High-Throughput Quantification and Characterization of Dual Payload mRNA/LNP Cargo via Deformulating Size Exclusion and Ion Pairing Reversed Phase AssaysClick to copy article linkArticle link copied!
- Mateusz Imiołek*Mateusz Imiołek*Email: [email protected]Waters Corporation, Rue Michel Servet 1 Geneva, 1211, SwitzerlandMore by Mateusz Imiołek
- Razvan CojocaruRazvan CojocaruAcuitas Therapeutics, 6190 Agronomy Rd. Suite 405, Vancouver, British Columbia V6T 1Z3, CanadaMore by Razvan Cojocaru
- Szabolcs FeketeSzabolcs FeketeWaters Corporation, Rue Michel Servet 1 Geneva, 1211, SwitzerlandMore by Szabolcs Fekete
- Jon Le HurayJon Le HurayAcuitas Therapeutics, 6190 Agronomy Rd. Suite 405, Vancouver, British Columbia V6T 1Z3, CanadaMore by Jon Le Huray
- Matthew LauberMatthew LauberWaters Corporation, 34 Maple St., Milford, Massachusetts 01757, United StatesMore by Matthew Lauber
Abstract
Therapeutic drugs and multivalent vaccines based on the delivery of mRNA via lipid nanoparticle (LNP) technologies are expected to dominate the biopharmaceutical industry landscape in the coming years. Many of these innovative therapies include several nucleic acid components (e.g., nuclease mRNA and guide RNA) posing unique analytical challenges when monitoring the quantity and quality of each individual payload substance in the formulated LNP. Current methods were optimized for single payload analysis and often lack resolving power needed to investigate nucleic acid mixtures. Ion pairing reversed phase (IP-RP) and size exclusion chromatography (SEC) are increasingly being used to characterize nucleic acids. Here, we studied their application for payload quantification in formulated LNP drug-like products. Using a detergent to disrupt the LNPs, the liberated payloads can be separated on an octadecyl RP column using a fast gradient. Reproducible results were obtained as lipids, and surfactants were efficiently eluted using a high organic solvent wash protocol. Alternatively, we also established an online SEC disruption analysis of the mRNA/LNPs wherein an alcohol and detergent containing a mobile phase was applied. Such conditions universally deformulated all tested LNP samples, indicating that a 5 min-long SEC separation can be used as a high-throughput platform method. In both approaches, the measurements facilitate a multiattribute analysis. Apart from quantitation, the characterization of specific impurities is achieved: IP-RP reveals mRNA-lipid adducts, while SEC informs on size variants, which in turn reduces a laboratory’s analytical workload. These easy-to-adopt LC-based assays are expected to fortify the analytical toolbox for emerging gene therapeutics.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*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 and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*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 and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*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
Material and Methods
Deformulating SEC Conditions
IP-RP Disruption Conditions
Results and Discussion
IP-RP Assay Method Development
Figure 1
Figure 1. IP-RP method development. Consecutive separations of unencapsulated FLuc mRNA interspersed by an empty LNP injection without (top) and with (bottom traces) an IPA column wash.
IP-RP Results and Discussion
Sample | Total [mg/mL] | RP assay [mg/mL] | gRNA [mg/mL] | mRNA [mg/mL] | Ratio gRNA/mRNA |
---|---|---|---|---|---|
sgRNA + FLuc mRNA(1:1) | |||||
LNP3 | 1.03 | 1.08 | 0.52 | 0.51 | 0.99 |
LNP4 | 1.03 | 1.07 | 0.52 | 0.51 | 0.98 |
LNP5 | 1.02 | 1.02 | 0.51 | 0.51 | 0.99 |
sgRNA + Cas9 mRNA (various ratios) | |||||
LNP3 (1:2) | 1.04 | 0.90 | 0.35 | 0.69 | 0.51 |
LNP3 (1:1) | 1.03 | 0.86 | 0.52 | 0.52 | 1.00 |
LNP3 (3:1) | 1.07 | 0.98 | 0.80 | 0.27 | 2.93 |
LNP3 (5:1) | 1.10 | 1.04 | 0.91 | 0.19 | 4.68 |
Figure 2
Figure 2. Use of a deformulating IP-RP assay for quantification of dual payload LNPs. A) Calibration curve for reference nucleic acids: sgRNA (green) and FLuc mRNA (fuchsia). B) Linear response of LNP3 loaded with the same RNAs across variable on column mass load. C) Overlay of IP-RP chromatograms showing response of LNP3 loaded with FLuc mRNA and sgRNA or formulated without any payload (empty - black trace). D) Overlay of IP-RP chromatograms for a fresh LNP3 sample (black) and a LNP3 sample stored at room temperature for 1 month (red) showing separation of lipid-adducted mRNA and loss of intact mRNA peak.
Sample | Total [mg/mL] | RP assay [mg/mL] | gRNA [mg/mL] | mRNA [mg/mL] | Ratio gRNA/mRNA |
---|---|---|---|---|---|
sgRNA + FLuc mRNA(1:1) | |||||
LNP3 | 1.08 | 1.08 | 0.54 | 0.54 | 1.01 |
LNP4 | 1.06 | 1.07 | 0.53 | 0.54 | 0.99 |
LNP5 | 1.02 | 1.02 | 0.50 | 0.52 | 0.96 |
sgRNA + Cas9 mRNA (various ratios) | |||||
LNP3 (1:2) | 0.96 | 0.90 | 0.34 | 0.62 | 0.55 |
LNP3 (1:1) | 0.91 | 0.86 | 0.47 | 0.44 | 1.05 |
LNP3 (3:1) | 0.97 | 0.98 | 0.73 | 0.24 | 3.04 |
LNP3 (5:1) | 0.99 | 1.04 | 0.83 | 0.16 | 5.06 |
SEC Assay Method Development
Figure 3
Figure 3. SEC chromatograms showing overlay of two UV signals recorded at 260 nm (continuous line) and 230 nm (dashed line) for A) intact LNP1 sample, B) its extracted mRNA payload, and C) LNP1 samples diluted with 0.2% Triton X-100 Surfactant. Separation was performed under native conditions with 100 ng of the injected sample.
Figure 4
Figure 4. Development of deformulating SEC analysis. A) Comparison of different deformulation conditions by analysis of 260 and 230 nm absorbance ratios (purity ratio). In all cases, the mobile phase based on 1× PBS was used with or without isopropanol (IPA) as the mobile phase additive on a water or detergent diluted sample. B) Comparison of deformulation efficiency between samples with different lipid formulations for Triton X-100 Surfactant diluted samples analyzed with IPA as a mobile phase additive. C) Purity ratios under optimized conditions for different intact LNP samples analyzed with 0.2% SDS and 20% IPA mobile phase.
SEC Results and Discussion
Figure 5
Figure 5. Use of the deformulating SEC assay for quantification of dual payload LNPs. A) Calibration curve for reference nucleic acids: sgRNA (green) and FLuc mRNA (fuchsia). B) Linear response of LNP3 loaded with the same RNAs across variable on column mass load. C) Overlay of SEC chromatograms showing response of LNP3 loaded with FLuc mRNA and sgRNA or formulated without any payload (empty - black trace). D) Quantification of LNP3 filled with a variable amount of Cas9 mRNA and sgRNA compared to the expected value used in formulation together with SEC chromatograms showing separation and relative amount of observed mRNA and sgRNA.
Conclusions
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.analchem.4c06296.
Additional figures, tables, experimental details and methods, including IP-RP gradient details (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.
Acknowledgments
ACQUITY, UPLC, GTxResolve, BEH and XBridge are trademarks of Waters Technologies Corporation. RiboGreen is a trademark of Molecular Probes, Inc. Triton is a trademark of Thermo Fisher Scientific Inc. Spikevax is a trademark of ModernaTx, Inc. Comirnaty is a trademark of BioNTech SE. All other trademarks are the property of their respective owner.
References
This article references 36 other publications.
- 1Parhiz, H.; Atochina-Vasserman, E. N.; Weissman, D. mRNA-based therapeutics: looking beyond COVID-19 vaccines. Lancet 2024, 403, 1192– 1204, DOI: 10.1016/S0140-6736(23)02444-3Google ScholarThere is no corresponding record for this reference.
