozzi, D.; Gioria, S.; Calzolai, L. Analytical Ultracentrifugation to Assess the Quality of LNP-mRNA Therapeutics. Int. J. Mol. Sci. 2024, 25 (11), 5718 DOI: 10.3390/ijms25115718There is no corresponding record for this reference.
- 10Schiel, J. E.; Hage, D. S. Density Measurements of Potassium Phosphate Buffer from 4 to 45 °C. Talanta 2005, 65 (2), 495– 500, DOI: 10.1016/j.talanta.2004.06.029There is no corresponding record for this reference.
- 11Henrickson, A.; Kulkarni, J. A.; Zaifman, J.; Gorbet, G. E.; Cullis, P. R.; Demeler, B. Density Matching Multi-Wavelength Analytical Ultracentrifugation to Measure Drug Loading of Lipid Nanoparticle Formulations. ACS Nano 2021, 15, 5068, DOI: 10.1021/acsnano.0c1006911https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXks1yjtLg%253D&md5=2b8a668202896863661f0971b05d51e3Density Matching Multi-wavelength Analytical Ultracentrifugation to Measure Drug Loading of Lipid Nanoparticle FormulationsHenrickson, Amy; Kulkarni, Jayesh A.; Zaifman, Josh; Gorbet, Gary E.; Cullis, Pieter R.; Demeler, BorriesACS Nano (2021), 15 (3), 5068-5076CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Previous work suggested that lipid nanoparticle (LNP) formulations, encapsulating nucleic acids, display electron-dense morphol. when examd. by cryogenic-transmission electron microscopy (cryo-TEM). Critically, the employed cryo-TEM method cannot differentiate between loaded and empty LNP formulations. Clin. relevant formulations contain high lipid-to-nucleic acid ratios (10-25 (wt./wt.)), and for systems that contain mRNA or DNA, it is anticipated that a substantial fraction of the LNP population does not contain a payload. Here, we present a method based on the global anal. of multi-wavelength sedimentation velocity anal. ultracentrifugation, using d. matching with heavy water, that not only measures the std. sedimentation and diffusion coeff. distributions of LNP mixts., but also reports the corresponding partial sp. vol. distributions and optically separates signal contributions from nucleic acid cargo and lipid shell. This makes it possible to reliably predict molar mass and anisotropy distributions, in particular, for systems that are heterogeneous in partial sp. vol. and have low to intermediate densities. Our method makes it possible to unambiguously measure the d. of nanoparticles and is motivated by the need to characterize the extent to which lipid nanoparticles are loaded with nucleic acid cargoes. Since the densities of nucleic acids and lipids substantially differ, the measured d. is directly proportional to the loading of nanoparticles. Hence, different loading levels will produce particles with variable d. and partial sp. vol. An UltraScan software module was developed to implement this approach for routine anal.
- 12Bepperling, A.; Richter, G. Determination of mRNA Copy Number in Degradable Lipid Nanoparticles via Density Contrast Analytical Ultracentrifugation. Eur. Biophys J. 2023, 52, 393, DOI: 10.1007/s00249-023-01663-y12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXhtF2iurnO&md5=ae9b4805103592dc6f27bfd07cef7c5bDetermination of mRNA copy number in degradable lipid nanoparticles via density contrast analytical ultracentrifugationBepperling, Alexander; Richter, GesaEuropean Biophysics Journal (2023), 52 (4-5), 393-400CODEN: EBJOE8; ISSN:0175-7571. (Springer International Publishing AG)Lipid nanoparticles as delivery system for mRNA have recently attracted attention to a broader audience as COVID-19 mRNA vaccines. Their low immunogenicity and capability to deliver a variety of nucleic acids renders them an interesting and complementary alternative to gene therapy vectors like AAVs. An important quality attribute of LNPs is the copy no. of the encapsulated cargo mol. This work describes how d. and mol. wt. distributions obtained by d. contrast sedimentation velocity can be used to calc. the mRNA copy no. of a degradable lipid nanoparticle formulation. The detd. av. copy no. of 5 mRNA mols. per LNP is consistent with the previous studies using other biophys. techniques, such as single particle imaging microscopy and multi-laser cylindrical illumination confocal spectroscopy (CICS).
