Nucleoside-modified messenger RNA

Last updated

A nucleoside-modified messenger RNA (modRNA) is a synthetic messenger RNA (mRNA) in which some nucleosides are replaced by other naturally modified nucleosides or by synthetic nucleoside analogues. [1] modRNA is used to induce the production of a desired protein in certain cells. An important application is the development of mRNA vaccines, of which the first authorized were COVID-19 vaccines (such as Comirnaty and Spikevax).

Contents

Background

A ribosome (depicted in green) creates a protein (depicted here as a string of beads representing amino acids) encoded in an mRNA (depicted as a ribbon of nucleotides) that may be modified to reduce inflammation in the cell. Ribosome mRNA translation en.svg
A ribosome (depicted in green) creates a protein (depicted here as a string of beads representing amino acids) encoded in an mRNA (depicted as a ribbon of nucleotides) that may be modified to reduce inflammation in the cell.

mRNA is produced by synthesising a ribonucleic acid (RNA) strand from nucleotide building blocks according to a deoxyribonucleic acid (DNA) template, a process that is called transcription. [2] When the building blocks provided to the RNA polymerase include non-standard nucleosides such as pseudouridine — instead of the standard adenosine, cytidine, guanosine, and uridine nucleosides — the resulting mRNA is described as nucleoside-modified. [3]

Production of protein begins with assembly of ribosomes on the mRNA, the latter then serving as a blueprint for the synthesis of proteins by specifying their amino acid sequence based on the genetic code in the process of protein biosynthesis called translation. [4]

Overview

To induce cells to make proteins that they do not normally produce, it is possible to introduce heterologous mRNA into the cytoplasm of the cell, bypassing the need for transcription. In other words, a blueprint for foreign proteins is "smuggled" into the cells. To achieve this goal, however, one must bypass cellular systems that prevent the penetration and translation of foreign mRNA. There are nearly-ubiquitous enzymes called ribonucleases (also called RNAses) that break down unprotected mRNA. [5] There are also intracellular barriers against foreign mRNA, such as innate immune system receptors, toll-like receptor (TLR) 7 and TLR8, located in endosomal membranes. RNA sensors like TLR7 and TLR8 can dramatically reduce protein synthesis in the cell, trigger release of cytokines such as interferon and TNF-alpha, and when sufficiently intense lead to programmed cell death. [6]

The inflammatory nature of exogenous RNA can be masked by modifying the nucleosides in mRNA. [7] For example, uridine can be replaced with a similar nucleoside such as pseudouridine (Ψ) or N1-methyl-pseudouridine (m1Ψ), [8] and cytosine can be replaced by 5-methylcytosine. [9] Some of these, such as pseudouridine and 5-methylcytosine, occur naturally in eukaryotes, [10] while m1Ψ occurs naturally in archaea. [11] Inclusion of these modified nucleosides alters the secondary structure of the mRNA, which can reduce recognition by the innate immune system while still allowing effective translation. [9]

Significance of untranslated regions

A normal mRNA starts and ends with sections that do not code for amino acids of the actual protein. These sequences at the 5′ and 3′ ends of an mRNA strand are called untranslated regions (UTRs). The two UTRs at their strand ends are essential for the stability of an mRNA and also of a modRNA as well as for the efficiency of translation, i.e. for the amount of protein produced. By selecting suitable UTRs during the synthesis of a modRNA, the production of the target protein in the target cells can be optimised. [5] [12]

Delivery

Comparing uptake of RNA and modRNA by the cell Comparing uptake of RNA and modRNA by the cell.jpg
Comparing uptake of RNA and modRNA by the cell

Various difficulties are involved in the introduction of modRNA into certain target cells. First, the modRNA must be protected from ribonucleases. [5] This can be accomplished, for example, by wrapping it in liposomes. Such "packaging" can also help to ensure that the modRNA is absorbed into the target cells. This is useful, for example, when used in vaccines, as nanoparticles are taken up by dendritic cells and macrophages, both of which play an important role in activating the immune system. [13]

Furthermore, it may be desirable that the modRNA applied is introduced into specific body cells. This is the case, for example, if heart muscle cells are to be stimulated to multiply. In this case, the packaged modRNA can be injected directly into an artery such as a coronary artery. [14]

Applications

An important field of application are mRNA vaccines.