- 2Kazemian, P.; Yu, S.-Y.; Thomson, S. B.; Birkenshaw, A.; Leavitt, B. R.; Ross, C. J. D. Lipid-Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing Components. Mol. Pharmaceutics 2022, 19, 1669– 1686, DOI: 10.1021/acs.molpharmaceut.1c00916Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtlalsbbJ&md5=5cc7ca317d5fb4f72fd0966cc9a10e41Lipid-Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing ComponentsKazemian, Pardis; Yu, Si-Yue; Thomson, Sarah B.; Birkenshaw, Alexandra; Leavitt, Blair R.; Ross, Colin J. D.Molecular Pharmaceutics (2022), 19 (6), 1669-1686CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)A review. Gene editing mediated by CRISPR/Cas9 systems is due to become a beneficial therapeutic option for treating genetic diseases and some cancers. However, there are challenges in delivering CRISPR components which necessitate sophisticated delivery systems for safe and effective genome editing. Lipid nanoparticles (LNPs) have become an attractive nonviral delivery platform for CRISPR-mediated genome editing due to their low immunogenicity and application flexibility. In this review, we provide a background of CRISPR-mediated gene therapy, as well as LNPs and their applicable characteristics for delivering CRISPR components. We then highlight the challenges of CRISPR delivery, which have driven the significant development of new, safe, and optimized LNP formulations in the past decade. Finally, we discuss considerations for using LNPs to deliver CRISPR and future perspectives on clin. translation of LNP-CRISPR gene editing.
- 3Bitounis, D.; Jacquinet, E.; Rogers, M. A.; Amiji, M. M. Strategies to reduce the risks of mRNA drug and vaccine toxicity. Nat. Rev. Drug Discovery 2024, 23, 281– 300, DOI: 10.1038/s41573-023-00859-3Google ScholarThere is no corresponding record for this reference.
- 4Wei, T.; Cheng, Q.; Min, Y.-L.; Olson, E. N.; Siegwart, D. J. Systemic nanoparticle delivery of CRISPR-Cas9 ribonucleoproteins for effective tissue specific genome editing. Nat. Commun. 2020, 11, 3232, DOI: 10.1038/s41467-020-17029-3Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlSitrvM&md5=8f53ca522c7d0be43edd8cde7912ff54Systemic nanoparticle delivery of CRISPR-Cas9 ribonucleoproteins for effective tissue specific genome editingWei, Tuo; Cheng, Qiang; Min, Yi-Li; Olson, Eric N.; Siegwart, Daniel J.Nature Communications (2020), 11 (1), 3232CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)CRISPR-Cas9 has emerged as a powerful technol. that relies on Cas9/sgRNA ribonucleoprotein complexes (RNPs) to target and edit DNA. However, many therapeutic targets cannot currently be accessed due to the lack of carriers that can deliver RNPs systemically. Here, we report a generalizable methodol. that allows engineering of modified lipid nanoparticles to efficiently deliver RNPs into cells and edit tissues including muscle, brain, liver, and lungs. I.v. injection facilitated tissue-specific, multiplexed editing of six genes in mouse lungs. High carrier potency was leveraged to create organ-specific cancer models in livers and lungs of mice though facile knockout of multiple genes. The developed carriers were also able to deliver RNPs to restore dystrophin expression in DMD mice and significantly decrease serum PCSK9 level in C57BL/6 mice. Application of this generalizable strategy will facilitate broad nanoparticle development for a variety of disease targets amenable to protein delivery and precise gene correction approaches.
- 5Yan, Q.; Trabulo, S.; Wolk, S.; Bridwell, H.; Burgers, P.; Thoryk, E.; Hamill, P.; Joest, E.; Ward, R.; Srinivasan, S.; . Defining the required critical quality attributes (CQAs) and phase requirements for mRNA/LNP product development and manufacture ; BioPhorum Operations Group Report 2023. See the following: https://cdnmedia.eurofins.com/corporate-eurofins/media/16371130/defining-the-required-cqas-and-phase-requirements-for-mrnalnp-product-development-and-manufacture-published.pdfGoogle ScholarThere is no corresponding record for this reference.
- 6Jia, X.; Liu, Y.; Wagner, A. M.; Chen, M.; Zhao, Y.; Smith, K. J.; Some, D.; Abend, A. M.; Pennington, J. Enabling online determination of the size-dependent RNA content of lipid nanoparticle-based RNA formulations. J. Chromatogr. B 2021, 1186, 123015 DOI: 10.1016/j.jchromb.2021.123015Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVanurrN&md5=6ff4ac31a4dfbab911ba0909d48c8737Enabling online determination of the size-dependent RNA content of lipid nanoparticle-based RNA formulationsJia, Xiujuan; Liu, Yong; Wagner, Angela M.; Chen, Michelle; Zhao, Yuejie; Smith, Katelyn J.; Some, Dan; Abend, Andreas M.; Pennington, JustinJournal of Chromatography B: Analytical Technologies in the Biomedical and Life Sciences (2021), 1186 (), 123015CODEN: JCBAAI; ISSN:1570-0232. (Elsevier B.V.)The potential of lipid nanoparticles (LNPs) as nucleic acid delivery vehicles has been demonstrated in recent years, culminating in the emergency use approval of LNP-based mRNA SARS-CoV-2 vaccines in late 2020. The detn. of RNA content relative to LNP size can be important to the understanding of efficacy and adverse effects. This work presents the first description of a facile and rapid anal. method for online, size-dependent RNA payload distribution measurement using data from multi-angle light scattering, UV and refractive index detectors following sepn. of the LNPs by size-exclusion chromatog. The anal. was validated by size-based fractionation of the LNPs with subsequent offline anal. of the fractions. Four LNPs formulated with different PEG-lipids and different lipid compns. were tested. Good agreement was obsd. between the online and offline size-based RNA distributions among all four LNPs, demonstrating the utility of the online method for LNP-encapsulated RNA in general, and suggesting a means for simplified biophys. quantitation of a dosing-related crit. quality attribute.
- 7Porterfield, J. Z.; Zlotnick, A. A simple and general method for determining the protein and nucleic acid content of viruses by UV absorbance. Virology 2010, 407, 281– 288, DOI: 10.1016/j.virol.2010.08.015Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXht1OmsrvK&md5=20e8fe486e645275404a624086b3f545A simple and general method for determining the protein and nucleic acid content of viruses by UV absorbancePorterfield, J. Zachary; Zlotnick, AdamVirology (2010), 407 (2), 281-288CODEN: VIRLAX; ISSN:0042-6822. (Elsevier B.V.)UV spectra of viruses are complicated by overlapping protein and RNA absorbance and light scattering. The authors describe and validate methodol. for estg. RNA and protein concn. from such spectra. Importantly, encapsidation did not substantially affect RNA absorbance. Combining absorbance data with a known T no., brome mosaic virus packages ∼3100 nucleotides/capsid, consistent with its genome. E. coli-expressed hepatitis B virus (HBV) packages host RNA based on capsid charge and vol. The authors examd. HBV capsid protein (Cp183, + 15 charge) and a less basic mutant (Cp183-EEE, + 12 charge) that mimics a phosphorylated state. Cp183-EEE packaged ∼ 3450 nucleotides per T = 4 capsid and Cp183 packaged ∼ 4800 nucleotides, correlating to the size of HBV's RNA pre-genome and mature DNA genome, resp. The RNA:protein charge ratio (∼1.4 phosphates per pos. charge) was consistent with that of other ssRNA viruses. This spectroscopic method is generalizable to any virus-like particle.
- 8Schober, G. B.; Story, S.; Arya, D. P. A careful look at lipid nanoparticle characterization: analysis of benchmark formulations for encapsulation of RNA cargo size gradient. Sci. Rep. 2024, 14, 2403, DOI: 10.1038/s41598-024-52685-1Google ScholarThere is no corresponding record for this reference.
- 9Jones, L. J.; Yue, S. T.; Cheung, C.-Y.; Singer, V. L. RNA Quantitation by Fluorescence-Based Solution Assay: RiboGreen Reagent Characterization. Anal. Biochem. 1998, 265, 368– 374, DOI: 10.1006/abio.1998.2914Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXitVymuw%253D%253D&md5=b4099f371234f82bf663fb1a37bbac00RNA quantitation by fluorescence-based solution assay: RiboGreen reagent characterizationJones, Laurie J.; Yue, Stephen T.; Cheung, Ching-Ying; Singer, Victoria L.Analytical Biochemistry (1998), 265 (2), 368-374CODEN: ANBCA2; ISSN:0003-2697. (Academic Press)We described the development of a sensitive fluorescence-based soln. assay for RNA using a new dye, RiboGreen RNA quantitation reagent. RiboGreen reagent exhibits > 1000-fold fluorescence enhancement and high quantum yield (0.65) upon binding nucleic acids, with excitation and emission maxima near those of fluorescein. Unbound dye is essentially nonfluorescent and has a large extinction coeff. (67,000 cm-1 M-1). The RiboGreen assay allows detection of as little as 1.0 ng/mL RNA in a std. fluorometer, filter fluorometer, or fluorescence microplate reader-surpassing the sensitivity achieved with ethidium bromide by 200-fold. The linear quantitation range for RiboGreen reagent extends over three orders of magnitude in RNA concn. Using 750 nM RiboGreen reagent, we quantitated 20 ng/mL to 1.0 μg/mL RNA. By dilg. the reagent to 75 nM, we could quantitate 1.0 to 50 ng/mL RNA. Both assay ranges exhibited linear fluorescence increases vs. RNA concn. (r2 = 0.999). Assay linearity was maintained in the presence of salts, protein, urea, ethanol, chloroform, agarose, and some detergents. Several different RNA types yielded similar signal intensities and detection sensitivities. The assay is easy to use, rapid, and readily adaptable for automation. (c) 1998 Academic Press.
