You are on page 1of 10

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/343650197

mRNA Vaccines: A Mini Review

Article · February 2017

CITATIONS READS

0 920

4 authors, including:

Amit Kumar Monu Karki


Indian Veterinary Research Institute Indian Veterinary Research Institute
46 PUBLICATIONS 322 CITATIONS 16 PUBLICATIONS 61 CITATIONS

SEE PROFILE SEE PROFILE

Gnanavel Venkatesan
Indian Veterinary Research Institute
182 PUBLICATIONS 2,118 CITATIONS

SEE PROFILE

All content following this page was uploaded by Monu Karki on 14 August 2020.

The user has requested enhancement of the downloaded file.


SCIFED
Publishers
Gnanavel Venkatesan, SF Virol Res J, 2017, 2:1

SciFed Virology Research Journal


Mini Review Open Access

mRNA Vaccines: A Mini Review


1
Sasi Kumar, 2Amit Kumar, 3Monu Karki, *4Gnanavel Venkatesan
*4
Division of Virology, ICAR-IVRI, Mukteswar 263 138, Nainital, Uttarakhand, India

Abstract
Nucleic acid-based vaccines are vaccines that containing DNA or RNA encoded with the
target antigen. These type of vaccines are novel, safe, effective, mimicking of live infection, and low
risk of reversion to virulence. RNA-based vaccines offer a safety in their delivery to the cytoplasm.The
characteristics feature of mRNA presents challenges as a nucleic acid-based vaccine approach. mRNA
platform offers strong safety profile and minimal genetic construction for expression of antigen. Here,
the review summarizes the concept of the recent mRNA vaccine development, mechanism of action and
their applications in immunotherapy and vaccine research.

Keywords and therefore it avoids oncogenesis and mutagenesis [6].


mRNA Vaccines Types; Self-Amplifying; These vaccines are momentary information carrier due to
Mechanism; Immune Response; Applications early metabolic degradation within few days. Any protein
can be encoded for development of therapeutic and
Introduction prophylactic vaccines, without affecting the properties of
Nucleic acid-based vaccines (NAVs) like DNA or the mRNA.
RNA- based vaccines consist of antigen-encoding genes.
Nucleic acid-based vaccines are more tolerable and safe Types of mRNA Vaccines
like other vaccines [1, 2]. DNA vaccines have been found There are two forms of mRNA vaccines: self-
to elicit a less immune response than other vaccines. RNA amplifying mRNA vaccines and conventional non-
vaccines induce both cellular and humoral immunity. amplifying mRNA-based vaccines [7]. Both of these
Unlike DNA, mRNA has more advantages than DNA vaccines are well tolerated, safe, and capable of inducing
vaccines [3]. The mRNA was first discovered in 1961. The immune responses [8, 9].
messenger RNA based applications and therapeutics were
first implemented in 1989. In vitro-transcribed mRNA
vaccines encode only required proteins for expression. In *Corresponding author: Gnanavel Venkatesan, Division of Virology,
this review, we provide the mRNA based vaccines, types, ICAR-IVRI, Mukteswar 263 138, Nainital, Uttarakhand, India. E-mail:
mechanism and delivery of mRNA based vaccines and gnanamvirol@gmail.com; Tel: +91 5942-286346
their applications. Received December 14, 2017; Accepted January 09, 2018; Published
January 24, 2018
mRNA Vaccines Citation: Gnanavel Venkatesan (2017) mRNA Vaccines: A Mini Review. SF
mRNA vaccines are safe, simple, inexpensive and Virol Res J 2:1.
possess maximum flexibility. Particularly compared with
Copyright: © 2017 Gnanavel Venkatesan. This is an open-access article
peptide vaccines, they have self-adjuvanting property, lack distributed under the terms of the Creative Commons Attribution License,
of MHC haplotype restriction and do not need to enter the which permits unrestricted use, distribution, and reproduction in any medium,
nucleus [4, 5]. mRNA does not integrate into the genome provided the original author and source are credited.

SF Virol Res J Volume 2 · Issue 1 · 1000010

ISSN:XXXX-XXXX SFJV, an open access journal page 1 of 9


Citation: Gnanavel Venkatesan (2017) mRNA Vaccines: A Mini Review. SF Virol Res J 2:1.

