Вы находитесь на странице: 1из 8

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

net/publication/340313439

Recent advances in mRNA vaccine technology

Article  in  Current Opinion in Immunology · August 2020


DOI: 10.1016/j.coi.2020.01.008

CITATIONS READS

0 66

3 authors, including:

Norbert Pardi Drew Weissman


University of Pennsylvania University of Pennsylvania
35 PUBLICATIONS   847 CITATIONS    134 PUBLICATIONS   9,295 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

HIV/AIDS Pathogenesis View project

l;k;'ghhghghggfhhfg View project

All content following this page was uploaded by Norbert Pardi on 02 April 2020.

The user has requested enhancement of the downloaded file.


Available online at www.sciencedirect.com

ScienceDirect

Recent advances in mRNA vaccine technology


Norbert Pardi1, Michael J Hogan2 and Drew Weissman1

Messenger RNA (mRNA) vaccines represent a relatively new including the incorporation of modified nucleosides
vaccine class showing great promise for the future. This [4,5] (particularly modified uridine), optimization of cod-
optimism is built on recently published studies demonstrating ing sequences [6], and stringent purification of IVT
the efficacy of mRNA vaccines in combatting several types of mRNA by high performance liquid chromatography
cancer and infectious pathogens where conventional vaccine (HPLC) [7] to remove double-stranded RNA (dsRNA)
platforms may fail to induce protective immune responses. contaminants; all of these techniques serve to dampen the
These results would not have been possible without critical innate sensing of synthetic mRNA, thus reducing toxicity
recent innovations in the field, such as the development of safe and improving translation of the mRNA [8]. The final
and efficient materials for in vivo mRNA delivery and advanced impediment to the viability of mRNA therapeutics has
protocols for the production of high quality mRNA. This review been inefficient cytoplasmic delivery. Although several
summarizes the most important developments in mRNA approaches such as ex vivo-loaded dendritic cells (DCs),
vaccines from the past few years and discusses the challenges intranodal delivery of mRNA, and mechanical methods
and future directions for the field. (gene gun, electroporation) were developed to deliver
naked mRNA for vaccination [9], these approaches are
Addresses either complicated and expensive (ex vivo loading of DCs)
1
Department of Medicine, University of Pennsylvania, Philadelphia, PA, or difficult to use in humans (intranodal delivery, elec-
19104, USA
2
Department of Pathology and Laboratory Medicine, Children’s Hospital
troporation); thus, the ideal way to deliver mRNA would
of Philadelphia, Philadelphia, PA, 19104, USA be with a material that protects it from degradation and
facilitates efficient cellular uptake after simple injection.
Corresponding authors: The past few years have witnessed a surge in develop-
Pardi, Norbert (pnorbert@pennmedicine.upenn.edu), Weissman, ment of highly efficacious delivery materials for nucleic
Drew (dreww@pennmedicine.upenn.edu)
acids, with some remarkable results [10].

Current Opinion in Immunology 2020, 65:14–20 Recent innovations


This review comes from a themed issue on Vaccines The most important innovations in mRNA vaccine tech-
Edited by Bali Pulendran and Rino Rappuoli
nology in recent years have been in the areas of: 1)
engineering of mRNA sequences, 2) development of
methods that enable simple, rapid and large-scale cGMP
production of mRNA; and 3) development of highly
https://doi.org/10.1016/j.coi.2020.01.008 efficient and safe mRNA vaccine delivery materials.
0952-7915/ã 2020 Published by Elsevier Ltd.
Engineering of mRNA sequences
Given that the 5’ and 3’ untranslated regions (UTRs) of
mRNA can significantly influence the rate of translation
and half-life of the transcript, optimization of the UTRs is
of interest to mRNA vaccine design [11–13]. A recent
Introduction: overcoming early challenges for study used a cell culture-based systematic selection pro-
RNA vaccines cess to identify new UTRs that significantly increase
The concept of genetic (DNA and RNA) vaccines was protein expression from IVT mRNA [14]. The authors
raised decades ago with the hope that a flexible, easy-to- identified several 3’ UTR sequences that could induce
produce, safe and effective vaccine class could be gener- approximately threefold higher protein production from
ated. Until the late 2000s, the emphasis was on the the associated transcript compared to the benchmark of
development of DNA-based approaches [1] due to the the human b-globin 3’ UTR. The novel 3’ UTR motifs
hurdles stemming from RNA’s instability, inefficient in were validated in mRNA vaccination and genetic repro-
vivo delivery, and its stimulation of excessive inflamma- gramming studies where they induced more potent ther-
tory responses. Producing in vitro-transcribed (IVT) mes- apeutic effects compared to mRNA with the b-globin 3’
senger RNA (mRNA) is a fairly straightforward process UTR.
[2,3], but making high quality ‘therapeutic’ mRNA that is
highly translatable and does not induce serious inflam- As mentioned above, the vast majority of effective mRNA
mation were major limitations in the field until very vaccines use some kind of a delivery material, but two
recently. By the early 2010s, the latter problem was recent publications reported on an interesting new vac-
largely resolved by a number of critical innovations, cine format that showed great potency in the absence of a