- 13Leung, A. K. K.; Hafez, I. M.; Baoukina, S.; Belliveau, N. M.; Zhigaltsev, I. V.; Afshinmanesh, E.; Tieleman, D. P.; Hansen, C. L.; Hope, M. J.; Cullis, P. R. Lipid Nanoparticles Containing siRNA Synthesized by Microfluidic Mixing Exhibit an Electron-Dense Nanostructured Core. J. Phys. Chem. C 2012, 116 (34), 18440– 18450, DOI: 10.1021/jp303267y13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVOjs7zL&md5=0f25938ac9ee92e7c8dc34ccc0d0f27aLipid Nanoparticles Containing siRNA Synthesized by Microfluidic Mixing Exhibit an Electron-Dense Nanostructured CoreLeung, Alex K. K.; Hafez, Ismail M.; Baoukina, Svetlana; Belliveau, Nathan M.; Zhigaltsev, Igor V.; Afshinmanesh, Elham; Tieleman, D. Peter; Hansen, Carl L.; Hope, Michael J.; Cullis, Pieter R.Journal of Physical Chemistry C (2012), 116 (34), 18440-18450CODEN: JPCCCK; ISSN:1932-7447. (American Chemical Society)Lipid nanoparticles (LNP) contg. ionizable cationic lipids are the leading systems for enabling therapeutic applications of siRNA; however, the structure of these systems has not been defined. Here we examine the structure of LNP siRNA systems contg. DLinKC2-DMA(an ionizable cationic lipid), phospholipid, cholesterol and a polyethylene glycol (PEG) lipid formed using a rapid microfluidic mixing process. Techniques employed include cryo-transmission electron microscopy, 31P NMR, membrane fusion assays, d. measurements, and mol. modeling. The exptl. results indicate that these LNP siRNA systems have an interior lipid core contg. siRNA duplexes complexed to cationic lipid and that the interior core also contains phospholipid and cholesterol. Consistent with exptl. observations, mol. modeling calcns. indicate that the interior of LNP siRNA systems exhibits a periodic structure of aq. compartments, where some compartments contain siRNA. It is concluded that LNP siRNA systems formulated by rapid mixing of an ethanol soln. of lipid with an aq. medium contg. siRNA exhibit a nanostructured core. The results give insight into the mechanism whereby LNP siRNA systems are formed, providing an understanding of the high encapsulation efficiencies that can be achieved and information on methods of constructing more sophisticated LNP systems.
- 14Vaidya, A.; Parande, D.; Khadse, N.; Vargas-Montoya, N.; Agarwal, V.; Ortiz, C.; Ellis, G.; Kaushal, N.; Sarode, A.; Karve, S.; DeRosa, F. Analytical Characterization of Heterogeneities in mRNA-Lipid Nanoparticles Using Sucrose Density Gradient Ultracentrifugation. Anal. Chem. 2024, 96, 5570, DOI: 10.1021/acs.analchem.4c00031There is no corresponding record for this reference.
- 15Zhao, H.; Sousa, A. A.; Schuck, P. Flotation Coefficient Distributions of Lipid Nanoparticles by Sedimentation Velocity Analytical Ultracentrifugation. ACS Nano 2024, 18, 18663, DOI: 10.1021/acsnano.4c05322There is no corresponding record for this reference.
