You are on page 1of 5

Vaccine 30 (2012) 4414–4418

Contents lists available at SciVerse ScienceDirect

Vaccine
journal homepage: www.elsevier.com/locate/vaccine

Review

RNA-based vaccines
Jeffrey B. Ulmer ∗ , Peter W. Mason, Andrew Geall, Christian W. Mandl
Novartis Vaccines, Cambridge, MA 02139, United States

a r t i c l e i n f o a b s t r a c t

Article history: Nucleic acid vaccines consisting of plasmid DNA, viral vectors or RNA may change the way the next
Received 3 February 2012 generation vaccines are produced, as they have the potential to combine the benefits of live-attenuated
Received in revised form 10 April 2012 vaccines, without the complications often associated with live-attenuated vaccine safety and manufac-
Accepted 18 April 2012
turing. Over the past two decades, numerous clinical trials of plasmid DNA and viral vector-based vaccines
Available online 28 April 2012
have shown them to be safe, well-tolerated and immunogenic. Yet, sufficient potency for general utility
in humans has remained elusive for DNA vaccines and the feasibility of repeated use of viral vectors
Keywords:
has been compromised by anti-vector immunity. RNA vaccines, including those based on mRNA and
Nucleic acid vaccine
Viral vector
self-amplifying RNA replicons, have the potential to overcome the limitations of plasmid DNA and viral
vectors. Possible drawbacks related to the cost and feasibility of manufacturing RNA vaccines are being
addressed, increasing the likelihood that RNA-based vaccines will be commercially viable. Proof of con-
cept for RNA vaccines has been demonstrated in humans and the prospects for further development into
commercial products are very encouraging.
© 2012 Elsevier Ltd. All rights reserved.

Contents

1. Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4414
2. Nucleic acid vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4414
2.1. DNA vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4415
2.2. Viral vectors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4415
2.3. RNA vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4415
3. Prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4417
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4417

1. Background immunization with a live organism vaccine, particularly for induc-


tion of T cell immunity [2]. In addition, the manufacture of nucleic
The discovery and development of new and improved vaccines acid-based vaccines offered the potential to be relatively simple,
have been greatly facilitated by the application of new technolo- inexpensive and generic. Since then, clinical trials have amply
gies to identify protective antigens, to optimally present antigens demonstrated the safety and tolerability of nucleic acid vaccines
to the immune system, and to manufacture vaccines using highly [3], and robust manufacturing processes have been developed [4].
characterized, synthetic methods of production. This progress has However, potency in humans has been disappointing, which has
been spurred by a need to move beyond empirical approaches to led to extensive activity to identify enabling technologies. The main
vaccines research and development, and has ushered in several areas for improvement have been directed toward the nucleic acid
new paradigms including reverse, structural and synthetic vacci- vector, targeting the innate immune system to enhance immuno-
nology approaches, respectively [1]. The use of nucleic acid-based genicity, and delivery systems to overcome the barriers to efficient
vaccines to combine the benefits of in situ expression of antigens, transfection of host cells in vivo. Significant progress has been made
with the safety of inactivated and subunit vaccines, has been a key on all these fronts. This review paper will focus on the use of an
advancement. Upon their discovery more than 20 years ago, nucleic alternative nucleic acid vector, namely RNA, as the basis of a new
acid vaccines promised to be a safe and effective means to mimic generation of vaccines.

2. Nucleic acid vaccines


∗ Corresponding author at: Novartis Vaccines, 350 Massachusetts Ave., Cam-
bridge, MA 02139, United States. By definition, nucleic acid vaccines are based on DNA or RNA
E-mail address: jeffrey.ulmer@novartis.com (J.B. Ulmer). encoding the antigen(s) of interest. In their simplest form, they can

0264-410X/$ – see front matter © 2012 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.vaccine.2012.04.060
J.B. Ulmer et al. / Vaccine 30 (2012) 4414–4418 4415