Current Opinion in Immunology 2020, 65:14–20 www.sciencedirect.com


Recent advances in mRNA vaccine technology Pardi, Hogan and Weissman 15

delivery material [15,16]. This method utilizes the co- Figure 1


delivery of an mRNA encoding an alphavirus RNA-
dependent RNA polymerase plus a second mRNA that Linearized DNA template
encodes the antigen of interest and alphavirus genomic (a) 2017 methods (b) 2019 methods
features enabling its replication in the cytoplasm. This so-
In vitro transcription
called ‘transreplicon’ or ‘splitzicon’ system builds on the In vitro transcription
with CleanCap®
dose-sparing properties of self-amplifying mRNA [9], as a
very low dose (50 ng) was effective in inducing protective
immune responses in mice, even when delivered as AAA AAA
ppp cap1
unformulated mRNA. These findings are particularly
attractive as the utilization of low doses decreases the
Enzymatic capping
cost of the vaccine production. The absence of a delivery
material further decreases the cost, simplifies manufactur- cap1
AAA

ing and raises the possibility of vaccine lyophilization and


Cellulose
storage at ambient temperature. adsorption
HPLC
Optimization of mRNA production purification
Development of methods that enable rapid, simple, large-
scale and inexpensive production of high-quality mRNA
is a critical requirement for the future implementation of AAA AAA
cap1 cap1
mRNA vaccines. Although production of ‘therapeutic’
quality mRNA is not particularly challenging at present
Formulation of mRNA
[2,3], current protocols may not be ideal for mass produc-
tion. Most current mRNA production protocols (summa-
rized in Figure 1) entail separate enzymatic reactions for Vaccination
DNA template preparation, IVT, and 5’ capping, with Current Opinion in Immunology

nucleic acid precipitation after each step to remove other


reaction components. This iterative process involves Schematic of recent innovations in mRNA preparation technology.
sample loss at each step and increases the time and cost Shown are (a) methods used by some groups to prepare non-
inflammatory mRNA circa 2017 (e.g. [35]) and (b) simplified methods
of production. Additionally, some current mRNA purifi- to prepare the same, available as of the writing of this manuscript in
cation methods — notably, HPLC purification — are not 2019.
easily scalable, further bottlenecking mass production.
While current approaches work well at small scale, they
are suboptimal for large-scale manufacturing. Ideally, the manufacture of therapeutic mRNA, and it is likely that
final mRNA product would be prepared in a ‘one-pot’ additional research in this vein will further improve the
reaction with a highly scalable purification method. Two simplicity and cost-efficiency of mRNA production.
recently described innovations have made progress
towards this end. The first pertains to a co-transcriptional Development of materials for efficient in vivo mRNA
capping strategy called CleanCap1, which adds a natural delivery
5’ cap1 structure to a specific transcription start sequence As discussed above, the vast majority of mRNA vaccines
during IVT [17]. This development is significant are designed to be injected with a carrier molecule that
because previous protocols used either enzymatic cap- protects mRNA from rapid degradation and delivers it to
ping [2] — adding additional reaction components and the cytoplasm without significant toxicity. Recent work
purification steps to manufacturing — or co-transcrip- has resulted in the development of multiple types of
tional capping with cap0 resulting innate immune activa- lipid-based carriers, polymers, and peptides that have
tion when the IVT mRNA preparation contains dsRNA all showed promise in preclinical and some clinical stud-
contaminant [18,19]. The second methodological innova- ies [10]. It is worth emphasizing that no efficient broadly
tion has offered an attractive alternative to HPLC purifi- applicable in vivo mRNA delivery materials were avail-
cation that is useful at laboratory and industrial scales able until very recently; thus, we believe that the most
[20]: Baiersdorfer et al. described a simple method for significant recent progress in the mRNA vaccine field has
mRNA purification via adsorption of double-stranded occurred in this area and that it has played a critical role in
RNA contaminants to cellulose, a cheap and abundant advancing mRNA vaccines through several landmark
polysaccharide. The authors demonstrated that this efficacy studies published within the last three years.
highly scalable and inexpensive method works just as
well as HPLC to remove dsRNA contaminants from IVT Polymers
mRNA samples. Taken together, research in the past Some early studies successfully used non-lipid polymers
couple of years has greatly facilitated the large-scale such as polyethylenimine (PEI) for mRNA vaccine