1. Self-Amplifying mRNA Vaccines (SAM) for 2 months [11]. The vector systems like alpha virus,
The self-amplifying vaccines are larger molecules picornavirus, flavivirus provide inherent adjuvant effects
that encode self-amplifying alphavirus genomes and and high transient transgene expression. The delivery
antigen of interest [10]. The structural proteins in the system consists of viral and non-viral delivery system.
genome are replaced by a gene of interest. Compared with The viral delivery system is facilitated by the alpha virus
non-amplifying vaccines, self-amplifying vaccines have a genome. The non-viral delivery system is achieved by
high level of expression of encoded antigen in the host. emulsions, lipids, and polymers, or other compounds.
The duration of expression by self-amplifying vaccines Table 1 enlists the pre-clinical self-amplifying vaccines.
with non-immunogenic luciferase reporter proteins lasts
Table 1: Pre-Clinical Self-Amplifying mRNA Vaccines for Different Infectious/Non-Infectious Diseases

Disease Target Antigen References


Influenza Nucleoprotein (NP) Lazzaro et al., 2015 [12]
Influenza Hemagglutinin and nucleocapsid De´ Moulins et al., 2015 [13]
Influenza Hemagglutinin and nucleocapsid McCullough et al., 2014 [14]
Influenza Hemagglutinin Brazzoli et al., 2015 [15]
Influenza Hemagglutinin Oreshkova et al., 2014 [16]
Influenza Hemagglutinin Hu et al., 2017 [17]
Influenza Hemagglutinin Bahl et al., 2017 [18]
Rabies Glycoprotein Brito et al., 2015 [19]
Dengue virus Ectodomain of E85 Khalil et al., 2014 [20]
HPV E6 and E7 van de Wall et al., 2015 [21]
RSV Fusion glycoprotein Pepini et al., 2017 [22]
HIV HIV gp140 Bogers et al., 2015 [23]
HCV HCV glycoprotein (E1 and E2) Brito et al., 2015 [19]
HCV HCV Non-structural proteins Ip et al., 2014 [24]
RSV RSV fusion glycoprotein Brito et al., 2014 [25]
Cytomegalovirus gH/gL glycoprotein complex Brito et al., 2015 [19]
Enterovirus-71 Epitope SP70 Huang et al., 2015 [26]
Ebola virus Glycoprotein Pyankov et al., 2015 [27]
Cytomegalovirus Pentamer Wen et al., 2014 [28]
Cytomegalovirus hCMVgB and pp65-IE1 fusion Brito et al., 2014 [25]
Zika virus glycoproteins Pardi et al., 2017 [29]
HIV-1 glycoproteins Pardi et al., 2017 [30]
HIV-1 glycoproteins Pardi et al., 2017 [30]
Hemophilia B hFIX DeRosa et al., 2016;
Cancer/Tumour Tumour specific antigens (TSAs) Ramaswamy et al., 2017 [31, 32]
Cancer/Tumour Tumour specific antigens (TSAs) Oberli et al., 2017; Kranz et al., 2016;
Defrancesco, 2017
[33, 34, 35]
RSV: Respiratory Syncytial Virus; HIV: Human Immunodeficiency Virus; HCV: Hepatitis C Virus; HPV: Human Papilloma
Virus.

2. Conventional Non-Amplifying mRNA-Based polymerase enzyme [36]. A regular cap analog (‘m7G(5′)-
Vaccines ppp-(5′)G’) is included [37] or a poly(A) tail and the cap
mRNA synthesis is obtained from ex-vivo may be added by post-transcription. The contaminants like
transcription of a cDNA from purified and invariant nucleotides, abortive transcripts, and oligonucleotides [38]
plasmid DNA by a Bacteriophages T3, T7 or SP6 RNA are removed from samples by extraction and precipitation

SF Virol Res J Volume 2 · Issue 1 · 1000010

ISSN:XXXX-XXXX SFJV, an open access journal page 2 of 9


Citation: Gnanavel Venkatesan (2017) mRNA Vaccines: A Mini Review. SF Virol Res J 2:1.

steps. Pure mRNA products are separated by a single delivered by non-viral methods. Table 2 enlists the pre-
chromatographic step or HPLC [39] for increased protein clinical conventional non-amplifying vaccines reported
expression [40]. After transcription, plasmid DNA is worldwide.
digested by DNAse. The non-amplifying vaccines are

Table 2: Pre-Clinical Non-Amplifying mRNA Vaccines for Different Infectious/Non-Infectious Diseases

Disease Target Antigen References


HIV HIV-1 gag Zhao et al., 2014; Phua et al., 2014 [41, 42]
Allergy Grass pollen allergen Hattinger et al., 2015 [43]
Cancer Fβ2
Van der Jeught et al., 2014 [44]
Cancer Ovalbumin Fotin-Mleczek et al., 2014 [45]
Cervical cancer HPV-16 E7 Peptide Heidenreich et al., 2015 [46]

HIV: Human Immunodeficiency Virus; HPV: Human Papilloma Virus.