- 10Malburet, C.; Carboni, A.; Guinamand, S.; Naik, H.; Fertier-Prizzon, S. mRNA extraction from lipid nanoparticles. J. Chromatogr. A 2024, 1714, 464545 DOI: 10.1016/j.chroma.2023.464545Google ScholarThere is no corresponding record for this reference.
- 11Gao, R. Y.; Riley, C. M.; Toth, E.; Blair, R. H.; Gerold, M. N.; McCormick, C.; Taylor, A. W.; Hu, T.; Rowlen, K. L.; Dawson, E. D. Rapid Identity and Quantity CQA Test for Multivalent mRNA Drug Product Formulations. Vaccines 2022, 10, 1704, DOI: 10.3390/vaccines10101704Google ScholarThere is no corresponding record for this reference.
- 12Pulido, M. A.; Gutierrez, M.; Wang, F.; Borgos, S. E.; Parot, J. mRNA-LNP nucleic acid assessment from distinct formulations by multi-capillary gel electrophoresis ; Sciex Application Note 2022. DOI: https://sciex.com/tech-notes/biopharma/mrna-lnp-nucleic-acid-assessment-from-distinct-formulations-by-m.Google ScholarThere is no corresponding record for this reference.
- 13Fekete, S.; Aebischer, M. K.; Imiołek, M.; Graf, T.; Ruppert, R.; Lauber, M.; D’Atri, V.; Guillarme, D. Chromatographic strategies for the analytical characterization of adeno-associated virus vector-based gene therapy products. TrAC, Trends Anal. Chem. 2023, 164, 117088 DOI: 10.1016/j.trac.2023.117088Google ScholarThere is no corresponding record for this reference.
- 14Fekete, S.; Doneanu, C.; Addepalli, B.; Gaye, M.; Nguyen, J.; Alden, B.; Birdsall, R.; Han, D.; Isaac, G.; Lauber, M. Challenges and emerging trends in liquid chromatography-based analyses of mRNA pharmaceuticals. J. Pharm. Biomed. Anal. 2023, 224, 115174 DOI: 10.1016/j.jpba.2022.115174Google ScholarThere is no corresponding record for this reference.
- 15Kanavarioti, A. HPLC methods for purity evaluation of man-made single-stranded RNAs. Sci. Rep. 2019, 9, 1019, DOI: 10.1038/s41598-018-37642-zGoogle Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cjns1Chuw%253D%253D&md5=fd7e78e9b5b3a11acf31d1bebdc77bb4HPLC methods for purity evaluation of man-made single-stranded RNAsKanavarioti AnastassiaScientific reports (2019), 9 (1), 1019 ISSN:.Synthetic RNA oligos exhibit purity decreasing as a function of length, because the efficiency of the total synthesis is the numerical product of the individual step efficiencies, typically below 98%. Analytical methods for RNAs up to the 60 nucleotides (nt) have been reported, but they fall short for purity evaluation of 100nt long, used as single guide RNA (sgRNA) in CRISPR technology, and promoted as pharmaceuticals. In an attempt to exploit a single HPLC method and obtain both identity as well as purity, ion-pair reversed-phase chromatography (IP-RP) at high temperature in the presence of an organic cosolvent is the current analytical strategy. Here we report that IP-RP is less suitable compared to the conventional ion-exchange (IEX) for analysis of 100nt RNAs. We demonstrate the relative stability of RNA in the denaturing/basic IEX mobile phase, lay out a protocol to determine the on-the-column stability of any RNA, and establish the applicability of this method for quality testing of sgRNA, tRNA, and mRNA. Unless well resolving HPLC methods are used for batch-to-batch evaluation of man-made RNAs, process development will remain shortsighted, and observed off-target effects in-vitro or in-vivo may be partially related to low purity and the presence of shorter sequences.
- 16Camperi, J.; Lippold, S.; Ayalew, L.; Roper, B.; Shao, S.; Freund, E.; Nissenbaum, A.; Galan, C.; Cao, Q.; Yang, F. Comprehensive Impurity Profiling of mRNA: Evaluating Current Technologies and Advanced Analytical Techniques. Anal. Chem. 2024, 96, 3886– 3897, DOI: 10.1021/acs.analchem.3c05539Google ScholarThere is no corresponding record for this reference.
- 17Welbourne, E. N.; Loveday, K. A.; Nair, A.; Nourafkan, E.; Qu, J.; Cook, K.; Kis, Z.; Dickman, M. J. Anion exchange HPLC monitoring of mRNA in vitro transcription reactions to support mRNA manufacturing process development. Front. Mol. Biosci. 2024, 11, 1250833, DOI: 10.3389/fmolb.2024.1250833Google ScholarThere is no corresponding record for this reference.
- 18Gilar, M.; Doneanu, C.; Gaye, M. M. Liquid Chromatography Methods for Analysis of mRNA Poly(A) Tail Length and Heterogeneity. Anal. Chem. 2023, 95, 14308– 14316, DOI: 10.1021/acs.analchem.3c02552Google ScholarThere is no corresponding record for this reference.
- 19D’Atri, V.; Imiołek, M.; Quinn, C.; Finny, A.; Lauber, M.; Fekete, S.; Guillarme, D. Size exclusion chromatography of biopharmaceutical products: From current practices for proteins to emerging trends for viral vectors, nucleic acids and lipid nanoparticles. J. Chromatogr. A 2024, 1722, 464862 DOI: 10.1016/j.chroma.2024.464862Google ScholarThere is no corresponding record for this reference.
- 20Lokras, A.; Chakravarty, A.; Rades, T.; Christensen, D.; Franzyk, H.; Thakur, A.; Foged, C. Simultaneous quantification of multiple RNA cargos co-loaded into nanoparticle-based delivery systems. Int. J. Pharm. 2022, 626, 122171 DOI: 10.1016/j.ijpharm.2022.122171Google ScholarThere is no corresponding record for this reference.
- 21Zhang, J.; Haas, R. M.; Leone, A. M. Polydispersity Characterization of Lipid Nanoparticles for siRNA Delivery Using Multiple Detection Size-Exclusion Chromatography. Anal. Chem. 2012, 84, 6088– 6096, DOI: 10.1021/ac3007768Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xot1Olt7o%253D&md5=be22cbff7a62f6d84c570a38f9bd1572Polydispersity Characterization of Lipid Nanoparticles for siRNA Delivery Using Multiple Detection Size-Exclusion ChromatographyZhang, Jingtao; Haas, R. Matthew; Leone, Anthony M.Analytical Chemistry (Washington, DC, United States) (2012), 84 (14), 6088-6096CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)The development of lipid nanoparticle (LNP) based small interfering RNA (siRNA) therapeutics presents unique pharmaceutical and regulatory challenges. In contrast to small mol. drugs that are highly pure and well-defined, LNP drug products can exhibit heterogeneity in size, compn., surface property, or morphol. The potential for batch heterogeneity introduces a complexity that must be confronted in order to successfully develop and ensure quality in LNP pharmaceuticals. Currently, there is a lack of scientific knowledge in the heterogeneity of LNPs as well as high-resoln. techniques that permit this evaluation. This article reports a size-exclusion chromatog. (SEC) method that permits the high-resoln. anal. of LNP size distribution in its native soln. condition. When coupled with multiple detection systems including UV-vis, multi-angle light scattering, and refractive index, online characterization of the distributions in size, mol. wt., and siRNA cargo loading of LNPs could be achieved. Six LNPs with sizes in the rang of 60-140 nm were evaluated and it was found that the SEC sepn. is efficient, highly reproducible, and can be broadly applied to a diverse range of LNPs. A comparison between the current SEC method and asym. field flow fractionation (FFF) shows that the current method provides similar size distribution results on LNPs compared to FFF. Two representative LNPs with similar bulk properties were evaluated in-depth using the SEC method along with two other sizing techniques-dynamic light scattering and cryo-TEM. Profound differences in batch polydispersity were obsd. between them. Despite the similarity in the particle assembly process, it was found that one LNP (A) possessed a narrow size and mol. wt. distribution while the other (B) was polydisperse. The present results suggest that LNP drug products are highly complex and diverse in nature, and care should be taken in examg. and understanding them to ensure quality and consistency. The method developed here can not only serve as a method for understanding LNP product property, permitting control on product quality, but also could serve as a potential manufg. method for product purifn. Understandings obtained in this work can help to facilitate the development of LNPs as a well-defined pharmaceutical product.