www.sciencedirect.com Current Opinion in Immunology 2020, 65:14–20


16 Vaccines

delivery in preclinical models [9] but those studies did not [42] to an influenza vaccine trial with inactivated influenza
reach clinical phase. Polymers used for mRNA delivery virus vaccines [43]. The magnitude and durability of
are often modified with fatty acid chains to improve the immune responses were relatively modest compared to
safety profile of the delivery material [21]. There has been preclinical studies [29] and will need to be improved. Of
significant progress in the field: Haabeth et al. and McKin- note,theionizable LNPsused intheabovestudiesappearto
lay et al. have recently developed novel lipid-containing contribute to effective immune responses through multiple
polymers called charge-altering releasable transporters mechanisms that are not fully appreciated, including effi-
(CARTs) that have efficiently targeted T cells and cient cellular uptake andtranslation of mRNA [27] as well as
resulted in clearance of established tumors in mice a recently reported adjuvant effect [34,44].
[22,23]. Manipulation of T cells is difficult and often
requires ex vivo operations (purification of T cells Of note, the above-mentioned preclinical and clinical
obtained from donors, electroporation with nucleic acid, trials were initiated years ago; thus, we presume that
expansion and reinfusion); thus, CARTs are very attrac- more effective LNP formulations will become available.
tive delivery materials with great potential in the areas of In this regard, similarly to the polymer-based CART
mRNA vaccines and gene therapy. Chahal et al. have platform, selective T cell targeting has also been achieved
described branched polyamine-based polymers called by mRNA-LNPs. Veiga et al. have used a novel platform,
dendrimers formulated with a lipid-anchored polyethyl- called ASSET (Anchored Secondary scFv Enabling Tar-
ene glycol (PEG) and an antigen-encoding, self-amplify- geting), in which a T cell-specific monoclonal antibody is
ing mRNA. Immunization with a single intramuscular linked to LNPs in order to target leukocytes [45]. This
dose of dendrimer-RNA nanoparticles elicited antigen- flexible platform holds great potential for mRNA vaccine
specific CD8+ T cell and neutralizing antibody responses and other applications as well.
against Zika, Ebola and influenza viruses and Toxoplasma
gondii in mice [24]. Kranz et al. have reported on lipoplexes that preferentially
target DCs after systemic delivery [46]. Selective DC
Peptides targeting with mRNA vaccines to induce strong immune
Cell-penetrating peptides (CPPs) are rarely used for responses is a potentially critical finding, and this platform
mRNA vaccines but there has been some recent progress has already demonstrated promise in clinical trials and has
in the field: Udhayakumar et al. developed CPPs contain- been actively investigated in the context of personalized
ing amphipathic Arg-Ala-Leu-Ala motifs to condense cancer vaccines [47,48].
mRNA into particles that can disrupt and penetrate
membranes and demonstrated potent cytolytic T cell Future directions and outstanding questions
responses after immunizing mice with CPP-complexed regarding mRNA vaccines
mRNA [25]. It will be important for future studies to While the past several years have witnessed a rapid pace
explore whether this platform can induce potent antibody of innovation in mRNA manufacturing, in vivo delivery,
responses or protection from pathogen infection. and immunogenicity, there remains much room for
improvement and investigation. Here, we briefly high-
Lipid nanoparticles light three interrelated topics that, if better understood,
Ionizable lipid-containing nanoparticles (LNPs), initially could propel the field further: (1) differences in mRNA
developed for siRNA delivery, are the most widely used preparation, (2) differences between animal models and
in vivo mRNA delivery materials at present [26]. After a humans, and (3) mechanisms of immunogenicity of
proof-of-concept for the in vivo translation of mRNA-LNPs mRNA vaccines.
was demonstrated in 2015 [27], multiple vaccine studies
have used LNPs with unmodified or nucleoside-modified mRNA preparation
mRNA that induced durable, protective immune responses It is currently unknown whether certain formats of
against multiple infectious pathogens, often after a single mRNA vaccines are more effective for certain types of
dose [28–34,35,36–40], and have been effective in com- immune responses or certain disease applications. This
batting cancer as well [41]. Several clinical trials using question is prompted by the fact that the inflammatory
mRNA-LNPs are underway, and some published data from profile of synthetic mRNA can be dramatically altered by
two Phase I influenza virus vaccine trials (NCT03076385 multiple variables, including modified nucleosides [4,5],
and NCT03345043) are available [29,42]. In these trials, purification to remove dsRNA species [7,20], sequence
healthy adults were immunized twice, three weeks apart, engineering [6] intracellular RNA replication [49], and
with placebo or nucleoside-modified mRNA-LNPs encod- delivery material [32,44,49]. The resulting cytokine and
ing full-length influenza virus H10 and H7 hemagglutinin chemokine milieu could be reasonably expected to sig-
(HA), and safety and immunogenicity were evaluated. nificantly impact the magnitude and/or quality of T and B
Nucleoside-modified mRNA-LNP influenza vaccines cell responses in ways that may be more useful for some
induced humoral immune responses in the vaccine recip- disease applications than others. Relevant mechanisms
ients and the safety profile of the vaccines was comparable may include the impact of type I IFNs on the inhibition of