Replacing uridine with pseudouridine to evade the innate immune system was pioneered by Karikó and Weissman in 2005. [15] [16] They won the 2023 Nobel Prize in Physiology or Medicine as a result of their work. [17]

Another milestone was achieved by demonstrating the life-saving efficacy of nucleoside modified mRNA in a mouse model of a lethal lung disease by the team of Kormann and others in 2011. [18]

N1-methyl-pseudouridine was used in vaccine trials against Zika, [19] [20] [21] HIV-1, [21] influenza, [21] and Ebola [22] in 2017–2018. [23] :5

The first authorized for use in humans were COVID-19 vaccines to address SARS-CoV-2. [24] [25] [26] [27] [28] [29] [30] Examples of COVID-19 vaccines using modRNA include those developed by the cooperation of BioNTech/Pfizer (BNT162b2), and by Moderna (mRNA-1273). [31] [32] [33] The zorecimeran vaccine developed by Curevac, however, uses unmodified RNA, [34] instead relying on codon optimization to minimize the presence of uridine. This vaccine is less effective, however. [35] [16]

Other possible uses of modRNA include the regeneration of damaged heart muscle tissue, [36] [37] an enzyme-replacement tool [38] and cancer therapy. [39] [40]

Related Research Articles

<i>Adenoviridae</i> Family of viruses

Adenoviruses are medium-sized, nonenveloped viruses with an icosahedral nucleocapsid containing a double-stranded DNA genome. Their name derives from their initial isolation from human adenoids in 1953.

Transfection is the process of deliberately introducing naked or purified nucleic acids into eukaryotic cells. It may also refer to other methods and cell types, although other terms are often preferred: "transformation" is typically used to describe non-viral DNA transfer in bacteria and non-animal eukaryotic cells, including plant cells. In animal cells, transfection is the preferred term as transformation is also used to refer to progression to a cancerous state (carcinogenesis) in these cells. Transduction is often used to describe virus-mediated gene transfer into eukaryotic cells.

<span class="mw-page-title-main">RNA editing</span> Molecular process

RNA editing is a molecular process through which some cells can make discrete changes to specific nucleotide sequences within an RNA molecule after it has been generated by RNA polymerase. It occurs in all living organisms and is one of the most evolutionarily conserved properties of RNAs. RNA editing may include the insertion, deletion, and base substitution of nucleotides within the RNA molecule. RNA editing is relatively rare, with common forms of RNA processing not usually considered as editing. It can affect the activity, localization as well as stability of RNAs, and has been linked with human diseases.

<span class="mw-page-title-main">Pseudouridine</span> Chemical compound

Pseudouridine is an isomer of the nucleoside uridine in which the uracil is attached via a carbon-carbon instead of a nitrogen-carbon glycosidic bond.

<span class="mw-page-title-main">Small nucleolar RNA SNORA44</span> Non-coding RNA molecule which functions in the biogenesis of other small nuclear RNAs

In molecular biology, Small nucleolar RNA SNORA44 is a non-coding RNA (ncRNA) molecule which functions in the biogenesis (modification) of other small nuclear RNAs (snRNAs). This type of modifying RNA is located in the nucleolus of the eukaryotic cell which is a major site of snRNA biogenesis. It is known as a small nucleolar RNA (snoRNA) and also often referred to as a 'guide RNA'.

