Analysis of the Effects of Different Synthesis and Processing Methods on Circular RNA Integrity, Protein Expression, and Removal of Immunogenic Impurities
Download PDF
$currentUrl="http://$_SERVER[HTTP_HOST]$_SERVER[REQUEST_URI]"

Keywords

Nucleic acid therapeutics
RNA manufacturing
Circular RNA
Gene delivery

DOI

10.26689/jcnr.v8i9.8393

Submitted : 2024-08-28
Accepted : 2024-09-12
Published : 2024-09-27

Abstract

Circular RNAs (circRNAs) are emerging as a promising alternative to messenger RNAs (mRNAs) in gene delivery applications due to their enhanced stability and translation. Developing circRNA-based therapeutic platforms requires efficient manufacturing of circRNA with broad scalability. However, the permuted intron-exon (PIE)-based circRNA production commonly used to date involves complex RNA synthesis, circularization, precursor RNA digestion, and impurity removal steps that have limited practical applications. While co-transcriptional circularization could effectively streamline circRNA production, and both cellulose/phosphatase treatment and high-performance liquid chromatography (HPLC) have demonstrated their reliability in mRNA manufacturing, their potential effects on the quality, translation, and reactogenicity of circRNA remained to be fully investigated. Here, using circRNAs systematically manufactured through three independent workflows, we comprehensively examined the utilities of these RNA synthesis and processing methods in circRNA production by comparing the integrity, translation, and immunogenicity of their circRNA products. We began by manufacturing a mNeonGreen (mNG)-encoding circRNA through these workflows and subsequently assessed circRNA integrity via E-gel EX electrophoresis. Protein expression was then monitored in HEK 293T, A549, and DC2.4 cells at 72 hours post-transfection. Finally, we evaluated the immunogenicity of these circRNAs by measuring their interferon beta (IFN-β) induction in A549 cells at 4 hours post-transfection. Using HPLC purification over cellulose and phosphatase treatment resulted in 10–14% higher circRNA enrichment by reducing nicking associated with processing conditions. Protein expression remained consistent across circRNAs from different workflows (P > 0.05), demonstrating that co-transcriptional circularization produces circRNA with translation levels comparable to those obtained from the conventional PIE method. Moreover, both cellulose/phosphatase treatment and HPLC purification effectively minimized IFN-β induction of the purified circRNAs, confirming their reliability in removing immunogenic impurities introduced during in vitro transcription and their compatibility with the co-transcriptional circularization strategy. Collectively, our results provide valuable insights for improving the production efficiency and scalability of circRNA manufacturing that are crucial for addressing key bottlenecks in the development of circRNA-based therapeutic applications.

References

To KKW, Cho WCS, 2021, An Overview of Rational Design of mRNA-Based Therapeutics and Vaccines. Expert Opin Drug Discov, (16): 1307–1317.

Rohner E, Yang R, Foo KS, et al., 2022, Unlocking the Promise of mRNA Therapeutics. Nat Biotechnol, (40): 1586–1600.

Weng Y, Li C, Yang T, et al., 2020, The Challenge and Prospect of mRNA Therapeutics Landscape. Biotechnol Adv, (40): 107534.

Garneau NL, Wilusz J, Wilusz CJ, 2007, The Highways and Byways of mRNA Decay. Nat Rev Mol Cell Biol, (8): 113–126.

von Niessen AGO, Poleganov MA, Rechner C, et al., 2019, Improving mRNA-Based Therapeutic Gene Delivery by Expression-Augmenting 3’ UTRs Identified by Cellular Library Screening. Mol Ther, (27): 824–836.

Barrett SP, Salzman J, 2016, Circular RNAs: Analysis, Expression and Potential Functions. Development, (143): 1838–1847.

Enuka Y, Lauriola M, Feldman ME, et al., 2016, Circular RNAs Are Long-Lived and Display Only Minimal Early Alterations in Response to a Growth Factor. Nucleic Acids Research, (44): 1370–1383.

Wesselhoeft RA, Kowalski PS, Anderson DG, 2018, Engineering Circular RNA for Potent and Stable Translation in Eukaryotic Cells. Nat Commun, (9): 2629.

Chen R, Wang SK, Belk JA, et al., 2023, Engineering Circular RNA for Enhanced Protein Production. Nat Biotechnol, (41): 262–272.