Current Opinion in Immunology 2020, 65:14–20 www.sciencedirect.com


Recent advances in mRNA vaccine technology Pardi, Hogan and Weissman 17

mRNA translation and the chemoattraction or functional of Tfh, GC, and neutralizing antibody responses to these
modulation (activation, differentiation, etc.) of antigen- vaccines [55]. It remains to be seen whether these various
presenting cells, lymphocytes, or other immune cells at vaccine formats have truly distinct immunogenic proper-
the site of vaccination or draining lymphoid tissues [50]. ties. We would like to particularly emphasize that very
In recent reports, nucleoside-modified mRNA-LNP vac- few studies have directly compared the in vivo efficacy of
cines have been particularly associated with potent T different mRNA vaccine formats [34,56], and these stud-
follicular helper (Tfh) cell, germinal center (GC) B cell, ies in general have not compared the protective or thera-
and neutralizing antibody responses [34,51]. However, it peutic efficacy of the various vaccines; therefore, this will
is currently unknown whether nucleoside modification is be a critical area of future research.
explicitly required for strong Tfh and GC responses to
mRNA vaccines or if other methods might be able to Human versus animal studies
achieve a similar outcome. Less clear is what impact A major hurdle in bringing mRNA vaccines to the clinic is
nucleoside modification has on cytotoxic CD8+ T cell to translate the often remarkable immunogenicity seen in
responses. Unmodified non-replicating mRNA, on the preclinical studies to human vaccine trials. This has been
other hand, has been shown to generate robust CD8+ illustrated by two mRNA vaccines made by different
T cell responses against tumor antigens, in which case methods: a rabies vaccine made using unmodified mRNA
antibody responses are generally not relevant to measure with a protamine/mRNA adjuvant [57,58], and two influ-
[46,52]. Self-amplifying mRNA vaccines, meanwhile, enza vaccines made with nucleoside-modified mRNA-
appear to stimulate balanced T and B cell responses LNPs encoding hemagglutinin from H7N9 or H10N8
[53,54], but there is limited information about the quality viruses [29,42]. In both cases, the preclinical data were

Figure 2

(a) Input variables (b) Output variables

modified vs. unmodified


ribonucleosides Cytokine and
chemokine milieu
delivery
materials B cell activation

ssRNA
B cell help

dsRNA

Differentiation signals
sequence
(Th1,Tfh,etc.)
engineering, UTRs
5’ 3’

CD8+T
cell help
Species differences in
immune sensing(?)

Current Opinion in Immunology

Representation of unknown factors in mRNA vaccine design. Shown are (a) a variety of potential features in mRNA vaccines that may influence
the immune response, and (b) a number of relevant immune pathways that might be affected. A transfected antigen-presenting cell is depicted as
an important mediator of many of these effects. Abbreviations: APC, antigen-presenting cell; ssRNA, single-stranded RNA; dsRNA, double-
stranded RNA; UTRs, untranslated regions; Th1, type 1 helper cell; Tfh, T follicular helper cell.