- 16Semple, S. C.; Akinc, A.; Chen, J.; Sandhu, A. P.; Mui, B. L.; Cho, C. K.; Sah, D. W. Y.; Stebbing, D.; Crosley, E. J.; Yaworski, E.; Hafez, I. M.; Dorkin, J. R.; Qin, J.; Lam, K.; Rajeev, K. G.; Wong, K. F.; Jeffs, L. B.; Nechev, L.; Eisenhardt, M. L.; Jayaraman, M.; Kazem, M.; Maier, M. A.; Srinivasulu, M.; Weinstein, M. J.; Chen, Q.; Alvarez, R.; Barros, S. A.; De, S.; Klimuk, S. K.; Borland, T.; Kosovrasti, V.; Cantley, W. L.; Tam, Y. K.; Manoharan, M.; Ciufolini, M. A.; Tracy, M. A.; de Fougerolles, A.; MacLachlan, I.; Cullis, P. R.; Madden, T. D.; Hope, M. J. Rational Design of Cationic Lipids for siRNA Delivery. Nat. Biotechnol. 2010, 28 (2), 172– 176, DOI: 10.1038/nbt.160216https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXntVCqtg%253D%253D&md5=8ed6986f3ed898efe69f4576f762b3adRational design of cationic lipids for siRNA deliverySemple, Sean C.; Akinc, Akin; Chen, Jianxin; Sandhu, Ammen P.; Mui, Barbara L.; Cho, Connie K.; Sah, Dinah W. Y.; Stebbing, Derrick; Crosley, Erin J.; Yaworski, Ed; Hafez, Ismail M.; Dorkin, J. Robert; Qin, June; Lam, Kieu; Rajeev, Kallanthottathil G.; Wong, Kim F.; Jeffs, Lloyd B.; Nechev, Lubomir; Eisenhardt, Merete L.; Jayaraman, Muthusamy; Kazem, Mikameh; Maier, Martin A.; Srinivasulu, Masuna; Weinstein, Michael J.; Chen, Qingmin; Alvarez, Rene; Barros, Scott A.; De, Soma; Klimuk, Sandra K.; Borland, Todd; Kosovrasti, Verbena; Cantley, William L.; Tam, Ying K.; Manoharan, Muthiah; Ciufolini, Marco A.; Tracy, Mark A.; de Fougerolles, Antonin; MacLachlan, Ian; Cullis, Pieter R.; Madden, Thomas D.; Hope, Michael J.Nature Biotechnology (2010), 28 (2), 172-176CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)We adopted a rational approach to design cationic lipids for use in formulations to deliver small interfering RNA (siRNA). Starting with the ionizable cationic lipid 1,2-dilinoleyloxy-3-dimethylaminopropane (DLinDMA), a key lipid component of stable nucleic acid lipid particles (SNALP) as a benchmark, we used the proposed in vivo mechanism of action of ionizable cationic lipids to guide the design of DLinDMA-based lipids with superior delivery capacity. The best-performing lipid recovered after screening (DLin-KC2-DMA) was formulated and characterized in SNALP and demonstrated to have in vivo activity at siRNA doses as low as 0.01 mg/kg in rodents and 0.1 mg/kg in nonhuman primates. To our knowledge, this represents a substantial improvement over previous reports of in vivo endogenous hepatic gene silencing.
- 17Svedberg, T.; Rinde, H. The Ultra-centrifuge, a New Instrument for the Determination of Size and Distribution of Size of Particle In Amicroscopic Colloids. J. Am. Chem. Soc. 1924, 46 (12), 2677– 2693, DOI: 10.1021/ja01677a011There is no corresponding record for this reference.
- 18Kim, J.; Eygeris, Y.; Gupta, M.; Sahay, G. Self-Assembled mRNA Vaccines. Adv. Drug Delivery Rev. 2021, 170, 83, DOI: 10.1016/j.addr.2020.12.01418https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslWgtr0%253D&md5=d213939af17262d61f009547d94627feSelf-assembled mRNA vaccinesKim, Jeonghwan; Eygeris, Yulia; Gupta, Mohit; Sahay, GauravAdvanced Drug Delivery Reviews (2021), 170 (), 83-112CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. MRNA vaccines have evolved from being a mere curiosity to emerging as COVID-19 vaccine front-runners. Recent advancements in the field of RNA technol., vaccinol., and nanotechnol. have generated interest in delivering safe and effective mRNA therapeutics. In this review, we discuss design and self-assembly of mRNA vaccines. Self-assembly, a spontaneous organization of individual mols., allows for design of nanoparticles with customizable properties. We highlight the materials commonly utilized to deliver mRNA, their physicochem. characteristics, and other relevant considerations, such as mRNA optimization, routes of administration, cellular fate, and immune activation, that are important for successful mRNA vaccination. We also examine the COVID-19 mRNA vaccines currently in clin. trials. mRNA vaccines are ready for the clinic, showing tremendous promise in the COVID-19 vaccine race, and have pushed the boundaries of gene therapy.
- 19Gilbert, J.; Sebastiani, F.; Arteta, M. Y.; Terry, A.; Fornell, A.; Russell, R.; Mahmoudi, N.; Nylander, T. Evolution of the Structure of Lipid Nanoparticles for Nucleic Acid Delivery: From in Situ Studies of Formulation to Colloidal Stability. J. Colloid Interface Sci. 2024, 660, 66– 76, DOI: 10.1016/j.jcis.2023.12.165There is no corresponding record for this reference.