mRNA Optimization viral mediated methods, Ex vivo transfected mRNA is an


For translation, cytoplasmic availability and alternative to the naked mRNA vaccination. In this method,
stability, mRNA requires optimization. Cap and poly mRNAs are transfected with monocytes, macrophages, T
(A) tail are essential for cytoplasmic stability. mRNA is cells, dendritic cells (DCs) and mesenchymal stem cells
capped by using regular cap analog or by using vaccinia (MSC) [60] before administration. A strong immune
virus capping complex [47]. The correct orientation of the response may be induced by ex vivo transfected mRNA
cap is obtained by the use of anti-reverse’’ cap analogs vaccination when compared to naked mRNA vaccination
(ARCAs) [48]. Phosphorothioate modified ARCA is used which offers only optimal expression.
for increasing the half-life and translation efficiency [49].
The length of the poly (A) tail should be between 120- Mechanism of Action
150 nucleotides [50, 51]. The efficiency of polysome mRNA vaccines stimulate host machinery system
formation is increased by increasing the length of poly (A) through their self-adjuvanting property. mRNA induces
tail. In mRNA, poly (A) tail is added by recombinant poly cytolyticCD8+ T cell response and type-I-IFN-mediated
(A) polymerase and by encoding poly (A) on the pDNA innate immune response [34]. The innate immune response
template [52]. α globin 3’-UTR is used for stabilization results in cytokine and chemokine production at the
of mRNA and β globin 5’-UTR is used for improving site of injection [22, 61]. Both non-amplifying and self-
translation efficiency [53]. Examples of 5’-UTRs include amplifying vaccines can promote CD4+ T cells, CD8+
tobacco etch virus 5’-UTR [54], human heat shock protein T- cells and antibody-producing B-cells [7, 25, 62]. The
70 5’-UTR [55]. Pseudouridine, chemically modified activation of B cells through MYD88/TLR7-dependent
nucleoside is also used to enhance the translation and signalling pathway by RNA may produce a stimulus
stability [56] to regulate adaptive immunity. A schematic diagram
indicating the mechanism of action of mRNA vaccines in
Delivery System for mRNA Vaccines host immune system is shown as Figure 1.
The non-viral delivery system has more potential
and advantageous than the viral delivery system [19, 57]. mRNA Vaccines Adjuvants
Non-viral methods are preferred over the viral delivery As discussed earlier, mRNA vaccines are found
system for their safety and cost-effectiveness [58, 59]. The to possess self-adjuvanting property. mRNA vaccines
non-viral methods for delivery of vaccines include naked supplemented with the adjuvant molecules are used to
mRNA vaccines, gene gun, protamine condensation; stimulate CTL activity and pAPCs. Some of the adjuvant
adjuvant based vaccines, and encapsulated mRNA vaccines. molecules supplemented with mRNA vaccines are
The positive-sense RNA viruses, alpha viruses are used for protamine, CPG motifs, poly I:C RNA, invariant chain/
the viral delivery system. The glycoproteins (E1 and E2) LAMP-1, GM-CSF and Co-stimulatory molecules like
of alpha virus used for endosomal escape and cell targeting CD40. Among these Adjuvants, GM-CSF mRNA and
in the host. In addition to direct delivery by viral or non- CD40 mRNA improved the induction of CTL activity and

SF Virol Res J Volume 2 · Issue 1 · 1000010

ISSN:XXXX-XXXX SFJV, an open access journal page 3 of 9


Citation: Gnanavel Venkatesan (2017) mRNA Vaccines: A Mini Review. SF Virol Res J 2:1.

Figure 1: Schematic Diagram Showing the Preparation of mRNA Vaccines and its Mechanism of Action on Host Immune System after Delivery

pAPCs respectively [63-65]. include prophylactic and therapeutic vaccines, cancer


immunotherapy, allergy tolerization, protein replacement,
Clinical Applications of mRNA Vaccines genome engineering, gene editing, genetic reprogramming
The clinical applications of the mRNA of cells and tissue engineering. Potenatial broad area of
application of mRNA vaccines is depicted as Figure 2.

Figure 2: Potentials of mRNA Vaccines in Biomedical Sciences

SF Virol Res J Volume 2 · Issue 1 · 1000010

ISSN:XXXX-XXXX SFJV, an open access journal page 4 of 9


Citation: Gnanavel Venkatesan (2017) mRNA Vaccines: A Mini Review. SF Virol Res J 2:1.