www.sciencedirect.com Current Opinion in Immunology 2020, 65:14–20


18 Vaccines

extremely promising in both mice and larger animals (pigs material. Further research into all of these areas will
for rabies and ferrets and cynomolgus macaques for contribute both to the further improvement of mRNA
influenza), and two doses of vaccine were sufficient to vaccines and to our basic understanding of immunology.
stimulate strong and sustained titers of neutralizing anti-
bodies. In contrast, when the same vaccines were tested Conclusions
in human volunteers, two doses of vaccine stimulated The past several years yielded critically important
unexpectedly low neutralizing antibody titers and sero- advancements in the field of mRNA vaccines and pro-
conversion frequencies. The discrepancies between the vided evidence for the viability of this novel vaccine
animal models and human studies prompt a number of modality. New manufacturing methods and delivery
important questions: are mRNA vaccines or their adju- materials will facilitate the rapid, inexpensive mass pro-
vants sensed or translated differently in humans versus duction of next-generation mRNA vaccines. Data from
other animals? Are there different biological require- human trials for both cancer and infectious disease
ments for potent antibody responses in humans compared mRNA vaccines are encouraging, but further improve-
to these animals? And does pre-existing immunity, for ments of the delivery materials and a more complete
example, against prior influenza virus exposures in understanding of the mechanisms of action of various
humans, impact the immunogenicity of a nucleoside- mRNA vaccine types are needed to rationally manipulate
modified mRNA-LNP vaccine? Animal models for influ- these formulations in order to increase efficacy while
enza virus vaccine studies may potentially be improved minimizing adverse events after vaccine administration.
by including a prior inoculation with live and/or inacti-
vated virus to better recapitulate the immune landscape
Conflict of interest statement
in human adults receiving an mRNA vaccine. Future
In accordance with the University of Pennsylvania poli-
studies should also examine the issue of ‘original anti-
cies and procedures and our ethical obligations as
genic sin;’ in this phenomenon, sequential exposure to
researchers, we report that Drew Weissman is named
divergent influenza virus antigens tends to boost antibody
on patents that describe the use of nucleoside-modified
responses that preferentially recognize the first antigen, at
mRNA as a platform to deliver therapeutic proteins.
the expense of those that recognize the second with high
Drew Weissman and Norbert Pardi are also named on
affinity [59].
a patent describing the use of modified mRNA in lipid
nanoparticles as a vaccine platform. We have disclosed
Mechanisms of immunogenicity of mRNA vaccines
those interests fully to the University of Pennsylvania,
Given the many variables in mRNA vaccine formulation
and we have in place an approved plan for managing any
outlined above and shown on Figure 2, it will be impor-
potential conflicts arising from licensing of our patents.
tant to determine which features promote protective
immunity so these vaccines may be improved in the
future. One major outstanding question in mRNA vac- Acknowledgements
N.P. was supported by the National Institute of Allergy and Infectious
cine design is whether type I IFNs enhance or inhibit Diseases (1R01AI146101). D.W. was supported by the National Institute of
protective immunity; in fact, there is good evidence for Allergy and Infectious Diseases (R01-AI050484, R01-AI124429 and R01-
both. A detrimental effect of type I IFN has been AI084860). M.J.H. is a Cancer Research Institute Irvington Fellow
supported by the Cancer Research Institute. The authors apologize to all
explicitly demonstrated in the context of self-amplifying colleagues whose great studies could not be cited here owing to space
mRNA vaccines. Pepini et al. showed that blocking the limitations.
activity of the type I IFN receptor improved both the
expression of mRNA-encoded protein as well as antigen- References and recommended reading
specific antibody and CD8+ T cell responses [49]. In Papers of particular interest, published within the period of review,
have been highlighted as:
contrast, a report by Kranz et al. showed that type I
IFN signaling was required for optimal antitumor immu-  of special interest
nity induced by an unmodified mRNA-LNP vaccine [46].  of outstanding interest
Taken together, it seems likely that type I IFNs may be
1. Suschak JJ, Williams JA, Schmaljohn CS: Advancements in DNA
detrimental for some types of mRNA vaccine and useful vaccine vectors, non-mechanical delivery methods, and
for others. Our current knowledge about the topic is molecular adjuvants to increase immunogenicity. Hum Vaccine
Immunother 2017, 13:2837-2848.
discussed in detail in a recent publication [50].
2. Pardi N, Muramatsu H, Weissman D, Kariko K: In vitro
transcription of long RNA containing modified nucleosides.
Other mechanisms that may influence the B or T cell Methods Mol Biol 2013, 969:29-42.
response against all mRNA vaccines include the half-life 3. Weissman D, Pardi N, Muramatsu H, Kariko K: HPLC purification
of antigen availability, the kinetics or magnitude of anti- of in vitro transcribed long RNA. Methods Mol Biol 2013, 969:43-
gen presentation on MHC class I and II, contributions 54.
from other components of the innate immune system 4. Kariko K, Buckstein M, Ni H, Weissman D: Suppression of RNA
recognition by Toll-like receptors: the impact of nucleoside
(NK cells, neutrophils, macrophages, etc.), and the cyto- modification and the evolutionary origin of RNA. Immunity
kine milieu induced by the mRNA and/or the delivery 2005, 23:165-175.