- 20Szebeni, J.; Kiss, B.; Bozó, T.; Turjeman, K.; Levi-Kalisman, Y.; Barenholz, Y.; Kellermayer, M. Insights into the Structure of Comirnaty Covid-19 Vaccine: A Theory on Soft, Partially Bilayer-Covered Nanoparticles with Hydrogen Bond-Stabilized mRNA–Lipid Complexes. ACS Nano 2023, 17 (14), 13147– 13157, DOI: 10.1021/acsnano.2c11904There is no corresponding record for this reference.
- 21Forchette, L.; Sebastian, W.; Liu, T. A Comprehensive Review of COVID-19 Virology, Vaccines, Variants, and Therapeutics. Curr. Med. Sci. 2021, 41 (6), 1037– 1051, DOI: 10.1007/s11596-021-2395-121https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsFGhurbE&md5=d90b78150fd6761fc4a02e09ff4d1254A Comprehensive Review of COVID-19 Virology, Vaccines, Variants, and TherapeuticsForchette, Lauren; Sebastian, William; Liu, TuoenCurrent Medical Science (2021), 41 (6), 1037-1051CODEN: CMSUCM; ISSN:2523-899X. (Huazhong University of Science and Technology)A review. Summary: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative pathogen of the coronavirus disease 2019 (COVID-19), has caused more than 179 million infections and 3.8 million deaths worldwide. Throughout the past year, multiple vaccines have already been developed and used, while some others are in the process of being developed. However, the emergence of new mutant strains of SARS-CoV-2 that have demonstrated immune-evading characteristics and an increase in infective capabilities leads to potential ineffectiveness of the vaccines against these variants. The purpose of this review article is to highlight the current understanding of the immunol. mechanisms of the virus and vaccines, as well as to investigate some key variants and mutations of the virus driving the current pandemic and their impacts on current management guidelines. We also discussed new technologies being developed for the prevention, treatment, and detection of SARS-CoV-2. In this paper, we thoroughly reviewed and provided crucial information on SARS-CoV-2 virol., vaccines and drugs being used and developed for its prevention and treatment, as well as important variant strains. Our review paper will be beneficial to health care professionals and researchers so they can have a better understanding of the basic sciences, prevention, and clin. treatment of COVID-19 during the pandemic. This paper consists of the most updated information that has been available as of June 21, 2021.
- 22Heinz, F. X.; Stiasny, K. Distinguishing Features of Current COVID-19 Vaccines: Knowns and Unknowns of Antigen Presentation and Modes of Action. npj Vaccines 2021, 6 (1), 104 DOI: 10.1038/s41541-021-00369-622https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVemurzL&md5=818b1fef015ae9bbffda5db8b50a7f09Distinguishing features of current COVID-19 vaccines: knowns and unknowns of antigen presentation and modes of actionHeinz, Franz X.; Stiasny, Karinnpj Vaccines (2021), 6 (1), 104CODEN: VACCBC; ISSN:2059-0105. (Nature Research)A review. COVID-19 vaccines were developed with an unprecedented pace since the beginning of the pandemic. Several of them have reached market authorization and mass prodn., leading to their global application on a large scale. This enormous progress was achieved with fundamentally different vaccine technologies used in parallel. mRNA, adenoviral vector as well as inactivated whole-virus vaccines are now in widespread use, and a subunit vaccine is in a final stage of authorization. They all rely on the native viral spike protein (S) of SARS-CoV-2 for inducing potently neutralizing antibodies, but the presentation of this key antigen to the immune system differs substantially between the different categories of vaccines. In this article, we review the relevance of structural modifications of S in different vaccines and the different modes of antigen expression after vaccination with genetic adenovirus-vector and mRNA vaccines. Distinguishing characteristics and unknown features are highlighted in the context of protective antibody responses and reactogenicity of vaccines.