Peptide-based synthetic vaccines are subunit vaccines made from peptides. The peptides mimic the epitopes of the antigen that triggers direct or potent immune responses. Peptide vaccines can not only induce protection against infectious pathogens and non-infectious diseases but also be utilized as therapeutic cancer vaccines, where peptides from tumor-associated antigens are used to induce an effective anti-tumor T-cell response.

mRNA vaccine Type of vaccine

An mRNAvaccine is a type of vaccine that uses a copy of a molecule called messenger RNA (mRNA) to produce an immune response. The vaccine delivers molecules of antigen-encoding mRNA into immune cells, which use the designed mRNA as a blueprint to build foreign protein that would normally be produced by a pathogen or by a cancer cell. These protein molecules stimulate an adaptive immune response that teaches the body to identify and destroy the corresponding pathogen or cancer cells. The mRNA is delivered by a co-formulation of the RNA encapsulated in lipid nanoparticles that protect the RNA strands and help their absorption into the cells.

<span class="mw-page-title-main">COVID-19 vaccine</span> Vaccine against SARS-CoV-2

A COVID‑19 vaccine is a vaccine intended to provide acquired immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID‑19).

<span class="mw-page-title-main">Katalin Karikó</span> Hungarian-American biochemist (born 1955)

Katalin "Kati" Karikó is a Hungarian-American biochemist who specializes in ribonucleic acid (RNA)-mediated mechanisms, particularly in vitro-transcribed messenger RNA (mRNA) for protein replacement therapy. Karikó laid the scientific groundwork for mRNA vaccines, overcoming major obstacles and skepticism in the scientific community. Karikó received the Nobel Prize in Physiology or Medicine in 2023 for her work, along with American immunologist Drew Weissman.

RNA therapeutics are a new class of medications based on ribonucleic acid (RNA). Research has been working on clinical use since the 1990s, with significant success in cancer therapy in the early 2010s. In 2020 and 2021, mRNA vaccines have been developed globally for use in combating the coronavirus disease. The Pfizer–BioNTech COVID-19 vaccine was the first mRNA vaccine approved by a medicines regulator, followed by the Moderna COVID-19 vaccine, and others.

<span class="mw-page-title-main">Uğur Şahin</span> German oncologist and immunologist (born 1965)

Uğur Şahin is a German oncologist and immunologist. He is the founder and CEO of BioNTech, which developed one of the major vaccines against COVID-19. His main fields of research are cancer research and immunology.

<span class="mw-page-title-main">Pfizer–BioNTech COVID-19 vaccine</span> Type of vaccine for humans

The Pfizer–BioNTech COVID-19 vaccine, sold under the brand name Comirnaty, is an mRNA-based COVID-19 vaccine developed by the German biotechnology company BioNTech. For its development, BioNTech collaborated with the American company Pfizer to carry out clinical trials, logistics, and manufacturing. It is authorized for use in humans to provide protection against COVID-19, caused by infection with the SARS-CoV-2 virus. The vaccine is given by intramuscular injection. It is composed of nucleoside-modified mRNA (modRNA) encoding a mutated form of the full-length spike protein of SARS-CoV-2, which is encapsulated in lipid nanoparticles. Initial advice indicated that vaccination required two doses given 21 days apart, but the interval was later extended to up to 42 days in the US, and up to four months in Canada.

<span class="mw-page-title-main">CureVac COVID-19 vaccine</span> Vaccine candidate against COVID-19

The CureVac COVID-19 vaccine was a COVID-19 vaccine candidate developed by CureVac N.V. and the Coalition for Epidemic Preparedness Innovations (CEPI). The vaccine showed inadequate results in its Phase III trials with only 47% efficacy. In October 2021 CureVac abandoned further development and production plans for CVnCoV and refocused efforts on a cooperation with GlaxoSmithKline.

<span class="mw-page-title-main">ZyCoV-D</span> Vaccine candidate against COVID-19

ZyCoV-D is a DNA plasmid-based COVID-19 vaccine developed by Indian pharmaceutical company Cadila Healthcare, with support from the Biotechnology Industry Research Assistance Council. It is approved for emergency use in India.