Briefly, the applications of mRNA vaccines in different mRNA vaccination using three different influenza antigens,
biomedical field is mentioned as below. namely HA, NA, and NP is found to protect against the
clinical disease. This new vaccine method induced strong
Infectious Diseases antigen-specific B and T cell responses demonstrating
For vaccination against the infectious diseases, that mRNA vaccine is superior for vaccination against
mRNA encoded with the virus associated antigens is infectious pathogens and potentially other vaccine targets.
injected and the first mRNA vaccine protective efficacy mRNA vaccines for some of the infectious diseases like
was demonstrated against influenza as a paradigm [66]. The dengue virus, human papilloma virus, cytomegalovirus,
Table 3: Applications for mRNA Vaccines, Targeted Antigens and their Methods of Delivery

Purpose Antigen Method


Infectious disease -virus-associated antigens mRNA injection
vaccines -Ex vivo DCs transfected mRNA
injection
Cancer Immunotherapy -Standardized preselected -mRNA injection
-Individualized -Ex vivo DCs transfected mRNA
shared antigens/neo antigens injection
-Ex vivo T cells transfected mRNA
injection
Allergy tolerization -Allergens -mRNA injection
Protein replacement -Associated proteins -Ex vivo transfection
(vasopressin, P-selectin, -Ex vivo DCs transfected mRNA
VEGF-A, Erythropoietin) injection
-Ex vivo monocytes, macrophages and
MSCs transfected mRNA injection
Gene editing and genome engineering -Nucleases and transposases -Ex vivo transfection
-Ex vivo gene defects -gene editing
-Ex vivo autologous/allogenic T cells-
gene editing
Genetic reprogramming -Transcription factors -Ex vivo transfection
of cells and tissue -progerin
engineering

DC: Dendritic Cell; VEGFA: Vascular Endothelial Growth Factor A; MSC: Mesenchymal Stem Cell.

influenza, ebola virus, entero virus, rabies and HIV are The mRNA based vaccines for cancer immunotherapy in
under pre-clinical trials as listed in Table 3. clinical trialsinclude prostate cancer, colorectal carcinoma,
melanoma, glioblastoma, hepatocellular carcinoma and
Cancer Immunotherapy acute lymphoblastic leukaemia [57].
mRNA encoded with a tumour associated antigens
(TAA) or total tumor RNA [6] can be then delivered to Allergy Tolerization
the host in order to elicit an immune response against the The immunoglobulin E (IgE) mediated type
antigen of interest. Similarly, mRNA encoded with chimeric I allergic diseases are treated with antigen-specific
antigen receptor (CAR) or TAA-specific T cell receptor immunotherapy. As the mRNA has the strong TH1
(TCR) has been transfected into natural killer cells or T immuno stimulatory capacity [70], encoded mRNA can
cells by electro oration [67]. Transfected cells carrying such induce IgG antibodies and T cell response that compete
mRNA-encoded receptors are able to recognize and kill with the binding sites in IgE antibodies.
tumor cells that express the target antigen. By this mRNA
encoded with various antigen-specific receptors and their Genome Editing
antitumor activity was evaluated in animal models [68, 69]. Genome editing is the best alternative for the gene

SF Virol Res J Volume 2 · Issue 1 · 1000010

ISSN:XXXX-XXXX SFJV, an open access journal page 5 of 9


Citation: Gnanavel Venkatesan (2017) mRNA Vaccines: A Mini Review. SF Virol Res J 2:1.

therapy. Transcription activator-like effectors nucleases References


(TALENs), zinc finger nucleases (ZFNs) and clustered 1. Ulmer JB, Mason PW, Geall A, et al. (2012) RNA based
regularly spaced short palindromic repeat-CRISPR- vaccines. Vaccine 4414-4418.
associated protein 9 (CRISPR-Cas9) are the powerful
2. Kutzler MA, Weiner DB (2008) DNA vaccines: ready for
tools for site-specific modification of genomes [71-73].
prime time?. Nat Rev Genet 776-788.
But these methods have disadvantages like nonspecific
editing which can be minimized by mRNAs encoded 3. Pascolo S (2006) Vaccination with messenger RNA. Methods
with ZFNs, TALENs and Cas9 that can edit genomes by Mol Med 127: 23-40.
disrupting or integrating sequences [57, 74-77]. Similarly,
transposase-encoded mRNA can be used for transposon- 4. Fotin-Mleczek M, Duchardt KM, Lorenz C, et al. (2011)
mediated stable gene transfer [56]. Messenger RNA based vaccines with dual activity induce
balanced TLR-7 dependent adaptive immune responses and
Protein Replacement Therapy provides antitumor activity. J Immunother 34: 1-15.
The proteins which are not expressed, non-
5. Schlake T, Thess A, Fotin-Mleczek M, et al. (2012) Developing
functional foreign protein can be supplemented by protein
mRNA-vaccine technologies. RNA Biol 9: 11, 1319-1330.
replacement therapy through encoded mRNAs. As an
example, mRNA encoded with surfactant protein B (SPB) 6. McNamara MA, Nair SK, Holl EK (2015) RNA-Based
was used against the congenital lethal lung disease caused Vaccines in Cancer Immunotherapy. J Immunol Res Article ID
by the lack of the SPB protein [78], Regulatory T cell 794528.
transcription factor fork head box protein P3 (FOXP3)
used against the airway hyperresponsiveness and allergen- 7. Ulmer JB, Geall AJ (2016) Recent innovations in mRNA
induced tissue inflammation [79], vascular endothelial vaccines. Curr Opin Immunol 41: 18-22.
growth factor A (VEGF-A) against myocardial infarction in
mice [80]. For mRNA based protein replacement delivery, 8. Ljungberg K, Liljeström P (2015) Self-replicating alpha virus
RNA vaccines. Expert Rev Vaccines 14: 177-194.
post-translational modifications have to be considered
like proteolytic processing and protein convertases or 9. Van Lint S, Renmans D, Broos K (2015) The RNA isan Ce of
endoprotease for protein cleavage. mRNA-based cancer therapy. Expert Rev Vaccines 14: 235-251.