- 23Buschmann, M. D.; Carrasco, M. J.; Alishetty, S.; Paige, M.; Alameh, M. G.; Weissman, D. Nanomaterial Delivery Systems for mRNA Vaccines. Vaccines 2021, 9 (1), 65 DOI: 10.3390/vaccines901006523https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhs1GgsrjP&md5=e09076471c269d29078c6b2445d542d8Nanomaterial delivery systems for mRNA vaccinesBuschmann, Michael D.; Carrasco, Manuel J.; Alishetty, Suman; Paige, Mikell; Alameh, Mohamad Gabriel; Weissman, DrewVaccines (Basel, Switzerland) (2021), 9 (1), 65CODEN: VBSABP; ISSN:2076-393X. (MDPI AG)The recent success of mRNA vaccines in SARS-CoV-2 clin. trials is in part due to the development of lipid nanoparticle delivery systems that not only efficiently express the mRNA-encoded immunogen after i.m. injection, but also play roles as adjuvants and in vaccine reactogenicity. We present an overview of mRNA delivery systems and then focus on the lipid nanoparticles used in the current SARS-CoV-2 vaccine clin. trials. The review concludes with an anal. of the determinants of the performance of lipid nanoparticles in mRNA vaccines.
- 24Tenchov, R.; Bird, R.; Curtze, A. E.; Zhou, Q. Lipid Nanoparticles─From Liposomes to mRNA Vaccine Delivery, a Landscape of Research Diversity and Advancement. ACS Nano 2021, 15 (11), 16982– 17015, DOI: 10.1021/acsnano.1c0499624https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsVSqsr%252FM&md5=c824f1ae324b984f94d2e33741ceef8bLipid Nanoparticles-From Liposomes to mRNA Vaccine Delivery, a Landscape of Research Diversity and AdvancementTenchov, Rumiana; Bird, Robert; Curtze, Allison E.; Zhou, QiongqiongACS Nano (2021), 15 (11), 16982-17015CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)A review. Lipid nanoparticles (LNPs) have emerged across the pharmaceutical industry as promising vehicles to deliver a variety of therapeutics. Currently in the spotlight as vital components of the COVID-19 mRNA vaccines, LNPs play a key role in effectively protecting and transporting mRNA to cells. Liposomes, an early version of LNPs, are a versatile nanomedicine delivery platform. A no. of liposomal drugs have been approved and applied to medical practice. Subsequent generations of lipid nanocarriers, such as solid lipid nanoparticles, nanostructured lipid carriers, and cationic lipid-nucleic acid complexes, exhibit more complex architectures and enhanced phys. stabilities. With their ability to encapsulate and deliver therapeutics to specific locations within the body and to release their contents at a desired time, LNPs provide a valuable platform for treatment of a variety of diseases. We present a landscape of LNP-related scientific publications, including patents and journal articles, based on anal. of the CAS Content Collection, the largest human-curated collection of published scientific knowledge. Rising trends are identified, such as nanostructured lipid carriers and solid lipid nanoparticles becoming the preferred platforms for numerous formulations. Recent advancements in LNP formulations as drug delivery platforms, such as antitumor and nucleic acid therapeutics and vaccine delivery systems, are discussed. Challenges and growth opportunities are also evaluated in other areas, such as medical imaging, cosmetics, nutrition, and agrochems. This report is intended to serve as a useful resource for those interested in LNP nanotechnologies, their applications, and the global research effort for their development.
- 25Kloczewiak, M.; Banks, J. M.; Jin, L.; Brader, M. L. A Biopharmaceutical Perspective on Higher-Order Structure and Thermal Stability of mRNA Vaccines. Mol. Pharmaceutics 2022, 19 (7), 2022– 2031, DOI: 10.1021/acs.molpharmaceut.2c0009225https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsFejt7vM&md5=ce77c9d38757cb5a47a66a583008e2d9A Biopharmaceutical Perspective on Higher-Order Structure and Thermal Stability of mRNA VaccinesKloczewiak, Marek; Banks, Jessica M.; Jin, Lin; Brader, Mark L.Molecular Pharmaceutics (2022), 19 (7), 2022-2031CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)A review. Preservation of the integrity of macromol. higher-order structure is a tenet central to achieving biol. drug and vaccine product stability toward manufg., 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 biol. drugs, at least at 1st 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 biol. 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 mols. We extend those findings by considering the effect of encapsulation on mRNA thermal unfolding with the observation that encapsulation in LNPs increases mRNA unfolding temps.