Dousis A, Ravichandran K, Hobert EM, et al., 2023, An Engineered T7 RNA Polymerase that Produces mRNA Free of Immunostimulatory Byproducts. Nat Biotechnol, (41): 560–568.

Cheng ZF, Deutscher MP, 2002, Purification and Characterization of the Escherichia coli Exoribonuclease RNase R: Comparison with RNase II. Journal of Biological Chemistry, (277): 21624–21629.

Vincent HA, Deutscher MP, 2009, Insights into How RNase R Degrades Structured RNA: Analysis of the Nuclease Domain. J Mol Biol, (387): 570–583.

Baiersdorfer M, Boros G, Muramatsu H, et al., 2019, A Facile Method for the Removal of dsRNA Contaminant from In Vitro-Transcribed mRNA. Mol Ther Nucleic Acids, (15): 26–35.

Wesselhoeft RA, Kowalski PS, Parker-Hale FC, et al., 2019, RNA Circularization Diminishes Immunogenicity and Can Extend Translation Duration In Vivo. Molecular Cell, (74): 508–520.

Arnaud-Barbe N, Cheynet-Sauvion V, Oriol G, et al., 1998, Transcription of RNA Templates by T7 RNA Polymerase. Nucleic Acids Res, (26): 3550–3554.

Wu W, Zhang W, Duggan ES, et al., 2015, RIG-I and TLR3 Are Both Required for Maximum Interferon Induction by Influenza Virus in Human Lung Alveolar Epithelial Cells. Virology, (482): 181–188.

Abe BT, Wesselhoeft RA, Chen R, et al., 2022, Circular RNA Migration in Agarose Gel Electrophoresis. Molecular Cell, (82): 1768–1777.

Takeda K, Akira S, 2004, TLR signaling pathways. Seminars in Immunology, (16): 3–9.

Hartman ZC, Black EP, Amalfitano A, 2007, Adenoviral Infection Induces a Multi-Faceted Innate Cellular Immune Response that Is Mediated by the Toll-Like Receptor Pathway in A549 Cells. Virology, (358): 357–372.

Thery C, Amigorena S, 2001, The Cell Biology of Antigen Presentation in Dendritic Cells. Current Opinion in Immunology, (13): 45–51.

Liu X, Zhang Y, Zhou S, et al., 2022, Circular RNA: An Emerging Frontier in RNA Therapeutic Targets, RNA Therapeutics, and mRNA Vaccines. J Control Release, (348): 84–94.

Kariko K, Muramatsu H, Ludwig J, et al., 2011, Generating the Optimal mRNA for Therapy: HPLC Purification Eliminates Immune Activation and Improves Translation of Nucleoside-Modified, Protein-Encoding mRNA. Nucleic Acids Res, (39): e142.

Liu CX, Guo SK, Nan F, et al., 2022, RNA Circles with Minimized Immunogenicity as Potent PKR inhibitors. Molecular Cell, (82): 420–434.

Chen R, Wang SK, Belk JA, et al., 2023, Engineering Circular RNA for Enhanced Protein Production. Nat Biotechnol, (41): 262–272.

Schlee M, Hartmann G, 2016, Discriminating Self from Non-Self in Nucleic Acid Sensing. Nat Rev Immunol, (16): 566–580.

Kim KQ, Burgute BD, Tzeng SC, et al., 2022, N1-Methylpseudouridine Found Within COVID-19 mRNA Vaccines Produces Faithful Protein Products. Cell Rep, (40): 111300.

Nance KD, Meier JL, 2021, Modifications in an Emergency: The Role of N1-Methylpseudouridine in COVID-19 Vaccines. ACS Cent. Sci, (70): 748–756.

AbouHaidar MG, Ivanov IG, 1999, Non-Enzymatic RNA Hydrolysis Promoted by the Combined Catalytic Activity of Buffers and Magnesium Ions. Zeitschrift für Naturforschung C, (54): 542–548.

Bain JD, Switzer C, 1992, Regioselective Ligation of Oligoribonucleotides Using DNA Splints. Nucleic Acids Research, (20): 4372.

Kim YS, Kim DH, An D, et al., 2023, The RNA Ligation Method Using Modified Splint DNAs Significantly Improves the Efficiency of Circular RNA Synthesis. Animal Cells and Systems, (27): 208–218.