Reprogramming of Cells 10. Kim DY, Atasheva S, McAuley AJ, et al. (2014) Enhancement
mRNA is used to modulate the cell phenotype of protein expression by alpha virus replicons by designing self-
and cell function. mRNA encoded with Yamanaka stem replicating sub genomic RNAs. Proc Natl Acad Sci USA 111:
cell factors are inducing pluripotency and also used for 10708-10713.
differentiation of induced pluripotent stem cells (iPSCs)
[81, 82]. For an example, mRNA encoded with myoblast 11. Geall AJ, Verma A, Otten GR, et al. (2012) Nonviral delivery
of self-amplifying RNA vaccines. Proc Natl Acad Sci USA 109:
determination protein into iPSCs results in the direct
14604-14609.
differentiation of myocyte-like cells.
12. Lazzaro S, Giovani C, Mangiavacchi S, et al. (2015) CD8
Conclusion T-cell priming upon mRNA vaccination is restricted to bone-
In the initial pre-clinical studies, it was clear that marrow-derived antigen-presenting cells and may involve
mRNA vaccines are safe, cost-effective and do not require antigen transfer from myocytes. Immunology 146: 312-326.
the cold chain. The activity observed by mRNA vaccines in
prophylactic and therapeutic vaccines and immunotherapy 13. De Moulins T, Milona P, Englezou PC, et al. (2015)
in the models have high impressive effects. mRNA Polyethylenimine-based polyplex delivery of self-replicating
based vaccines will become a groundbreaking platform RNA vaccines. Nanomedicine 12: 711-722.
for vaccination against infectious diseases and cancer
14. McCullough KC, Bassi I, Milona P, et al. (2014) Self-
immunotherapy in future area of immunoprophylaxis. replicating replicon- RNA delivery to dendritic cells by
chitosan-nanoparticles for translation in vitro and in vivo. Mol
Ther Nucleic Acids 3: e173.

SF Virol Res J Volume 2 · Issue 1 · 1000010

ISSN:XXXX-XXXX SFJV, an open access journal page 6 of 9


Citation: Gnanavel Venkatesan (2017) mRNA Vaccines: A Mini Review. SF Virol Res J 2:1.

15. Brazzoli M, Magini D, Bonci A, et al. (2015) Induction of virus replicon-based vaccine expressing enterovirus-71 epitope
broad-based immunity and protective efficacy by self-amplifying confers dual protection from lethal challenges. J Biomed Sci 22:
mRNA vaccines encoding influenza virus hemagglutinin. J Virol 74.
01786-01815.
27. Pyankov OV, Bodnev SA, Pyankova OG, et al. (2015) A
16. Oreshkova N, Cornelissen LAHM, De Haan CAM, et al. Kunjin replicon virus-like particle vaccine provides protection
(2014) Evaluation of nonspreading Rift Valley fever virus as a against ebola virus infection in nonhuman primates. J Infect Dis
vaccine vector using influenza virus hemagglutinin as a model Suppl S368-S371.
antigen. Vaccine 32: 5323-5329.
28. Wen Y, Monroe J, Linton C, et al. (2014) Human
17. Hu Z, Jiao X, Liu X (2017) Antibody Immunity Induced by cytomegalovirus gH/gL/UL128/UL130/UL131A complex
H7N9 Avian Influenza Vaccines: Evaluation Criteria, Affecting elicits potently neutralizing antibodies in mice. Vaccine 32:
Factors, and Implications for Rational Vaccine Design. Front 3796-3804.
Microbiol 8: 1898.
29. Pardi N, Hogan MJ, Pelc RS, et al. (2017) Zika virus
18. Bahl K, Senn JJ, Yuzhakov O, et al. (2017) Pre-clinical and protection by a single low-dose nucleoside-modified mRNA
Clinical Demonstration of Immunogenicity by mRNA Vaccines vaccination. Nature 543: 248-251.
against H10N8 and H7N9 Influenza Viruses. Mol Ther 25:1316-
1327. 30. Pardi N, Secreto AJ, Shan X, et al. (2017) Administration
of nucleoside-modified mRNA encoding broadly neutralizing
19. Brito LA, Kommareddy S, Maione D, et al. (2015) Self- antibody protects humanized mice from HIV-1 challenge. Nat
amplifying mRNA vaccines. Adv Genet 89: 179-233. Commun 8: 14630.