<span class="mw-page-title-main">Viral vector vaccine</span> Type of vaccine

A viral vector vaccine is a vaccine that uses a viral vector to deliver genetic material (DNA) that can be transcribed by the recipient's host cells as mRNA coding for a desired protein, or antigen, to elicit an immune response. As of April 2021, six viral vector vaccines, four COVID-19 vaccines and two Ebola vaccines, have been authorized for use in humans.

<span class="mw-page-title-main">Walvax COVID-19 vaccine</span> Vaccine candidate against COVID-19

AWcorna, originally termed ARCoV and also known as the Walvax COVID-19 vaccine, is an mRNA COVID-19 vaccine developed by Walvax Biotechnology, Suzhou Abogen Biosciences, and the PLA Academy of Military Science. In contrast to other mRNA COVID vaccines, such as those by Pfizer-BioNtech and Moderna, this vaccine primarily targets the Sars-CoV-2 receptor-binding domain of the spike protein, rather than the entire spike protein. It is approved for Phase III trials in China, Mexico, Indonesia, and Nepal.

<span class="mw-page-title-main">NDV-HXP-S</span> Vaccine candidate against COVID-19

NDV-HXP-S is a COVID-19 vaccine candidate developed under the leadership of Peter Palese, Adolfo García-Sastre, and Florian Krammer at the Icahn School of Medicine at Mount Sinai.

<span class="mw-page-title-main">N1-Methylpseudouridine</span> Chemical compound

N1-Methylpseudouridine is a natural archaeal tRNA component, component of mammalian ribosomal RNA, and "hypermodified" pyrimidine nucleoside used in biochemistry and molecular biology for in vitro transcription and is found in the SARS-CoV-2 mRNA vaccines tozinameran (Pfizer–BioNTech) and elasomeran (Moderna).

<span class="mw-page-title-main">COVID-19 vaccine clinical research</span> Clinical research to establish the characteristics of COVID-19 vaccines

COVID-19 vaccine clinical research uses clinical research to establish the characteristics of COVID-19 vaccines. These characteristics include efficacy, effectiveness, and safety. As of November 2022, 40 vaccines are authorized by at least one national regulatory authority for public use:

<span class="mw-page-title-main">Riccardo Cortese</span> Italian scientist and entrepreneur

Riccardo Cortese was an Italian scientist, entrepreneur, and innovator in the field of gene expression, drug discovery and genetic vaccines. His work led to the development of novel therapeutic strategies for the prevention and cure of viral infections, including HIV, HCV, Ebola and RSV. He pioneered a novel platform technology based on simian adenoviral vectors for prophylactic and therapeutic vaccines, and authored more than 300 publications in peer-reviewed journals in the field of gene expression, transcriptional control, molecular virology and immunology.