Current Opinion in Immunology 2020, 65:14–20 www.sciencedirect.com


Recent advances in mRNA vaccine technology Pardi, Hogan and Weissman 19

5. Kariko K, Muramatsu H, Welsh FA, Ludwig J, Kato H, Akira S, 21. Zhao M, Li M, Zhang Z, Gong T, Sun X: Induction of HIV-1 gag
Weissman D: Incorporation of pseudouridine into mRNA yields specific immune responses by cationic micelles mediated
superior nonimmunogenic vector with increased translational delivery of gag mRNA. Drug Deliv 2016, 23:2596-2607.
capacity and biological stability. Mol Ther 2008, 16:1833-1840.
22. Haabeth OAW, Blake TR, McKinlay CJ, Waymouth RM,
6. Thess A, Grund S, Mui BL, Hope MJ, Baumhof P, Fotin-Mleczek M, Wender PA, Levy R: mRNA vaccination with charge-altering
Schlake T: Sequence-engineered mRNA without chemical releasable transporters elicits human T cell responses and
nucleoside modifications enables an effective protein therapy cures established tumors in mice. Proc Natl Acad Sci U S A
in large animals. Mol Ther 2015, 23:1456-1464. 2018, 115:E9153-E9161.
7. Kariko K, Muramatsu H, Ludwig J, Weissman D: Generating the 23. McKinlay CJ, Benner NL, Haabeth OA, Waymouth RM,
optimal mRNA for therapy: HPLC purification eliminates Wender PA: Enhanced mRNA delivery into lymphocytes
immune activation and improves translation of nucleoside- enabled by lipid-varied libraries of charge-altering releasable
modified, protein-encoding mRNA. Nucleic Acids Res 2011, 39: transporters. Proc Natl Acad Sci U S A 2018, 115:E5859-E5866.
e142.
24. Chahal JS, Khan OF, Cooper CL, McPartlan JS, Tsosie JK,
8. Sahin U, Kariko K, Tureci O: mRNA-based therapeutics — Tilley LD, Sidik SM, Lourido S, Langer R, Bavari S et al.:
developing a new class of drugs. Nat Rev Drug Discov 2014, Dendrimer-RNA nanoparticles generate protective immunity
13:759-780. against lethal Ebola, H1N1 influenza, and Toxoplasma gondii
challenges with a single dose. Proc Natl Acad Sci U S A 2016,
9. Pardi N, Hogan MJ, Porter FW, Weissman D: mRNA vaccines - a 113:E4133-4142.
new era in vaccinology. Nat Rev Drug Discov 2018, 17:261-279.
25. Udhayakumar VK, De Beuckelaer A, McCaffrey J, McCrudden CM,
10. Kowalski PS, Rudra A, Miao L, Anderson DG: Delivering the Kirschman JL, Vanover D, Van Hoecke L, Roose K, Deswarte K, De
messenger: advances in technologies for therapeutic mRNA Geest BG et al.: Arginine-rich peptide-based mRNA
delivery. Mol Ther 2019, 27:710-728. nanocomplexes efficiently instigate cytotoxic T cell immunity
dependent on the amphipathic organization of the peptide.
11. Asrani KH, Farelli JD, Stahley MR, Miller RL, Cheng CJ, Adv Healthc Mater 2017, 6.
Subramanian RR, Brown JM: Optimization of mRNA
untranslated regions for improved expression of therapeutic 26. Semple SC, Akinc A, Chen J, Sandhu AP, Mui BL, Cho CK,
mRNA. RNA Biol 2018, 15:756-762. Sah DW, Stebbing D, Crosley EJ, Yaworski E et al.: Rational
design of cationic lipids for siRNA delivery. Nat Biotechnol
12. Sample PJ, Wang B, Reid DW, Presnyak V, McFadyen IJ, 2010, 28:172-176.
Morris DR, Seelig G: Human 5’ UTR design and variant effect
prediction from a massively parallel translation assay. Nat 27. Pardi N, Tuyishime S, Muramatsu H, Kariko K, Mui BL, Tam YK,
Biotechnol 2019, 37:803-809. Madden TD, Hope MJ, Weissman D: Expression kinetics of
nucleoside-modified mRNA delivered in lipid nanoparticles to
13. Trepotec Z, Aneja MK, Geiger J, Hasenpusch G, Plank C, mice by various routes. J Control Release 2015, 217:345-351.
Rudolph C: Maximizing the translational yield of mRNA
therapeutics by minimizing 5’-UTRs. Tissue Eng Part A 2019, 28. Awasthi S, Hook LM, Pardi N, Wang F, Myles A, Cancro MP,
25:69-79. Cohen GH, Weissman D, Friedman HM: Nucleoside-modified
mRNA encoding HSV-2 glycoproteins C, D, and E prevents
14. Orlandini von Niessen AG, Poleganov MA, Rechner C, Plaschke A, clinical and subclinical genital herpes. Sci Immunol 2019, 4.
Kranz LM, Fesser S, Diken M, Lower M, Vallazza B, Beissert T
et al.: Improving mRNA-based therapeutic gene delivery by 29. Bahl K, Senn JJ, Yuzhakov O, Bulychev A, Brito LA, Hassett KJ,
expression-augmenting 3’ UTRs identified by cellular library Laska ME, Smith M, Almarsson O, Thompson J et al.: Preclinical
screening. Mol Ther 2019, 27:824-836. and clinical demonstration of immunogenicity by mRNA
vaccines against H10N8 and H7N9 influenza viruses. Mol Ther
15. Beissert T, Perkovic M, Vogel A, Erbar S, Walzer KC, Hempel T, 2017, 25:1316-1327.
Brill S, Haefner E, Becker R, Tureci O et al.: A trans-amplifying
RNA vaccine strategy for induction of potent protective 30. Jagger BW, Dowd KA, Chen RE, Desai P, Foreman B,
immunity. Mol Ther 2020, 28:119-128. Burgomaster KE, Himansu S, Kong WP, Graham BS, Pierson TC
et al.: Protective efficacy of nucleic acid vaccines against
16. Blakney AK, McKay PF, Shattock RJ: Structural components for transmission of Zika virus during pregnancy in mice. J Infect
amplification of positive and negative strand VEEV splitzicons. Dis 2019, 220:1577-1588.
Front Mol Biosci 2018, 5:71.
31. John S, Yuzhakov O, Woods A, Deterling J, Hassett K, Shaw CA,
17. Vaidyanathan S, Azizian KT, Haque A, Henderson JM, Hendel A, Ciaramella G: Multi-antigenic human cytomegalovirus mRNA
 Shore S, Antony JS, Hogrefe RI, Kormann MSD, Porteus MH et al.: vaccines that elicit potent humoral and cell-mediated
Uridine depletion and chemical modification increase Cas9 immunity. Vaccine 2018, 36:1689-1699.
mRNA activity and reduce immunogenicity without HPLC
purification. Mol Ther Nucleic Acids 2018, 12:530-542. 32. Lutz J, Lazzaro S, Habbeddine M, Schmidt KE, Baumhof P,
This study reports on CleanCap1, a natural cap1 structure that can be Mui BL, Tam YK, Madden TD, Hope MJ, Heidenreich R et al.:
added to the mRNA during the IVT reaction, thus reducing the number of Unmodified mRNA in LNPs constitutes a competitive
steps of mRNA production. technology for prophylactic vaccines. NPJ Vaccines 2017, 2:29.
18. Devarkar SC, Wang C, Miller MT, Ramanathan A, Jiang F, 33. Meyer M, Huang E, Yuzhakov O, Ramanathan P, Ciaramella G,
Khan AG, Patel SS, Marcotrigiano J: Structural basis for m7G Bukreyev A: Modified mRNA-based vaccines elicit robust
recognition and 2’-O-methyl discrimination in capped RNAs immune responses and protect guinea pigs from Ebola virus
by the innate immune receptor RIG-I. Proc Natl Acad Sci U S A disease. J Infect Dis 2018, 217:451-455.
2016, 113:596-601.
34. Pardi N, Hogan MJ, Naradikian MS, Parkhouse K, Cain DW,
19. Schuberth-Wagner C, Ludwig J, Bruder AK, Herzner AM, Jones L, Moody MA, Verkerke HP, Myles A, Willis E et al.:
Zillinger T, Goldeck M, Schmidt T, Schmid-Burgk JL, Kerber R, Nucleoside-modified mRNA vaccines induce potent T
Wolter S et al.: A conserved histidine in the RNA sensor RIG-I follicular helper and germinal center B cell responses. J Exp
controls immune tolerance to N1-2’O-methylated self RNA. Med 2018, 215:1571-1588.
Immunity 2015, 43:41-51.
35. Pardi N, Hogan MJ, Pelc RS, Muramatsu H, Andersen H,
20. Baiersdorfer M, Boros G, Muramatsu H, Mahiny A, Vlatkovic I,  DeMaso CR, Dowd KA, Sutherland LL, Scearce RM, Parks R et al.:
 Sahin U, Kariko K: A facile method for the removal of dsRNA Zika virus protection by a single low-dose nucleoside-
contaminant from in vitro-transcribed mRNA. Mol Ther Nucleic modified mRNA vaccination. Nature 2017, 543:248-251.
Acids 2019, 15:26-35. This is the first published study that demonstrates the outstanding
This study reports on a simple, inexpensive, scalable mRNA purification immunogenicity and protective efficacy of nucleoside-modified mRNA-
protocol. LNP vaccines.