20. Khalil SM, Tonkin DR, Mattocks MD, et al. (2014) A 31. De Rosa F, Guild B, Karve S, et al. (2016) Therapeutic
tetravalent alpha virus-vector based dengue vaccine provides efficacy in a haemophilia B model using a biosynthetic mRNA
effective immunity in an early life mouse model. Vaccine 32: liver depot system. Gene Ther 23: 699-707.
4068-4074.
32. Ramaswamy S, Tonnu N, Tachikawa K, et al. (2017) Systemic
21. Van De Wall S, Walczak M, Van Rooij N, et al. (2015) Tattoo delivery of factor IX messenger RNA for protein replacement
delivery of a semliki forest virus-based vaccine encoding human therapy. Proc Natl Acad Sci USA 114: E1941-E1945.
papilloma virus E6 and E7. Vaccines 3: 221-238.
33. Oberli MA, Reichmuth AM, Dorkin JR, et al. (2017)
Lipid nano particle assisted mRNA delivery for potent cancer
22. Pepini T, Pulichino AM, Carsillo T, et al. (2017) Induction immunotherapy. Nano Lett 17: 1326-1335.
of an IFN-Mediated Antiviral Response by a Self-Amplifying
RNA Vaccine: Implications for Vaccine Design. J Immunol 34. Kranz LM, Diken M, Haas H, et al. (2016) Systemic RNA
198:4012-4024. delivery to dendritic cells exploits antiviral defence for cancer
immunotherapy. Nature 534: 396-401.
23. Bogers WM, Oostermeijer H, Mooij P, et al. (2015) Potent
immune responses in rhesus macaques induced by non viral 35. Defrancesco L (2017) The “anti-hype” vaccine. Nat
delivery of a self-amplifying RNA vaccine expressing HIV type Biotechnol 35: 193-197.
1 envelope with a cationic nano emulsion. J Infect Dis 211: 947-
955. 36. Krieg PA, Melton DA (1984) Functional messenger RNAs
are produced by SP6 in vitro transcription of cloned cDNAs.
24. Ip PP, Boerma A, Regts J, et al. (2014) Alpha virus-based Nucleic Acids Res 12: 7057-7070.
vaccines encoding nonstructural proteins of hepatitis C virus
induce robust and protective T-cell responses. Mol Ther 22: 881- 37. Konarska MM, Padgett RA, Sharp PA (1984) Recognition
890. of cap structure in splicing in vitro of mRNA precursors. Cell
38: 731-736.
25. Brito LA, Chan M, Shaw CA, et al. (2014) A cationic nano
emulsion for the delivery of next-generation RNA vaccines. Mol 38. Gong P, Martin CT (2006) Mechanism of instability in
Ther 22: 2118-2129. abortive cycling by T7 RNA polymerase. J Biol Chem 281:
23533-23544.
26. Huang YT, Liao JT, Yen LC, et al. (2015) Japanese encephalitis

SF Virol Res J Volume 2 · Issue 1 · 1000010

ISSN:XXXX-XXXX SFJV, an open access journal page 7 of 9


Citation: Gnanavel Venkatesan (2017) mRNA Vaccines: A Mini Review. SF Virol Res J 2:1.