References

  1. Chien KR, Zangi L, Lui KO (October 2014). "Synthetic chemically modified mRNA (modRNA): toward a new technology platform for cardiovascular biology and medicine". Cold Spring Harbor Perspectives in Medicine. 5 (1): a014035. doi:10.1101/cshperspect.a014035. PMC   4292072 . PMID   25301935.
  2. Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P (2002). From DNA to RNA (4 ed.). Garland Science.
  3. Pardi N, Weissman D (2017). "Nucleoside Modified mRNA Vaccines for Infectious Diseases". RNA Vaccines. Methods in Molecular Biology. Vol. 1499. Clifton, N.J. pp. 109–121. doi:10.1007/978-1-4939-6481-9_6. ISBN   978-1-4939-6479-6. PMID   27987145.{{cite book}}: CS1 maint: location missing publisher (link)
  4. Lodish H, Berk A, Zipursky SL, Matsudaira P, Baltimore D, Darnell J (2000). The Three Roles of RNA in Protein Synthesis (4th ed.). New York: W. H. Freeman. pp. Sec 4.4.
  5. 1 2 3 Schlake T, Thess A, Fotin-Mleczek M, Kallen KJ (November 2012). "Developing mRNA-vaccine technologies". RNA Biology. 9 (11): 1319–1330. doi:10.4161/rna.22269. PMC   3597572 . PMID   23064118.
  6. Dalpke A, Helm M (June 2012). "RNA mediated Toll-like receptor stimulation in health and disease". RNA Biology. 9 (6): 828–842. doi:10.4161/rna.20206. PMC   3495747 . PMID   22617878.
  7. Karikó K, Buckstein M, Ni H, Weissman D (August 2005). "Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA". Immunity. 23 (2): 165–175. doi: 10.1016/j.immuni.2005.06.008 . PMID   16111635.
  8. Andries O, Mc Cafferty S, De Smedt SC, Weiss R, Sanders NN, Kitada T (November 2015). "N(1)-methylpseudouridine-incorporated mRNA outperforms pseudouridine-incorporated mRNA by providing enhanced protein expression and reduced immunogenicity in mammalian cell lines and mice". Journal of Controlled Release. 217: 337–344. doi:10.1016/j.jconrel.2015.08.051. hdl: 1854/LU-6993270 . PMID   26342664.
  9. 1 2 Svitkin YV, Cheng YM, Chakraborty T, Presnyak V, John M, Sonenberg N (June 2017). "N1-methyl-pseudouridine in mRNA enhances translation through eIF2α-dependent and independent mechanisms by increasing ribosome density". Nucleic Acids Research. 45 (10): 6023–6036. doi:10.1093/nar/gkx135. PMC   5449617 . PMID   28334758.
  10. Hoernes TP, Clementi N, Faserl K, Glasner H, Breuker K, Lindner H, et al. (January 2016). "Nucleotide modifications within bacterial messenger RNAs regulate their translation and are able to rewire the genetic code". Nucleic Acids Research. 44 (2): 852–862. doi:10.1093/nar/gkv1182. PMC   4737146 . PMID   26578598.
  11. Wurm JP, Griese M, Bahr U, Held M, Heckel A, Karas M, et al. (March 2012). "Identification of the enzyme responsible for N1-methylation of pseudouridine 54 in archaeal tRNAs". RNA. 18 (3): 412–420. doi:10.1261/rna.028498.111. PMC   3285930 . PMID   22274954. In contrast, in most archaea this position is occupied by another hypermodified nucleotide: the isosteric N1-methylated pseudouridine.
  12. Orlandini von Niessen AG, Poleganov MA, Rechner C, Plaschke A, Kranz LM, Fesser S, et al. (April 2019). "Improving mRNA-Based Therapeutic Gene Delivery by Expression-Augmenting 3' UTRs Identified by Cellular Library Screening". Molecular Therapy. 27 (4): 824–836. doi:10.1016/j.ymthe.2018.12.011. PMC   6453560 . PMID   30638957.
  13. Zhao L, Seth A, Wibowo N, Zhao CX, Mitter N, Yu C, Middelberg AP (January 2014). "Nanoparticle vaccines". Vaccine. 32 (3): 327–337. doi: 10.1016/j.vaccine.2013.11.069 . PMID   24295808.