- 22D’Atri, V.; Lardeux, H.; Goyon, A.; Imiołek, M.; Fekete, S.; Lauber, M.; Zhang, K.; Guillarme, D. Optimizing Messenger RNA Analysis Using Ultra-Wide Pore Size Exclusion Chromatography Columns. Int. J. Mol. Sci. 2024, 25, 6254, DOI: 10.3390/ijms25116254Google ScholarThere is no corresponding record for this reference.
- 23Packer, M.; Gyawali, D.; Yerabolu, R.; Schariter, J.; White, P. A novel mechanism for the loss of mRNA activity in lipid nanoparticle delivery systems. Nat. Commun. 2021, 12, 6777, DOI: 10.1038/s41467-021-26926-0Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisFClsLzL&md5=9ea94bb1f8267764266f7419475c1255A novel mechanism for the loss of mRNA activity in lipid nanoparticle delivery systemsPacker, Meredith; Gyawali, Dipendra; Yerabolu, Ravikiran; Schariter, Joseph; White, PhilNature Communications (2021), 12 (1), 6777CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Lipid nanoparticle (LNP)-formulated mRNA vaccines were rapidly developed and deployed in response to the SARS-CoV-2 pandemic. Due to the labile nature of mRNA, identifying impurities that could affect product stability and efficacy is crucial to the long-term use of nucleic-acid based medicines. Herein, reversed-phase ion pair high performance liq. chromatog. (RP-IP HPLC) was used to identify a class of impurity formed through lipid:mRNA reactions; such reactions are typically undetectable by traditional mRNA purity anal. techniques. The identified modifications render the mRNA untranslatable, leading to loss of protein expression. Specifically, electrophilic impurities derived from the ionizable cationic lipid component are shown to be responsible. Mechanisms implicated in the formation of reactive species include oxidn. and subsequent hydrolysis of the tertiary amine. It thus remains crit. to ensure robust anal. methods and stringent manufg. control to ensure mRNA stability and high activity in LNP delivery systems.
- 24Levin, D. S.; Shepperd, B. T.; Gruenloh, C. J. Combining ion pairing agents for enhanced analysis of oligonucleotide therapeutics by reversed phase-ion pairing ultra performance liquid chromatography (UPLC). J. Chromatogr. B 2011, 879, 1587– 1595, DOI: 10.1016/j.jchromb.2011.03.051Google ScholarThere is no corresponding record for this reference.
- 25Hashiba, K.; Taguchi, M.; Sakamoto, S.; Otsu, A.; Maeda, Y.; Ebe, H.; Okazaki, A.; Harashima, H.; Sato, Y. Overcoming thermostability challenges in mRNA–lipid nanoparticle systems with piperidine-based ionizable lipids. Commun. Biol. 2024, 7, 556, DOI: 10.1038/s42003-024-06235-0Google ScholarThere is no corresponding record for this reference.
- 26Birdsall, R. E.; Han, D.; DeLaney, K.; Kowalczyk, A.; Cojocaru, R.; Lauber, M.; Huray, J. L. Monitoring stability indicating impurities and aldehyde content in lipid nanoparticle raw material and formulated drugs. J. Chromatogr. B 2024, 1234, 124005 DOI: 10.1016/j.jchromb.2024.124005Google ScholarThere is no corresponding record for this reference.
- 27De Vos, J.; Morreel, K.; Alvarez, P.; Vanluchene, H.; Vankeirsbilck, R.; Sandra, P.; Sandra, K. Evaluation of size-exclusion chromatography, multi-angle light scattering detection and mass photometry for the characterization of mRNA. J. Chromatogr. A 2024, 1719, 464756 DOI: 10.1016/j.chroma.2024.464756Google ScholarThere is no corresponding record for this reference.
- 28Imiołek, M.; Fekete, S.; Kizekai, L.; Addepalli, B.; Lauber, M. Fast and efficient size exclusion chromatography of adeno associated viral vectors with 2.5 micrometer particle low adsorption columns. J. Chromatogr. A 2024, 1714, 464587 DOI: 10.1016/j.chroma.2023.464587Google ScholarThere is no corresponding record for this reference.
- 29Fekete, S.; DeLano, M.; Harrison, A. B.; Shiner, S. J.; Belanger, J. L.; Wyndham, K. D.; Lauber, M. A. Size Exclusion and Ion Exchange Chromatographic Hardware Modified with a Hydrophilic Hybrid Surface. Anal. Chem. 2022, 94, 3360– 3367, DOI: 10.1021/acs.analchem.1c05466Google ScholarThere is no corresponding record for this reference.
- 30Kizekai, L.; Addepalli, B.; Jawdat, N.; Chumakov, V.; Gilar, M.; Lauber, M. A. Suitability of GTxResolve Premier BEH SEC 450 Å 2.5 μm Column for Size-based Separations of Nucleic Acids ; Waters Application Note 2023. See the following: https://www.waters.com/nextgen/us/en/library/application-notes/2023/suitability-of-xbridge-premier-gtx-beh-sec-450-25-m-column-for-size-based-separations-of-nucleic-acids.html.Google ScholarThere is no corresponding record for this reference.
- 31Fongaro, B.; Campara, B.; Moscatiello, G. Y.; De Luigi, A.; Panzeri, D.; Sironi, L.; Bigini, P.; Carretta, G.; Miolo, G.; Pasut, G. Assessing the physicochemical stability and intracellular trafficking of mRNA-based COVID-19 vaccines. Int. J. Pharm. 2023, 644, 123319 DOI: 10.1016/j.ijpharm.2023.123319Google ScholarThere is no corresponding record for this reference.
- 32Koetsier, G.; Cantor, E. A Practical Guide to Analyzing Nucleic Acid Concentration and Purity with Microvolume Spectrophotometers ; New England Biolabs Technical Note 2019. See the following: https://www.neb.com/en/-/media/nebus/files/application-notes/technote_mvs_analysis_of_nucleic_acid_concentration_and_purity.pdf?rev=c24cea043416420d84fb6bf7b554dbbb.Google ScholarThere is no corresponding record for this reference.
- 33Ramirez-Cuevas, F. V.; Gurunatha, K. L.; Parkin, I. P.; Papakonstantinou, I. Universal Theory of Light Scattering of Randomly Oriented Particles: A Fluctuational-Electrodynamics Approach for Light Transport Modeling in Disordered Nanostructures. ACS Photonics 2022, 9, 672– 681, DOI: 10.1021/acsphotonics.1c01710Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XislersL4%253D&md5=85ddef06713dee1fc3f47aa718ff11d2Universal Theory of Light Scattering of Randomly Oriented Particles: A Fluctuational-Electrodynamics Approach for Light Transport Modeling in Disordered NanostructuresRamirez-Cuevas, Francisco V.; Gurunatha, Kargal L.; Parkin, Ivan P.; Papakonstantinou, IoannnisACS Photonics (2022), 9 (2), 672-681CODEN: APCHD5; ISSN:2330-4022. (American Chemical Society)Disordered nanostructures are commonly encountered in many nanophotonic systems, from colloid dispersions for sensing to heterostructured photocatalysts. Randomness, however, imposes severe challenges for nanophotonics modeling, often constrained by the irregular geometry of the scatterers involved or the stochastic nature of the problem itself. In this Article, we resolve this conundrum by presenting a universal theory of averaged light scattering of randomly oriented objects. Specifically, we derive expansion-basis-independent formulas of the orientation-and-polarization-averaged absorption cross section, scattering cross section, and asymmetry parameter, for single or a collection of objects of arbitrary shape. These three parameters can be directly integrated into traditional unpolarized radiative energy transfer modeling, enabling a practical tool to predict multiple scattering and light transport in disordered nanostructured materials. Notably, the formulas of av. light scattering can be derived under the principles of fluctuational electrodynamics, allowing the analogous math. treatment to the methods used in thermal radiation, nonequil. electromagnetic forces, and other assocd. phenomena. The proposed modeling framework is validated against optical measurements of polymer composite films with metal-oxide microcrystals. Our work may contribute to a better understanding of light-matter interactions in disordered systems, such as plasmonics for sensing and photothermal therapy, photocatalysts for water splitting and CO2 dissocn., photonic glasses for artificial structural colors, and diffuse reflectors for radiative cooling, to name just a few.