39. Pascolo S (2004) Messenger RNA-based vaccines. Expert


Opin Biol Ther 4: 1285-1294. 51. Tcherepanova IY, Adams MD, Feng X (2008) Ectopic
expression of a truncated CD40L protein from synthetic post-
40. Probst J, Weide B, Scheel B, et al. (2007) Spontaneous transcriptionally capped RNA in dendritic cells induces high
cellular uptake of exogenous messenger RNA in vivo is nucleic levels of IL-12 secretion. BMC Mol Biol 90.
acid-specific, saturable and ion-dependent. Gene Ther 14: 1175-
1180. 52. Munroe D, Jacobson A (1990) mRNA Poly (A) tail, a3’
enhancer of translational initiation. J Cell Biol Mol Sci 3441-
41. Zhao M, Li M, Zhang Z, et al. (2015) Induction of HIV-1 3455.
gag specific immune responses by cationic micelles mediated
delivery of gag mRNA. Drug Deliv 23: 2596-2607. 53. Zohra FT, Chowdhury EH, Tada S, et al. (2007) Effective
delivery with enhanced translational activity synergistically
42. Phua KKL, Staats HF, Leong KW, et al. (2014) Intranasal accelerates mRNA-based transfection. Biochem Biophys Res
mRNA nano particle vaccination induces prophylactic and Commun 358: 373-378.
therapeutic anti-tumor immunity. Sci Rep 4: 5128.
54. Gallie DR, Tanguay RL, Leathers V (1995) The tobacco
43. Hattinger E, Scheiblhofer S, Roesler E, et al. (2015) etch viral 5′ leader and poly(A) tail are functionally synergistic
Prophylactic mRNA vaccination against allergy confers long- regulators of translation. Gene 165: 233-238.
term memory responses and persistent protection in mice. J
Immunol Res 1-12. 55. Vivinus S, Baulande S, Van Zanten M, et al. (2001) An
element within the 5′ untranslated region of human Hsp70
44. Van Der Jeught K, Joe PT, Bialkowski L, et al. (2014) mRNA which acts as a general enhancer of mRNA translation.
Intratumoral administration of mRNA encoding a fusokine Eur J Biochem 268: 1908-1917.
consisting of IFN-b and the ectodomain of the TGF-b receptor
II potentiates antitumor immunity. Onco Target 5: 10100-10113. 56. Kreiter S, Selmi A, Diken et al. (2010) Intranodal vaccination
with naked antigen-encoding RNA elicits potent prophylactic
45. Fotin-Mleczek M, Zanzinger K, Heidenreich R, et al. (2014) and therapeutic antitumoral immunity. Cancer Res 70: 9031-
mRNA-based vaccines synergize with radiation therapy to 9040.
eradicate established tumors. Radiat Oncol 9: 180.
57. Ulmer JB, Mansoura MK, Geall AJ (2015) Vaccines ‘on
46. Heidenreich R, Jasny E, Kowalczyk A, et al. (2015) A novel demand’: science fiction or a future reality. Expert Opin Drug
RNA-based adjuvant combines strong immuno stimulatory Discov 10: 101-106.
capacities with a favorable safety profile. J Cancer 137: 372-
384. 58. Juliano R, Alam MR, Dixit V, et al. (2008) Mechanisms
and strategies for effective delivery of antisense and siRNA
47. Venkatesan S, Gershowitz A, Moss B (1980) Modification of oligonucleotides. Nucleic Acids Res 36: 4158-4171.
the 5′ end of mRNA. Association of RNA triphosphatase with the
RNA guanylyltransferase-RNA (guanine-7-) methyltransferase 59. Pack DW, Hoffman AS, Pun S, et al. (2005) Design and
complex from vaccinia virus. J Biol Chem 255: 903-908. development of polymers for gene delivery. Nat Rev Drug
Discov 4: 581-593.
48. Stepinski J, Waddell C, Stolarski R, et al. (2001) Synthesis
and properties of mRNAs containing the novel “anti-reverse” 60. Sahin U, Karikó K, Türeci Ö. (2014) mRNA-based
cap analogs 7-methyl (3′-O-methyl) GpppG and 7-methyl therapeutics developing a new class of drugs. Nat Rev Drug
(3′-deoxy) GpppG. RNA 7: 1486-1495. Discov 13: 759-780.

49. Grudzien-Nogalska E, Jemielity J, Kowalska J, et al. (2007) 61. Edwards DK, Jasny E, Yoon H, et al. (2017) Adjuvant effects
Phosphorothioate cap analogs stabilize mRNA and increase of a sequence-engineered mRNA vaccine: translational profiling
translational efficiency in mammalian cells RNA 13: 1745-1755. demonstrates similar human and murine innate response. J
Transl Med 15: 1.
50. Mockey M, Goncalves C, Dupuy FP, et al. (2006) mRNA
transfection of dendritic cells: synergistic effect of ARCA 62. Iavarone C, O’Hagan DT, Yu D, et al. (2017) Mechanism of
mRNA capping with Poly (A) chains in cis and in trans for a action of mRNA-based vaccines. Expert Rev Vaccines 16.
high protein expression level. Biochem Biophys Res Commun
1062-1068. 63. Caux C, Massacrier C, Vanbervliet B, et al. (1994) Activation