- 26Brader, M. L.; Williams, S. J.; Banks, J. M.; Hui, W. H.; Zhou, Z. H.; Jin, L. Encapsulation State of Messenger RNA inside Lipid Nanoparticles. Biophys. J. 2021, 120 (14), 2766– 2770, DOI: 10.1016/j.bpj.2021.03.01226https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXns1Cjsbc%253D&md5=dca599c7e6b5129440e6ce1b9ff78b44Encapsulation state of messenger RNA inside lipid nanoparticlesBrader, Mark L.; Williams, Sean J.; Banks, Jessica M.; Hui, Wong H.; Zhou, Z. Hong; Jin, LinBiophysical Journal (2021), 120 (14), 2766-2770CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)Understanding the structure of mRNA (mRNA) lipid nanoparticles, and specifically the microenvironment of the mRNA mols. within these entities, is fundamental to advancing their biomedical potential. Here, we show that a permeating cationic dye, thionine, can serve as a cryogenic electron microscopy contrasting agent by binding selectively to encapsulated mRNA without disturbing lipid nanoparticle morphol. Cryo-electron microscopy images identify the mRNA location, revealing that mRNA may exist within solvent-filled cavities or may be substantially lipid assocd.
- 27Cheng, M. H. Y.; Leung, J.; Zhang, Y.; Strong, C.; Basha, G.; Momeni, A.; Chen, Y.; Jan, E.; Abdolahzadeh, A.; Wang, X.; Kulkarni, J. A.; Witzigmann, D.; Cullis, P. R. Induction of Bleb Structures in Lipid Nanoparticle Formulations of mRNA Leads to Improved Transfection Potency. Adv. Mater. 2023, 35 (31), 2303370 DOI: 10.1002/adma.20230337027https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXhtlSqtrnE&md5=6fadec23ad601f3cc8f817c2c8709265Induction of Bleb Structures in Lipid Nanoparticle Formulations of mRNA Leads to Improved Transfection PotencyCheng, Miffy Hok Yan; Leung, Jerry; Zhang, Yao; Strong, Colton; Basha, Genc; Momeni, Arash; Chen, Yihang; Jan, Eric; Abdolahzadeh, Amir; Wang, Xinying; Kulkarni, Jayesh A.; Witzigmann, Dominik; Cullis, Pieter R.Advanced Materials (Weinheim, Germany) (2023), 35 (31), 2303370CODEN: ADVMEW; ISSN:0935-9648. (Wiley-VCH Verlag GmbH & Co. KGaA)The transfection potency of lipid nanoparticle (LNP) mRNA systems is critically dependent on the ionizable cationic lipid component. LNP mRNA systems composed of optimized ionizable lipids often display distinctive mRNA-rich "bleb" structures. Here, it is shown that such structures can also be induced for LNPs contg. nominally less active ionizable lipids by formulating them in the presence of high concns. of pH 4 buffers such as sodium citrate, leading to improved transfection potencies both in vitro and in vivo. Induction of bleb structure and improved potency is dependent on the type of pH 4 buffer employed, with LNP mRNA systems prepd. using 300 mM sodium citrate buffer displaying max. transfection. The improved transfection potencies of LNP mRNA systems displaying bleb structure can be attributed, at least in part, to enhanced integrity of the encapsulated mRNA. It is concluded that enhanced transfection can be achieved by optimizing formulation parameters to improve mRNA stability and that optimization of ionizable lipids to achieve enhanced potency may well lead to improvements in mRNA integrity through formation of the bleb structure rather than enhanced intracellular delivery.