- 34Goyon, A.; Tang, S.; Fekete, S.; Nguyen, D.; Hofmann, K.; Wang, S.; Shatz-Binder, W.; Fernandez, K. I.; Hecht, E. S.; Lauber, M. Separation of Plasmid DNA Topological Forms, Messenger RNA, and Lipid Nanoparticle Aggregates Using an Ultrawide Pore Size Exclusion Chromatography Column. Anal. Chem. 2023, 95, 15017– 15024, DOI: 10.1021/acs.analchem.3c02944Google ScholarThere is no corresponding record for this reference.
- 35Muramatsu, H.; Lam, K.; Bajusz, C.; Laczkó, D.; Karikó, K.; Schreiner, P.; Martin, A.; Lutwyche, P.; Heyes, J.; Pardi, N. Lyophilization provides long-term stability for a lipid nanoparticle-formulated, nucleoside-modified mRNA vaccine. Mol. Ther. 2022, 30, 1941– 1951, DOI: 10.1016/j.ymthe.2022.02.001Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XjslSgsrs%253D&md5=65b435c164ca92084a2ca06ea701952bLyophilization provides long-term stability for a lipid nanoparticle-formulated, nucleoside-modified mRNA vaccineMuramatsu, Hiromi; Lam, Kieu; Bajusz, Csaba; Laczko, Dorottya; Kariko, Katalin; Schreiner, Petra; Martin, Alan; Lutwyche, Peter; Heyes, James; Pardi, NorbertMolecular Therapy (2022), 30 (5), 1941-1951CODEN: MTOHCK; ISSN:1525-0024. (Cell Press)Lipid nanoparticle (LNP)-formulated nucleoside-modified mRNA vaccines have proven to be very successful in the fight against the coronavirus disease 2019 (COVID-19) pandemic. They are effective, safe, and can be produced in large quantities. However, the long-term storage of mRNA-LNP vaccines without freezing is still a challenge. Here, we demonstrate that nucleoside-modified mRNA-LNPs can be lyophilized, and the physicochem. properties of the lyophilized material do not significantly change for 12 wk after storage at room temp. and for at least 24 wk after storage at 4°C. Importantly, we show in comparative mouse studies that lyophilized firefly luciferase-encoding mRNA-LNPs maintain their high expression, and no decrease in the immunogenicity of a lyophilized influenza virus hemagglutinin-encoding mRNA-LNP vaccine was obsd. after 12 wk of storage at room temp. or for at least 24 wk after storage at 4°C. Our studies offer a potential soln. to overcome the long-term storage-related limitations of nucleoside-modified mRNA-LNP vaccines.
- 36Camperi, J.; Moshref, M.; Dai, L.; Lee, H. Y. Physicochemical and Functional Characterization of Differential CRISPR-Cas9 Ribonucleoprotein Complexes. Anal. Chem. 2022, 94, 1432– 1440, DOI: 10.1021/acs.analchem.1c04795Google ScholarThere is no corresponding record for this reference.
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. IP-RP method development. Consecutive separations of unencapsulated FLuc mRNA interspersed by an empty LNP injection without (top) and with (bottom traces) an IPA column wash.
Figure 2
Figure 2. Use of a deformulating IP-RP assay for quantification of dual payload LNPs. A) Calibration curve for reference nucleic acids: sgRNA (green) and FLuc mRNA (fuchsia). B) Linear response of LNP3 loaded with the same RNAs across variable on column mass load. C) Overlay of IP-RP chromatograms showing response of LNP3 loaded with FLuc mRNA and sgRNA or formulated without any payload (empty - black trace). D) Overlay of IP-RP chromatograms for a fresh LNP3 sample (black) and a LNP3 sample stored at room temperature for 1 month (red) showing separation of lipid-adducted mRNA and loss of intact mRNA peak.
Figure 3
Figure 3. SEC chromatograms showing overlay of two UV signals recorded at 260 nm (continuous line) and 230 nm (dashed line) for A) intact LNP1 sample, B) its extracted mRNA payload, and C) LNP1 samples diluted with 0.2% Triton X-100 Surfactant. Separation was performed under native conditions with 100 ng of the injected sample.
Figure 4
Figure 4. Development of deformulating SEC analysis. A) Comparison of different deformulation conditions by analysis of 260 and 230 nm absorbance ratios (purity ratio). In all cases, the mobile phase based on 1× PBS was used with or without isopropanol (IPA) as the mobile phase additive on a water or detergent diluted sample. B) Comparison of deformulation efficiency between samples with different lipid formulations for Triton X-100 Surfactant diluted samples analyzed with IPA as a mobile phase additive. C) Purity ratios under optimized conditions for different intact LNP samples analyzed with 0.2% SDS and 20% IPA mobile phase.
Figure 5
Figure 5. Use of the deformulating SEC assay for quantification of dual payload LNPs. A) Calibration curve for reference nucleic acids: sgRNA (green) and FLuc mRNA (fuchsia). B) Linear response of LNP3 loaded with the same RNAs across variable on column mass load. C) Overlay of SEC chromatograms showing response of LNP3 loaded with FLuc mRNA and sgRNA or formulated without any payload (empty - black trace). D) Quantification of LNP3 filled with a variable amount of Cas9 mRNA and sgRNA compared to the expected value used in formulation together with SEC chromatograms showing separation and relative amount of observed mRNA and sgRNA.
References
This article references 36 other publications.
- 1Parhiz, H.; Atochina-Vasserman, E. N.; Weissman, D. mRNA-based therapeutics: looking beyond COVID-19 vaccines. Lancet 2024, 403, 1192– 1204, DOI: 10.1016/S0140-6736(23)02444-3There is no corresponding record for this reference.
- 2Kazemian, P.; Yu, S.-Y.; Thomson, S. B.; Birkenshaw, A.; Leavitt, B. R.; Ross, C. J. D. Lipid-Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing Components. Mol. Pharmaceutics 2022, 19, 1669– 1686, DOI: 10.1021/acs.molpharmaceut.1c009162https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtlalsbbJ&md5=5cc7ca317d5fb4f72fd0966cc9a10e41Lipid-Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing ComponentsKazemian, Pardis; Yu, Si-Yue; Thomson, Sarah B.; Birkenshaw, Alexandra; Leavitt, Blair R.; Ross, Colin J. D.Molecular Pharmaceutics (2022), 19 (6), 1669-1686CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)A review. Gene editing mediated by CRISPR/Cas9 systems is due to become a beneficial therapeutic option for treating genetic diseases and some cancers. However, there are challenges in delivering CRISPR components which necessitate sophisticated delivery systems for safe and effective genome editing. Lipid nanoparticles (LNPs) have become an attractive nonviral delivery platform for CRISPR-mediated genome editing due to their low immunogenicity and application flexibility. In this review, we provide a background of CRISPR-mediated gene therapy, as well as LNPs and their applicable characteristics for delivering CRISPR components. We then highlight the challenges of CRISPR delivery, which have driven the significant development of new, safe, and optimized LNP formulations in the past decade. Finally, we discuss considerations for using LNPs to deliver CRISPR and future perspectives on clin. translation of LNP-CRISPR gene editing.
- 3Bitounis, D.; Jacquinet, E.; Rogers, M. A.; Amiji, M. M. Strategies to reduce the risks of mRNA drug and vaccine toxicity. Nat. Rev. Drug Discovery 2024, 23, 281– 300, DOI: 10.1038/s41573-023-00859-3There is no corresponding record for this reference.
- 4Wei, T.; Cheng, Q.; Min, Y.-L.; Olson, E. N.; Siegwart, D. J. Systemic nanoparticle delivery of CRISPR-Cas9 ribonucleoproteins for effective tissue specific genome editing. Nat. Commun. 2020, 11, 3232, DOI: 10.1038/s41467-020-17029-34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlSitrvM&md5=8f53ca522c7d0be43edd8cde7912ff54Systemic nanoparticle delivery of CRISPR-Cas9 ribonucleoproteins for effective tissue specific genome editingWei, Tuo; Cheng, Qiang; Min, Yi-Li; Olson, Eric N.; Siegwart, Daniel J.Nature Communications (2020), 11 (1), 3232CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)CRISPR-Cas9 has emerged as a powerful technol. that relies on Cas9/sgRNA ribonucleoprotein complexes (RNPs) to target and edit DNA. However, many therapeutic targets cannot currently be accessed due to the lack of carriers that can deliver RNPs systemically. Here, we report a generalizable methodol. that allows engineering of modified lipid nanoparticles to efficiently deliver RNPs into cells and edit tissues including muscle, brain, liver, and lungs. I.v. injection facilitated tissue-specific, multiplexed editing of six genes in mouse lungs. High carrier potency was leveraged to create organ-specific cancer models in livers and lungs of mice though facile knockout of multiple genes. The developed carriers were also able to deliver RNPs to restore dystrophin expression in DMD mice and significantly decrease serum PCSK9 level in C57BL/6 mice. Application of this generalizable strategy will facilitate broad nanoparticle development for a variety of disease targets amenable to protein delivery and precise gene correction approaches.