  14. Carlsson L, Clarke JC, Yen C, Gregoire F, Albery T, Billger M, et al. (June 2018). "Biocompatible, Purified VEGF-A mRNA Improves Cardiac Function after Intracardiac Injection 1 Week Post-myocardial Infarction in Swine". Molecular Therapy. Methods & Clinical Development. 9: 330–346. doi:10.1016/j.omtm.2018.04.003. PMC   6054703 . PMID   30038937.
  15. Karikó K, Buckstein M, Ni H, Weissman D (August 2005). "Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA". Immunity. 23 (2): 165–175. doi: 10.1016/j.immuni.2005.06.008 . PMID   16111635.
  16. 1 2 Dolgin E (September 2021). "The tangled history of mRNA vaccines". Nature. 597 (7876): 318–324. Bibcode:2021Natur.597..318D. doi:10.1038/d41586-021-02483-w. PMID   34522017. S2CID   237515383.
  17. "The Nobel Prize in Physiology or Medicine 2023".
  18. Kormann, Michael S. D.; Hasenpusch, Günther; Aneja, Manish K.; Nica, Gabriela; Flemmer, Andreas W.; Herber-Jonat, Susanne; Huppmann, Marceline; Mays, Lauren E.; Illenyi, Marta; Schams, Andrea; Griese, Matthias; Bittmann, Iris; Handgretinger, Rupert; Hartl, Dominik; Rosenecker, Joseph (February 2011). "Expression of therapeutic proteins after delivery of chemically modified mRNA in mice". Nature Biotechnology. 29 (2): 154–157. doi:10.1038/nbt.1733. ISSN   1546-1696. PMID   21217696. S2CID   205275040.
  19. Pardi N, Hogan MJ, Pelc RS, Muramatsu H, Andersen H, DeMaso CR, et al. (March 2017). "Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination". Nature. 543 (7644): 248–251. Bibcode:2017Natur.543..248P. doi:10.1038/nature21428. PMC   5344708 . PMID   28151488. we designed a potent anti-ZIKV vaccine … containing the modified nucleoside 1-methylpseudouridine (m1Ψ)
  20. Richner JM, Himansu S, Dowd KA, Butler SL, Salazar V, Fox JM, et al. (March 2017). "Modified mRNA Vaccines Protect against Zika Virus Infection". Cell. 168 (6): 1114–1125.e10. doi:10.1016/j.cell.2017.02.017. PMC   5388441 . PMID   28222903. The mRNA was synthesized … where the UTP was substituted with 1-methylpseudoUTP
  21. 1 2 3 Pardi N, Hogan MJ, Naradikian MS, Parkhouse K, Cain DW, Jones L, et al. (June 2018). "Nucleoside-modified mRNA vaccines induce potent T follicular helper and germinal center B cell responses". The Journal of Experimental Medicine. 215 (6): 1571–1588. doi:10.1084/jem.20171450. PMC   5987916 . PMID   29739835. In this study, we characterize the immunogenicity of three vaccines consisting of m1Ψ-modified, FPLC-purified mRNA-LNPs encoding HIV-1 envelope (Env), ZIKV prM-E, and influenza virus hemagglutinin (HA)
  22. Meyer M, Huang E, Yuzhakov O, Ramanathan P, Ciaramella G, Bukreyev A (January 2018). "Modified mRNA-Based Vaccines Elicit Robust Immune Responses and Protect Guinea Pigs From Ebola Virus Disease". The Journal of Infectious Diseases. 217 (3): 451–455. doi:10.1093/infdis/jix592. PMC   5853918 . PMID   29281112. Two mRNA vaccines were synthesized … where the UTP were substituted with 1-methylpseudo UTP
  23. Morais P, Adachi H, Yu YT (2021-11-04). "The Critical Contribution of Pseudouridine to mRNA COVID-19 Vaccines". Frontiers in Cell and Developmental Biology. 9: 789427. doi: 10.3389/fcell.2021.789427 . PMC   8600071 . PMID   34805188.
  24. "Pfizer and BioNTech Celebrate Historic First Authorization in the U.S. of Vaccine to Prevent COVID-19". www.businesswire.com. 12 December 2020.