- 28Whitehead, K. A.; Matthews, J.; Chang, P. H.; Niroui, F.; Dorkin, J. R.; Severgnini, M.; Anderson, D. G. In Vitro–In Vivo Translation of Lipid Nanoparticles for Hepatocellular siRNA Delivery. ACS Nano 2012, 6 (8), 6922– 6929, DOI: 10.1021/nn301922x28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XpvFegtL8%253D&md5=011ddae7baf862301e24ac829334f599In Vitro-In Vivo Translation of Lipid Nanoparticles for Hepatocellular siRNA DeliveryWhitehead, Kathryn A.; Matthews, Jonathan; Chang, Philip H.; Niroui, Farnaz; Dorkin, J. Robert; Severgnini, Mariano; Anderson, Daniel G.ACS Nano (2012), 6 (8), 6922-6929CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)A significant challenge in the development of clin. viable siRNA delivery systems is a lack of in vitro-in vivo translatability: many delivery vehicles that are initially promising in cell culture do not retain efficacy in animals. Despite its importance, little information exists on the predictive nature of in vitro methodologies, most likely due to the cost and time assocd. with generating in vitro-in vivo data sets. Recently, high-throughput techniques have been developed that have allowed the examn. of hundreds of lipid nanoparticle formulations for transfection efficiency in multiple exptl. systems. The large resulting data set has allowed the development of correlations between in vitro and characterization data and in vivo efficacy for hepatocellular delivery vehicles. Consistency of formulation technique and the type of cell used for in vitro expts. was found to significantly affect correlations, with primary hepatocytes and HeLa cells yielding the most predictive data. Interestingly, in vitro data acquired using HeLa cells were more predictive of in vivo performance than mouse hepatoma Hepa1-6 cells. Of the characterization parameters, only siRNA entrapment efficiency was partially predictive of in vivo silencing potential, while zeta-potential and, surprisingly, nanoparticle size (when <300 nm) as measured by dynamic light scattering were not. These data provide guiding principles in the development of clin. viable siRNA delivery materials and have the potential to reduce exptl. costs while improving the translation of materials into animals.
- 29Paunovska, K.; Sago, C. D.; Monaco, C. M.; Hudson, W. H.; Castro, M. G.; Rudoltz, T. G.; Kalathoor, S.; Vanover, D. A.; Santangelo, P. J.; Ahmed, R.; Bryksin, A. V.; Dahlman, J. E. A Direct Comparison of in Vitro and in Vivo Nucleic Acid Delivery Mediated by Hundreds of Nanoparticles Reveals a Weak Correlation. Nano Lett. 2018, 18 (3), 2148– 2157, DOI: 10.1021/acs.nanolett.8b0043229https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjs1SqtLo%253D&md5=a951cf961c11aa4fb49ef2de448780edA direct comparison of in vitro and in vivo nucleic acid delivery mediated by hundreds of nanoparticles reveals a weak correlationPaunovska, Kalina; Sago, Cory D.; Monaco, Christopher M.; Hudson, William H.; Castro, Marielena Gamboa; Rudoltz, Tobi G.; Kalathoor, Sujay; Vanover, Daryll A.; Santangelo, Philip J.; Ahmed, Rafi; Bryksin, Anton V.; Dahlman, James E.Nano Letters (2018), 18 (3), 2148-2157CODEN: NALEFD; ISSN:1530-6984. (American Chemical Society)Endothelial cells and macrophages play active roles in disease and as a result are important targets for nucleic acid therapies. While thousands of chem. distinct lipid nanoparticles (LNPs) can be synthesized to deliver nucleic acids, studying more than a few LNPs in vivo is challenging. As a result, it is difficult to understand how nanoparticles target these cells in vivo. Using high throughput LNP barcoding, we quantified how well LNPs delivered DNA barcodes to endothelial cells and macrophages in vitro, as well as endothelial cells and macrophages isolated from the lung, heart, and bone marrow in vivo. We focused on two fundamental questions in drug delivery. First, does in vitro LNP delivery predict in vivo LNP delivery. By comparing how 281 LNPs delivered barcodes to endothelial cells and macrophages in vitro and in vivo, we found in vitro delivery did not predict in vivo delivery. Second, does LNP delivery change within the microenvironment of a tissue. We quantified how 85 LNPs delivered barcodes to eight splenic cell populations, and found that cell types derived from myeloid progenitors tended to be targeted by similar LNPs, relative to cell types derived from lymphoid progenitors. These data demonstrate that barcoded LNPs can elucidate fundamental questions about in vivo nanoparticle delivery.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsami.4c20077.
Mathematical deduction of eq 1; photos of DiD-labeled LNPs before and after ultracentrifugation (Figure S1); RiboGreen assay results show encapsulation efficiency (Figure S2); LNP size distribution by cryoEM (Figure S3); other representative cryoEM images (Figure S4); cellular uptake and functional delivery of mRNA in Huh7 cells (Figure S5); quantification of CLSM images (Figure S6); and colocalization analysis shown as Pearson’s coefficient (Figure S7) (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.