SF Virol Res J Volume 2 · Issue 1 · 1000010

ISSN:XXXX-XXXX SFJV, an open access journal page 8 of 9


Citation: Gnanavel Venkatesan (2017) mRNA Vaccines: A Mini Review. SF Virol Res J 2:1.

of human dendritic cells through CD40 cross-linking. J Exp Med


180: 1263-1272. 76. Wang H, Yang H, Shivalila CS et al. (2013) One-step
generation of mice carrying mutations in multiple genes by
64. Hess PR, Boczkowski D, Nair SK, et al. (2006) Vaccination CRISPR/Cas mediated genome engineering. Cell 153: 910-918.
with mRNAs encoding tumor-associated antigens and
granulocyte-macrophage colony stimulating factor efficiently 77. Wefers B, Panda SK, Ortiz O, et al. (2013) Generation of
primes CTL responses but is insufficient to overcome tolerance targeted mouse mutants by embryo microinjection of TALEN
to a model tumor/self-antigen. Cancer Immunol Immunother 55: mRNA. Nat Protoc 8: 2355-2379.
672-683.
78. Kormann MS, Hasenpusch G, Aneja MK, et al. (2011)
65. Weide B, Pascolo S, Scheel B, et al. (2009) Direct injection of Expression of therapeutic proteins after delivery of chemically
protamine-protected mRNA: results of a phase 1/2 vaccination modified mRNA in mice. Nature Biotech 29: 154-157.
trial in metastatic melanoma patients. J Immunother 32: 498-
507. 79. Mays LE, Ammon-Treiber S, Mothes B, et al. (2013)
Modified Foxp3 mRNA protects against asthma through an IL-
66. Petsch B, Schnee M, Vogel AB, et al. (2012) Protective 10-dependent mechanism. J Clin Invest 123: 1216-1228.
efficacy of in vitro synthesized, specific mRNA vaccines against
influenza A virus infection. Nat Biotechnol 30: 1210-1216. 80. Zangi L, Lui KO, Von Gise A, et al. (2013) Modified mRNA
directs the fate of heart progenitor cells and induces vascular
67. Sahin U, Karikó K, Türeci O (2014) mRNA-based regeneration after myocardial infarction. Nature Biotech 31:
therapeutics - developing a new class of drugs. Nat Rev Drug 898-907.
Discov 13: 759-780.
81. Warren L, Manos PD, Ahfeldt T, et al. (2010) Highly efficient
68. Zhao Y, Moon E, Carpenito C, et al. (2010) Multiple injections reprogramming topluripotency and directed differentiation of
of electro orated autologous T cells expressing a chimeric human cells with synthetic modified mRNA. Cell Stem Cell 7:
antigen receptor mediate regression of human disseminated 618-630.
tumor. Cancer Res 70: 9053-9061.
82. Human Vaccines and Immunotherapeutics: News, Human
69. Barrett DM, Liu X, Jiang S, et al. (2013) Regimen specific Vaccines and Immunotherapeutics 13: 1208-1209.
effects of RNA modified chimeric antigen receptor T cells in
mice with advanced leukemia. Hum Gene Ther 24: 717-727. Citation: Gnanavel Venkatesan (2017) mRNA Vaccines: A Mini Review.
SF Virol Res J 2:1.
70. Valenta R, Ferreira F, Focke-Tejkl M, et al. (2010) From
allergen genes to allergy vaccines. Annu Rev Immunol 28: 211-
241.

71. Miller JC, Holmes MC, Wang J, et al. (2007) An improved


zinc-finger nuclease architecture for highly specific genome
editing. Nature Biotech 25: 778-785.

72. Hockemeyer D, Wang H, Kiani S, et al. (2011) Genetic


engineering of human pluripotent cells using TALE nucleases.
Nature Biotech 29: 731-734.

73. Mali P, Yang L, Esvelt KM, et al. (2013) RNA-guided human


genome engineering via Cas9. Science 339: 823-826.

74. Doyon Y, McCammon JM, Miller JC et al. (2008) Heritable


targeted gene disruption in zebrafish using designed zinc-finger
nucleases. Nature Biotech 26: 702-708.

75. Tesson L, Usal C, Ménoret S, et al. (2011) Knockout rats


generated by embryo microinjection of TALENs. Nature Biotech
29: 695-696.

SF Virol Res J Volume 2 · Issue 1 · 1000010

ISSN:XXXX-XXXX SFJV, an open access journal page 9 of 9

View publication stats

You might also like