- 5Yan, Q.; Trabulo, S.; Wolk, S.; Bridwell, H.; Burgers, P.; Thoryk, E.; Hamill, P.; Joest, E.; Ward, R.; Srinivasan, S.; . Defining the required critical quality attributes (CQAs) and phase requirements for mRNA/LNP product development and manufacture ; BioPhorum Operations Group Report 2023. See the following: https://cdnmedia.eurofins.com/corporate-eurofins/media/16371130/defining-the-required-cqas-and-phase-requirements-for-mrnalnp-product-development-and-manufacture-published.pdfThere is no corresponding record for this reference.
- 6Jia, X.; Liu, Y.; Wagner, A. M.; Chen, M.; Zhao, Y.; Smith, K. J.; Some, D.; Abend, A. M.; Pennington, J. Enabling online determination of the size-dependent RNA content of lipid nanoparticle-based RNA formulations. J. Chromatogr. B 2021, 1186, 123015 DOI: 10.1016/j.jchromb.2021.1230156https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVanurrN&md5=6ff4ac31a4dfbab911ba0909d48c8737Enabling online determination of the size-dependent RNA content of lipid nanoparticle-based RNA formulationsJia, Xiujuan; Liu, Yong; Wagner, Angela M.; Chen, Michelle; Zhao, Yuejie; Smith, Katelyn J.; Some, Dan; Abend, Andreas M.; Pennington, JustinJournal of Chromatography B: Analytical Technologies in the Biomedical and Life Sciences (2021), 1186 (), 123015CODEN: JCBAAI; ISSN:1570-0232. (Elsevier B.V.)The potential of lipid nanoparticles (LNPs) as nucleic acid delivery vehicles has been demonstrated in recent years, culminating in the emergency use approval of LNP-based mRNA SARS-CoV-2 vaccines in late 2020. The detn. of RNA content relative to LNP size can be important to the understanding of efficacy and adverse effects. This work presents the first description of a facile and rapid anal. method for online, size-dependent RNA payload distribution measurement using data from multi-angle light scattering, UV and refractive index detectors following sepn. of the LNPs by size-exclusion chromatog. The anal. was validated by size-based fractionation of the LNPs with subsequent offline anal. of the fractions. Four LNPs formulated with different PEG-lipids and different lipid compns. were tested. Good agreement was obsd. between the online and offline size-based RNA distributions among all four LNPs, demonstrating the utility of the online method for LNP-encapsulated RNA in general, and suggesting a means for simplified biophys. quantitation of a dosing-related crit. quality attribute.
- 7Porterfield, J. Z.; Zlotnick, A. A simple and general method for determining the protein and nucleic acid content of viruses by UV absorbance. Virology 2010, 407, 281– 288, DOI: 10.1016/j.virol.2010.08.0157https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXht1OmsrvK&md5=20e8fe486e645275404a624086b3f545A simple and general method for determining the protein and nucleic acid content of viruses by UV absorbancePorterfield, J. Zachary; Zlotnick, AdamVirology (2010), 407 (2), 281-288CODEN: VIRLAX; ISSN:0042-6822. (Elsevier B.V.)UV spectra of viruses are complicated by overlapping protein and RNA absorbance and light scattering. The authors describe and validate methodol. for estg. RNA and protein concn. from such spectra. Importantly, encapsidation did not substantially affect RNA absorbance. Combining absorbance data with a known T no., brome mosaic virus packages ∼3100 nucleotides/capsid, consistent with its genome. E. coli-expressed hepatitis B virus (HBV) packages host RNA based on capsid charge and vol. The authors examd. HBV capsid protein (Cp183, + 15 charge) and a less basic mutant (Cp183-EEE, + 12 charge) that mimics a phosphorylated state. Cp183-EEE packaged ∼ 3450 nucleotides per T = 4 capsid and Cp183 packaged ∼ 4800 nucleotides, correlating to the size of HBV's RNA pre-genome and mature DNA genome, resp. The RNA:protein charge ratio (∼1.4 phosphates per pos. charge) was consistent with that of other ssRNA viruses. This spectroscopic method is generalizable to any virus-like particle.
- 8Schober, G. B.; Story, S.; Arya, D. P. A careful look at lipid nanoparticle characterization: analysis of benchmark formulations for encapsulation of RNA cargo size gradient. Sci. Rep. 2024, 14, 2403, DOI: 10.1038/s41598-024-52685-1There is no corresponding record for this reference.
- 9Jones, L. J.; Yue, S. T.; Cheung, C.-Y.; Singer, V. L. RNA Quantitation by Fluorescence-Based Solution Assay: RiboGreen Reagent Characterization. Anal. Biochem. 1998, 265, 368– 374, DOI: 10.1006/abio.1998.29149https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXitVymuw%253D%253D&md5=b4099f371234f82bf663fb1a37bbac00RNA quantitation by fluorescence-based solution assay: RiboGreen reagent characterizationJones, Laurie J.; Yue, Stephen T.; Cheung, Ching-Ying; Singer, Victoria L.Analytical Biochemistry (1998), 265 (2), 368-374CODEN: ANBCA2; ISSN:0003-2697. (Academic Press)We described the development of a sensitive fluorescence-based soln. assay for RNA using a new dye, RiboGreen RNA quantitation reagent. RiboGreen reagent exhibits > 1000-fold fluorescence enhancement and high quantum yield (0.65) upon binding nucleic acids, with excitation and emission maxima near those of fluorescein. Unbound dye is essentially nonfluorescent and has a large extinction coeff. (67,000 cm-1 M-1). The RiboGreen assay allows detection of as little as 1.0 ng/mL RNA in a std. fluorometer, filter fluorometer, or fluorescence microplate reader-surpassing the sensitivity achieved with ethidium bromide by 200-fold. The linear quantitation range for RiboGreen reagent extends over three orders of magnitude in RNA concn. Using 750 nM RiboGreen reagent, we quantitated 20 ng/mL to 1.0 μg/mL RNA. By dilg. the reagent to 75 nM, we could quantitate 1.0 to 50 ng/mL RNA. Both assay ranges exhibited linear fluorescence increases vs. RNA concn. (r2 = 0.999). Assay linearity was maintained in the presence of salts, protein, urea, ethanol, chloroform, agarose, and some detergents. Several different RNA types yielded similar signal intensities and detection sensitivities. The assay is easy to use, rapid, and readily adaptable for automation. (c) 1998 Academic Press.
- 10Malburet, C.; Carboni, A.; Guinamand, S.; Naik, H.; Fertier-Prizzon, S. mRNA extraction from lipid nanoparticles. J. Chromatogr. A 2024, 1714, 464545 DOI: 10.1016/j.chroma.2023.464545There is no corresponding record for this reference.
- 11Gao, R. Y.; Riley, C. M.; Toth, E.; Blair, R. H.; Gerold, M. N.; McCormick, C.; Taylor, A. W.; Hu, T.; Rowlen, K. L.; Dawson, E. D. Rapid Identity and Quantity CQA Test for Multivalent mRNA Drug Product Formulations. Vaccines 2022, 10, 1704, DOI: 10.3390/vaccines10101704There is no corresponding record for this reference.
- 12Pulido, M. A.; Gutierrez, M.; Wang, F.; Borgos, S. E.; Parot, J. mRNA-LNP nucleic acid assessment from distinct formulations by multi-capillary gel electrophoresis ; Sciex Application Note 2022. DOI: https://sciex.com/tech-notes/biopharma/mrna-lnp-nucleic-acid-assessment-from-distinct-formulations-by-m.There is no corresponding record for this reference.
- 13Fekete, S.; Aebischer, M. K.; Imiołek, M.; Graf, T.; Ruppert, R.; Lauber, M.; D’Atri, V.; Guillarme, D. Chromatographic strategies for the analytical characterization of adeno-associated virus vector-based gene therapy products. TrAC, Trends Anal. Chem. 2023, 164, 117088 DOI: 10.1016/j.trac.2023.117088There is no corresponding record for this reference.
- 14Fekete, S.; Doneanu, C.; Addepalli, B.; Gaye, M.; Nguyen, J.; Alden, B.; Birdsall, R.; Han, D.; Isaac, G.; Lauber, M. Challenges and emerging trends in liquid chromatography-based analyses of mRNA pharmaceuticals. J. Pharm. Biomed. Anal. 2023, 224, 115174 DOI: 10.1016/j.jpba.2022.115174There is no corresponding record for this reference.