www.sciencedirect.com Current Opinion in Immunology 2020, 65:14–20


20 Vaccines

36. Pardi N, Parkhouse K, Kirkpatrick E, McMahon M, Zost SJ, Mui BL, nanoparticles encoding shared tumor antigens for potent
Tam YK, Kariko K, Barbosa CJ, Madden TD et al.: Nucleoside- melanoma immunotherapy. Ann Oncol 2017, 28.
modified mRNA immunization elicits influenza virus
hemagglutinin stalk-specific antibodies. Nat Commun 2018, 48. Kloke BP, Kreiter S, Vormehr M, Diken M, Kuhn AN, Sahin U:
9:3361. Actively personalized cancer vaccines–the step into clinical
application. Pharmazie 2016, 71:43-47.
37. Richner JM, Himansu S, Dowd KA, Butler SL, Salazar V, Fox JM,
Julander JG, Tang WW, Shresta S, Pierson TC et al.: Modified 49. Pepini T, Pulichino AM, Carsillo T, Carlson AL, Sari-Sarraf F,
mRNA vaccines protect against Zika virus infection. Cell 2017, Ramsauer K, Debasitis JC, Maruggi G, Otten GR, Geall AJ et al.:
168:1114-1125 e1110. Induction of an IFN-mediated antiviral response by a self-
amplifying RNA vaccine: implications for vaccine design. J
38. Richner JM, Jagger BW, Shan C, Fontes CR, Dowd KA, Cao B, Immunol 2017, 198:4012-4024.
Himansu S, Caine EA, Nunes BTD, Medeiros DBA et al.: Vaccine
mediated protection against Zika virus-induced congenital 50. Verbeke R, Lentacker I, De Smedt SC, Dewitte H: Three decades
disease. Cell 2017, 170:273-283 e212. of messenger RNA vaccine development. Nano Today 2019, 28.