  25. Polack FP, Thomas SJ, Kitchin N, Absalon J, Gurtman A, Lockhart S, et al. (December 2020). "Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine". The New England Journal of Medicine. 383 (27): 2603–2615. doi:10.1056/NEJMoa2034577. PMC   7745181 . PMID   33301246. S2CID   228087117.
  26. Hohmann-Jeddi C (2020-11-10). "Hoffnungsträger BNT162b2: Wie funktionieren mRNA-Impfstoffe?". Pharmazeutische Zeitung (in German). Retrieved 2020-11-28.
  27. Abbasi J (September 2020). "COVID-19 and mRNA Vaccines-First Large Test for a New Approach". JAMA. 324 (12): 1125–1127. doi: 10.1001/jama.2020.16866 . PMID   32880613. S2CID   221476409.
  28. Vogel A, Kanevsky I, Che Y, Swanson K, Muik A, Vormehr M, et al. (8 September 2020). "A prefusion SARS-CoV-2 spike RNA vaccine is highly immunogenic and prevents lung infection in non-human primates" (PDF). bioRxiv. doi:10.1101/2020.09.08.280818. S2CID   221589144.
  29. "Conditions of Authorisation for Pfizer/BioNTech COVID-19 Vaccine" (Decision). Medicines & Healthcare Products Regulatory Agency. 8 December 2020.
  30. Office of the Commissioner (14 December 2020). "Pfizer-BioNTech COVID-19 Vaccine". www.fda.gov (Decision). US FDA.
  31. Krammer F (October 2020). "SARS-CoV-2 vaccines in development". Nature. 586 (7830): 516–527. Bibcode:2020Natur.586..516K. doi: 10.1038/s41586-020-2798-3 . PMID   32967006. S2CID   221887746.
  32. "Moderna's Pipeline". Moderna. Retrieved 2020-11-28.
  33. Dolgin E (November 2020). "COVID-19 vaccines poised for launch, but impact on pandemic unclear". Nature Biotechnology: d41587–020–00022-y. doi:10.1038/d41587-020-00022-y. PMID   33239758. S2CID   227176634.
  34. "COVID-19". CureVac. Retrieved 2020-12-21.
  35. Dolgin E (June 2021). "CureVac COVID vaccine let-down spotlights mRNA design challenges". Nature. 594 (7864): 483. Bibcode:2021Natur.594..483D. doi: 10.1038/d41586-021-01661-0 . PMID   34145413. S2CID   235480198.
  36. Kaur K, Zangi L (December 2020). "Modified mRNA as a Therapeutic Tool for the Heart". Cardiovascular Drugs and Therapy. 34 (6): 871–880. doi:10.1007/s10557-020-07051-4. PMC   7441140 . PMID   32822006.
  37. Zangi L, Lui KO, von Gise A, Ma Q, Ebina W, Ptaszek LM, et al. (October 2013). "Modified mRNA directs the fate of heart progenitor cells and induces vascular regeneration after myocardial infarction". Nature Biotechnology. 31 (10): 898–907. doi:10.1038/nbt.2682. PMC   4058317 . PMID   24013197.
  38. Ter Huurne, Menno; Parker, Benjamin L.; Liu, Ning Qing; Qian, Elizabeth Ling; Vivien, Celine; Karavendzas, Kathy; Mills, Richard J.; Saville, Jennifer T.; Abu-Bonsrah, Dad; Wise, Andrea F.; Hudson, James E.; Talbot, Andrew S.; Finn, Patrick F.; Martini, Paolo G. V.; Fuller, Maria (2023-09-07). "GLA-modified RNA treatment lowers GB3 levels in iPSC-derived cardiomyocytes from Fabry-affected individuals". American Journal of Human Genetics. 110 (9): 1600–1605. doi:10.1016/j.ajhg.2023.07.013. ISSN   1537-6605. PMC   10502840 . PMID   37607539.
  39. McNamara MA, Nair SK, Holl EK (2015). "RNA-Based Vaccines in Cancer Immunotherapy". Journal of Immunology Research. 2015: 794528. doi: 10.1155/2015/794528 . PMC   4668311 . PMID   26665011.
  40. Verbeke R, Lentacker I, Wayteck L, Breckpot K, Van Bockstal M, Descamps B, et al. (November 2017). "Co-delivery of nucleoside-modified mRNA and TLR agonists for cancer immunotherapy: Restoring the immunogenicity of immunosilent mRNA". Journal of Controlled Release. 266: 287–300. doi:10.1016/j.jconrel.2017.09.041. PMID   28987878. S2CID   20794075.

Further reading