- 15Kanavarioti, A. HPLC methods for purity evaluation of man-made single-stranded RNAs. Sci. Rep. 2019, 9, 1019, DOI: 10.1038/s41598-018-37642-z15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cjns1Chuw%253D%253D&md5=fd7e78e9b5b3a11acf31d1bebdc77bb4HPLC methods for purity evaluation of man-made single-stranded RNAsKanavarioti AnastassiaScientific reports (2019), 9 (1), 1019 ISSN:.Synthetic RNA oligos exhibit purity decreasing as a function of length, because the efficiency of the total synthesis is the numerical product of the individual step efficiencies, typically below 98%. Analytical methods for RNAs up to the 60 nucleotides (nt) have been reported, but they fall short for purity evaluation of 100nt long, used as single guide RNA (sgRNA) in CRISPR technology, and promoted as pharmaceuticals. In an attempt to exploit a single HPLC method and obtain both identity as well as purity, ion-pair reversed-phase chromatography (IP-RP) at high temperature in the presence of an organic cosolvent is the current analytical strategy. Here we report that IP-RP is less suitable compared to the conventional ion-exchange (IEX) for analysis of 100nt RNAs. We demonstrate the relative stability of RNA in the denaturing/basic IEX mobile phase, lay out a protocol to determine the on-the-column stability of any RNA, and establish the applicability of this method for quality testing of sgRNA, tRNA, and mRNA. Unless well resolving HPLC methods are used for batch-to-batch evaluation of man-made RNAs, process development will remain shortsighted, and observed off-target effects in-vitro or in-vivo may be partially related to low purity and the presence of shorter sequences.
- 16Camperi, J.; Lippold, S.; Ayalew, L.; Roper, B.; Shao, S.; Freund, E.; Nissenbaum, A.; Galan, C.; Cao, Q.; Yang, F. Comprehensive Impurity Profiling of mRNA: Evaluating Current Technologies and Advanced Analytical Techniques. Anal. Chem. 2024, 96, 3886– 3897, DOI: 10.1021/acs.analchem.3c05539There is no corresponding record for this reference.
- 17Welbourne, E. N.; Loveday, K. A.; Nair, A.; Nourafkan, E.; Qu, J.; Cook, K.; Kis, Z.; Dickman, M. J. Anion exchange HPLC monitoring of mRNA in vitro transcription reactions to support mRNA manufacturing process development. Front. Mol. Biosci. 2024, 11, 1250833, DOI: 10.3389/fmolb.2024.1250833There is no corresponding record for this reference.
- 18Gilar, M.; Doneanu, C.; Gaye, M. M. Liquid Chromatography Methods for Analysis of mRNA Poly(A) Tail Length and Heterogeneity. Anal. Chem. 2023, 95, 14308– 14316, DOI: 10.1021/acs.analchem.3c02552There is no corresponding record for this reference.
- 19D’Atri, V.; Imiołek, M.; Quinn, C.; Finny, A.; Lauber, M.; Fekete, S.; Guillarme, D. Size exclusion chromatography of biopharmaceutical products: From current practices for proteins to emerging trends for viral vectors, nucleic acids and lipid nanoparticles. J. Chromatogr. A 2024, 1722, 464862 DOI: 10.1016/j.chroma.2024.464862There is no corresponding record for this reference.
- 20Lokras, A.; Chakravarty, A.; Rades, T.; Christensen, D.; Franzyk, H.; Thakur, A.; Foged, C. Simultaneous quantification of multiple RNA cargos co-loaded into nanoparticle-based delivery systems. Int. J. Pharm. 2022, 626, 122171 DOI: 10.1016/j.ijpharm.2022.122171There is no corresponding record for this reference.
- 21Zhang, J.; Haas, R. M.; Leone, A. M. Polydispersity Characterization of Lipid Nanoparticles for siRNA Delivery Using Multiple Detection Size-Exclusion Chromatography. Anal. Chem. 2012, 84, 6088– 6096, DOI: 10.1021/ac300776821https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xot1Olt7o%253D&md5=be22cbff7a62f6d84c570a38f9bd1572Polydispersity Characterization of Lipid Nanoparticles for siRNA Delivery Using Multiple Detection Size-Exclusion ChromatographyZhang, Jingtao; Haas, R. Matthew; Leone, Anthony M.Analytical Chemistry (Washington, DC, United States) (2012), 84 (14), 6088-6096CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)The development of lipid nanoparticle (LNP) based small interfering RNA (siRNA) therapeutics presents unique pharmaceutical and regulatory challenges. In contrast to small mol. drugs that are highly pure and well-defined, LNP drug products can exhibit heterogeneity in size, compn., surface property, or morphol. The potential for batch heterogeneity introduces a complexity that must be confronted in order to successfully develop and ensure quality in LNP pharmaceuticals. Currently, there is a lack of scientific knowledge in the heterogeneity of LNPs as well as high-resoln. techniques that permit this evaluation. This article reports a size-exclusion chromatog. (SEC) method that permits the high-resoln. anal. of LNP size distribution in its native soln. condition. When coupled with multiple detection systems including UV-vis, multi-angle light scattering, and refractive index, online characterization of the distributions in size, mol. wt., and siRNA cargo loading of LNPs could be achieved. Six LNPs with sizes in the rang of 60-140 nm were evaluated and it was found that the SEC sepn. is efficient, highly reproducible, and can be broadly applied to a diverse range of LNPs. A comparison between the current SEC method and asym. field flow fractionation (FFF) shows that the current method provides similar size distribution results on LNPs compared to FFF. Two representative LNPs with similar bulk properties were evaluated in-depth using the SEC method along with two other sizing techniques-dynamic light scattering and cryo-TEM. Profound differences in batch polydispersity were obsd. between them. Despite the similarity in the particle assembly process, it was found that one LNP (A) possessed a narrow size and mol. wt. distribution while the other (B) was polydisperse. The present results suggest that LNP drug products are highly complex and diverse in nature, and care should be taken in examg. and understanding them to ensure quality and consistency. The method developed here can not only serve as a method for understanding LNP product property, permitting control on product quality, but also could serve as a potential manufg. method for product purifn. Understandings obtained in this work can help to facilitate the development of LNPs as a well-defined pharmaceutical product.
- 22D’Atri, V.; Lardeux, H.; Goyon, A.; Imiołek, M.; Fekete, S.; Lauber, M.; Zhang, K.; Guillarme, D. Optimizing Messenger RNA Analysis Using Ultra-Wide Pore Size Exclusion Chromatography Columns. Int. J. Mol. Sci. 2024, 25, 6254, DOI: 10.3390/ijms25116254There is no corresponding record for this reference.
- 23Packer, M.; Gyawali, D.; Yerabolu, R.; Schariter, J.; White, P. A novel mechanism for the loss of mRNA activity in lipid nanoparticle delivery systems. Nat. Commun. 2021, 12, 6777, DOI: 10.1038/s41467-021-26926-023https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisFClsLzL&md5=9ea94bb1f8267764266f7419475c1255A novel mechanism for the loss of mRNA activity in lipid nanoparticle delivery systemsPacker, Meredith; Gyawali, Dipendra; Yerabolu, Ravikiran; Schariter, Joseph; White, PhilNature Communications (2021), 12 (1), 6777CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Lipid nanoparticle (LNP)-formulated mRNA vaccines were rapidly developed and deployed in response to the SARS-CoV-2 pandemic. Due to the labile nature of mRNA, identifying impurities that could affect product stability and efficacy is crucial to the long-term use of nucleic-acid based medicines. Herein, reversed-phase ion pair high performance liq. chromatog. (RP-IP HPLC) was used to identify a class of impurity formed through lipid:mRNA reactions; such reactions are typically undetectable by traditional mRNA purity anal. techniques. The identified modifications render the mRNA untranslatable, leading to loss of protein expression. Specifically, electrophilic impurities derived from the ionizable cationic lipid component are shown to be responsible. Mechanisms implicated in the formation of reactive species include oxidn. and subsequent hydrolysis of the tertiary amine. It thus remains crit. to ensure robust anal. methods and stringent manufg. control to ensure mRNA stability and high activity in LNP delivery systems.
- 24Levin, D. S.; Shepperd, B. T.; Gruenloh, C. J. Combining ion pairing agents for enhanced analysis of oligonucleotide therapeutics by reversed phase-ion pairing ultra performance liquid chromatography (UPLC). J. Chromatogr. B 2011, 879, 1587– 1595, DOI: 10.1016/j.jchromb.2011.03.051There is no corresponding record for this reference.