39. Roth C, Cantaert T, Colas C, Prot M, Casademont I, Levillayer L, 51. Lindgren G, Ols S, Liang F, Thompson EA, Lin A, Hellgren F, Bahl K,
Thalmensi J, Langlade-Demoyen P, Gerke C, Bahl K et al.: A John S, Yuzhakov O, Hassett KJ et al.: Induction of robust B cell
modified mRNA vaccine targeting immunodominant NS responses after influenza mRNA vaccination is accompanied
epitopes protects against dengue virus infection in HLA class I by circulating hemagglutinin-specific ICOS+ PD-1+ CXCR3+ T
transgenic mice. Front Immunol 2019, 10:1424. follicular helper cells. Front Immunol 2017, 8:1539.
52. Kreiter S, Vormehr M, van de Roemer N, Diken M, Lower M,
40. VanBlargan LA, Himansu S, Foreman BM, Ebel GD, Pierson TC,
Diekmann J, Boegel S, Schrors B, Vascotto F, Castle JC et al.:
Diamond MS: An mRNA vaccine protects mice against multiple
Mutant MHC class II epitopes drive therapeutic immune
tick-transmitted Flavivirus infections. Cell Rep 2018, 25:3382-
responses to cancer. Nature 2015, 520:692-696.
3392 e3383.
53. Brazzoli M, Magini D, Bonci A, Buccato S, Giovani C, Kratzer R,
41. Hewitt SL, Bai A, Bailey D, Ichikawa K, Zielinski J, Karp R, Apte A, Zurli V, Mangiavacchi S, Casini D, Brito LM et al.: Induction of
Arnold K, Zacharek SJ, Iliou MS et al.: Durable anticancer broad-based immunity and protective efficacy by self-
immunity from intratumoral administration of IL-23, IL- amplifying mRNA vaccines encoding influenza virus
36gamma, and OX40L mRNAs. Sci Transl Med 2019, 11. hemagglutinin. J Virol 2016, 90:332-344.
42. Feldman RA, Fuhr R, Smolenov I, Mick Ribeiro A, Panther L, 54. Geall AJ, Verma A, Otten GR, Shaw CA, Hekele A, Banerjee K,
Watson M, Senn JJ, Smith M, Almarsson Ӧ, Pujar HS et al.: mRNA Cu Y, Beard CW, Brito LA, Krucker T et al.: Nonviral delivery of
vaccines against H10N8 and H7N9 influenza viruses of self-amplifying RNA vaccines. Proc Natl Acad Sci U S A 2012,
pandemic potential are immunogenic and well tolerated in 109:14604-14609.
healthy adults in phase 1 randomized clinical trials. Vaccine
2019, 37:3326-3334. 55. Baeza Garcia A, Siu E, Sun T, Exler V, Brito L, Hekele A, Otten G,
Augustijn K, Janse CJ, Ulmer JB et al.: Neutralization of the
43. Chen WH, Jackson LA, Edwards KM, Keitel WA, Hill H, Noah DL, Plasmodium-encoded MIF ortholog confers protective
Creech CB, Patel SM, Mangal B, Kotloff KL: Safety, immunity against malaria infection. Nat Commun 2018, 9:2714.
reactogenicity, and immunogenicity of inactivated
monovalent influenza A(H5N1) virus vaccine administered with 56. Kauffman KJ, Mir FF, Jhunjhunwala S, Kaczmarek JC, Hurtado JE,
or without AS03 adjuvant. Open Forum Infect Dis 2014, 1:ofu091. Yang JH, Webber MJ, Kowalski PS, Heartlein MW, DeRosa F et al.:
Efficacy and immunogenicity of unmodified and
44. Liang F, Lindgren G, Lin A, Thompson EA, Ols S, Rohss J, John S, pseudouridine-modified mRNA delivered systemically with
Hassett K, Yuzhakov O, Bahl K et al.: Efficient targeting and lipid nanoparticles in vivo. Biomaterials 2016, 109:78-87.
activation of antigen-presenting cells in vivo after modified
mRNA vaccine administration in rhesus macaques. Mol Ther 57. Alberer M, Gnad-Vogt U, Hong HS, Mehr KT, Backert L, Finak G,
2017, 25:2635-2647. Gottardo R, Bica MA, Garofano A, Koch SD et al.: Safety and
immunogenicity of a mRNA rabies vaccine in healthy adults:
45. Veiga N, Goldsmith M, Granot Y, Rosenblum D, Dammes N, an open-label, non-randomised, prospective, first-in-human
Kedmi R, Ramishetti S, Peer D: Cell specific delivery of modified phase 1 clinical trial. Lancet 2017, 390:1511-1520.
mRNA expressing therapeutic proteins to leukocytes. Nat
Commun 2018, 9:4493. 58. Schnee M, Vogel AB, Voss D, Petsch B, Baumhof P, Kramps T,
Stitz L: An mRNA vaccine encoding rabies virus glycoprotein
46. Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, induces protection against lethal infection in mice and
Meng M, Fritz D, Vascotto F, Hefesha H et al.: Systemic RNA correlates of protection in adult and newborn pigs. PLoS Negl
delivery to dendritic cells exploits antiviral defence for cancer Trop Dis 2016, 10:e0004746.
immunotherapy. Nature 2016, 534:396-401.
59. Linderman SL, Hensley SE: Antibodies with ‘Original Antigenic
47. Heesen L, Jabulowsky R, Loquai C, Utikal J, Gebhardt C, Hassel J, Sin’ properties are valuable components of secondary
Kaufmann R, Pinter A, Derhovanessian E, Diken M et al.: A first-in- immune responses to influenza viruses. PLoS Pathog 2016, 12:
human phase I/II clinical trial assessing novel mRNA-lipoplex e1005806.

Current Opinion in Immunology 2020, 65:14–20 www.sciencedirect.com

View publication stats

Вам также может понравиться