- 25Hashiba, K.; Taguchi, M.; Sakamoto, S.; Otsu, A.; Maeda, Y.; Ebe, H.; Okazaki, A.; Harashima, H.; Sato, Y. Overcoming thermostability challenges in mRNA–lipid nanoparticle systems with piperidine-based ionizable lipids. Commun. Biol. 2024, 7, 556, DOI: 10.1038/s42003-024-06235-0There is no corresponding record for this reference.
- 26Birdsall, R. E.; Han, D.; DeLaney, K.; Kowalczyk, A.; Cojocaru, R.; Lauber, M.; Huray, J. L. Monitoring stability indicating impurities and aldehyde content in lipid nanoparticle raw material and formulated drugs. J. Chromatogr. B 2024, 1234, 124005 DOI: 10.1016/j.jchromb.2024.124005There is no corresponding record for this reference.
- 27De Vos, J.; Morreel, K.; Alvarez, P.; Vanluchene, H.; Vankeirsbilck, R.; Sandra, P.; Sandra, K. Evaluation of size-exclusion chromatography, multi-angle light scattering detection and mass photometry for the characterization of mRNA. J. Chromatogr. A 2024, 1719, 464756 DOI: 10.1016/j.chroma.2024.464756There is no corresponding record for this reference.
- 28Imiołek, M.; Fekete, S.; Kizekai, L.; Addepalli, B.; Lauber, M. Fast and efficient size exclusion chromatography of adeno associated viral vectors with 2.5 micrometer particle low adsorption columns. J. Chromatogr. A 2024, 1714, 464587 DOI: 10.1016/j.chroma.2023.464587There is no corresponding record for this reference.
- 29Fekete, S.; DeLano, M.; Harrison, A. B.; Shiner, S. J.; Belanger, J. L.; Wyndham, K. D.; Lauber, M. A. Size Exclusion and Ion Exchange Chromatographic Hardware Modified with a Hydrophilic Hybrid Surface. Anal. Chem. 2022, 94, 3360– 3367, DOI: 10.1021/acs.analchem.1c05466There is no corresponding record for this reference.
- 30Kizekai, L.; Addepalli, B.; Jawdat, N.; Chumakov, V.; Gilar, M.; Lauber, M. A. Suitability of GTxResolve Premier BEH SEC 450 Å 2.5 μm Column for Size-based Separations of Nucleic Acids ; Waters Application Note 2023. See the following: https://www.waters.com/nextgen/us/en/library/application-notes/2023/suitability-of-xbridge-premier-gtx-beh-sec-450-25-m-column-for-size-based-separations-of-nucleic-acids.html.There is no corresponding record for this reference.
- 31Fongaro, B.; Campara, B.; Moscatiello, G. Y.; De Luigi, A.; Panzeri, D.; Sironi, L.; Bigini, P.; Carretta, G.; Miolo, G.; Pasut, G. Assessing the physicochemical stability and intracellular trafficking of mRNA-based COVID-19 vaccines. Int. J. Pharm. 2023, 644, 123319 DOI: 10.1016/j.ijpharm.2023.123319There is no corresponding record for this reference.
- 32Koetsier, G.; Cantor, E. A Practical Guide to Analyzing Nucleic Acid Concentration and Purity with Microvolume Spectrophotometers ; New England Biolabs Technical Note 2019. See the following: https://www.neb.com/en/-/media/nebus/files/application-notes/technote_mvs_analysis_of_nucleic_acid_concentration_and_purity.pdf?rev=c24cea043416420d84fb6bf7b554dbbb.There is no corresponding record for this reference.
- 33Ramirez-Cuevas, F. V.; Gurunatha, K. L.; Parkin, I. P.; Papakonstantinou, I. Universal Theory of Light Scattering of Randomly Oriented Particles: A Fluctuational-Electrodynamics Approach for Light Transport Modeling in Disordered Nanostructures. ACS Photonics 2022, 9, 672– 681, DOI: 10.1021/acsphotonics.1c0171033https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XislersL4%253D&md5=85ddef06713dee1fc3f47aa718ff11d2Universal Theory of Light Scattering of Randomly Oriented Particles: A Fluctuational-Electrodynamics Approach for Light Transport Modeling in Disordered NanostructuresRamirez-Cuevas, Francisco V.; Gurunatha, Kargal L.; Parkin, Ivan P.; Papakonstantinou, IoannnisACS Photonics (2022), 9 (2), 672-681CODEN: APCHD5; ISSN:2330-4022. (American Chemical Society)Disordered nanostructures are commonly encountered in many nanophotonic systems, from colloid dispersions for sensing to heterostructured photocatalysts. Randomness, however, imposes severe challenges for nanophotonics modeling, often constrained by the irregular geometry of the scatterers involved or the stochastic nature of the problem itself. In this Article, we resolve this conundrum by presenting a universal theory of averaged light scattering of randomly oriented objects. Specifically, we derive expansion-basis-independent formulas of the orientation-and-polarization-averaged absorption cross section, scattering cross section, and asymmetry parameter, for single or a collection of objects of arbitrary shape. These three parameters can be directly integrated into traditional unpolarized radiative energy transfer modeling, enabling a practical tool to predict multiple scattering and light transport in disordered nanostructured materials. Notably, the formulas of av. light scattering can be derived under the principles of fluctuational electrodynamics, allowing the analogous math. treatment to the methods used in thermal radiation, nonequil. electromagnetic forces, and other assocd. phenomena. The proposed modeling framework is validated against optical measurements of polymer composite films with metal-oxide microcrystals. Our work may contribute to a better understanding of light-matter interactions in disordered systems, such as plasmonics for sensing and photothermal therapy, photocatalysts for water splitting and CO2 dissocn., photonic glasses for artificial structural colors, and diffuse reflectors for radiative cooling, to name just a few.
- 34Goyon, A.; Tang, S.; Fekete, S.; Nguyen, D.; Hofmann, K.; Wang, S.; Shatz-Binder, W.; Fernandez, K. I.; Hecht, E. S.; Lauber, M. Separation of Plasmid DNA Topological Forms, Messenger RNA, and Lipid Nanoparticle Aggregates Using an Ultrawide Pore Size Exclusion Chromatography Column. Anal. Chem. 2023, 95, 15017– 15024, DOI: 10.1021/acs.analchem.3c02944There is no corresponding record for this reference.
- 35Muramatsu, H.; Lam, K.; Bajusz, C.; Laczkó, D.; Karikó, K.; Schreiner, P.; Martin, A.; Lutwyche, P.; Heyes, J.; Pardi, N. Lyophilization provides long-term stability for a lipid nanoparticle-formulated, nucleoside-modified mRNA vaccine. Mol. Ther. 2022, 30, 1941– 1951, DOI: 10.1016/j.ymthe.2022.02.00135https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XjslSgsrs%253D&md5=65b435c164ca92084a2ca06ea701952bLyophilization provides long-term stability for a lipid nanoparticle-formulated, nucleoside-modified mRNA vaccineMuramatsu, Hiromi; Lam, Kieu; Bajusz, Csaba; Laczko, Dorottya; Kariko, Katalin; Schreiner, Petra; Martin, Alan; Lutwyche, Peter; Heyes, James; Pardi, NorbertMolecular Therapy (2022), 30 (5), 1941-1951CODEN: MTOHCK; ISSN:1525-0024. (Cell Press)Lipid nanoparticle (LNP)-formulated nucleoside-modified mRNA vaccines have proven to be very successful in the fight against the coronavirus disease 2019 (COVID-19) pandemic. They are effective, safe, and can be produced in large quantities. However, the long-term storage of mRNA-LNP vaccines without freezing is still a challenge. Here, we demonstrate that nucleoside-modified mRNA-LNPs can be lyophilized, and the physicochem. properties of the lyophilized material do not significantly change for 12 wk after storage at room temp. and for at least 24 wk after storage at 4°C. Importantly, we show in comparative mouse studies that lyophilized firefly luciferase-encoding mRNA-LNPs maintain their high expression, and no decrease in the immunogenicity of a lyophilized influenza virus hemagglutinin-encoding mRNA-LNP vaccine was obsd. after 12 wk of storage at room temp. or for at least 24 wk after storage at 4°C. Our studies offer a potential soln. to overcome the long-term storage-related limitations of nucleoside-modified mRNA-LNP vaccines.
- 36Camperi, J.; Moshref, M.; Dai, L.; Lee, H. Y. Physicochemical and Functional Characterization of Differential CRISPR-Cas9 Ribonucleoprotein Complexes. Anal. Chem. 2022, 94, 1432– 1440, DOI: 10.1021/acs.analchem.1c04795There is no corresponding record for this reference.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.analchem.4c06296.
Additional figures, tables, experimental details and methods, including IP-RP gradient details (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.