consist of highly purified nucleic acids formulated in a buffer. Most issue. Second, because viral vectors contain, and in some cases
often, however, specialized delivery systems are utilized to increase express, viral antigens in addition to the target antigen of inter-
vaccine potency. Means to facilitate nucleic acid delivery involve est, such vectors are usually quite immunogenic (i.e., elicit immune
(1) viral particles to take advantage of the efficiency of viral entry responses against the vectors themselves). Pre-existing anti-vector
mechanisms, (2) non-viral formulations of DNA or RNA involving immunity (either due to prior infection with wild-type virus, vac-
lipids, polymers, emulsions or other synthetic approaches to avoid cines or immunization with the vector) has been shown to blunt
the use of viral vectors, and (3) physical delivery technologies, such the ability of the vector to launch production of the target anti-
as electroporation in situ. The majority of the preclinical and clinical gen and, hence, limits induction of immune responses against the
experiences with nucleic acid vaccines so far have been with DNA antigen of interest. Strategies to circumvent this limitation have
vaccines and DNA-based viral vectors. included use of certain adenovirus strains not commonly circulat-
ing in humans, to allow initial take of the viral vector vaccine, and
2.1. DNA vaccines heterologous prime-boost approaches involving different vectors,
to allow effective boosting of immune responses against the target
DNA vaccines have been widely evaluated in many animal mod- antigen. While these approaches can be effective, at least temporar-
els of infectious and non-infectious diseases with generally good ily, they complicate the vaccination regimen and do not provide an
success at eliciting potent immune responses against encoded anti- optimal solution.
gens, which have ranged from discrete T or B cell epitopes to large
polyprotein complexes. The utility of DNA vaccines in animals has 2.3. RNA vaccines
been further documented by the development and commercializa-
tion of plasmid DNA-based animal health products. These include Proof of concept for RNA vaccines was provided two decades
a West Nile virus vaccine for horses [5], an infectious hematopoi- ago, when intramuscular injection of mRNA in mice resulted in
etic necrosis virus vaccine for fish [6], a melanoma cancer vaccine local production of an encoded reporter protein [15] and induc-
for dogs [7], and a growth hormone releasing hormone gene ther- tion of immune responses against an encoded antigen [16]. In
apy for pigs [8]. In humans, proof of concept for induction of both a direct comparison with a corresponding plasmid DNA vaccine,
antibody and T cell responses has been demonstrated for various injection of similar doses of mRNA (on a mass basis) formulated
indications in multiple clinical trials. However, the magnitude of in sucrose resulted in similar levels of reporter gene expression,
these immune responses has been lower than those observed for suggesting equivalent efficiencies of cellular transfection in vivo
conventional vaccines consisting of live or inactivated whole organ- by the two types of nucleic acid vaccines [15]. These initial pub-
isms, or subunit proteins formulated with adjuvants. The reasons lications were followed by many more demonstrating the general
for this shortcoming of DNA vaccines are not clear, but are likely utility of eliciting immune responses by RNA vaccines (for review,
due, at least in part, to inefficient delivery of DNA into human cells see [17]). The variety of gene targets included reporter genes
and inadequate stimulation of the human immune system. To over- [15,18–20] viral antigens [16,21], tumor antigens [22–26], and
come these limitations, various technologies have been evaluated allergens [27,28]. In these animal models, both antibody and T
and the most promising current approaches involve facilitation of cell responses, including CD4+ and CD8+ , were elicited. Further-
DNA delivery by electroporation [9] and stimulation of the immune more, functional immunity, as measured by protective efficacy
system via the use of genetic adjuvants (i.e., in situ expression of against challenge with live pathogens or tumors, was achieved. In
immunologically active molecules encoded by the DNA vaccine) preclinical models of allergy, low doses of an RNA vaccine encod-
[10]. Combinations of these approaches have resulted in potent ing allergens induce a Th1-biased immune response that provided
induction of immunity in non-human primates [11,12] and pre- resistance against subsequent allergic sensitization. Induction of
liminary results of human clinical trials are encouraging [3,9]. antigen-specific immune responses can be achieved by adminis-
tration of RNA vaccines via various routes, including intramuscular
2.2. Viral vectors [15,19], intradermal [22], subcutaneous [16], intravenous [16],
intrasplenic [21], and intranodal [24], as well as delivery into the
In situ expression of antigens in a vaccinated host can be effec- skin by the gene gun [29,30]. In addition, a considerable amount
tively achieved through the use of recombinant vectors, often DNA of work has been done using mRNA vaccines to pulse dendritic
viruses, engineered to be safe and to encode the gene(s) of interest. cells in vitro, which are then administered as the immunizing
Vectors based on adenoviruses and poxviruses have been studied agent (for review, see [31]). Hence, like DNA vaccines, RNA vaccines
extensively, although several other viral vectors are being evalu- have demonstrated versatility in many animal models of infectious
ated at earlier stages of development. Both adenovirus and poxvirus and non-infectious diseases. However, RNA vaccines have several
vectors have demonstrated safety and immunogenicity in human attributes that provide potential advantages over DNA vaccines.
clinical trials [13]. Notably, a poxvirus vector encoding HIV enve- First, there is a finite chance that plasmid DNA vaccines can inte-
lope protein used in a prime-boost regimen with recombinant grate into the genome of the immunized host. Although there has
envelope protein plus adjuvant elicited modest protection in a been little evidence so far that integration occurs after DNA vaccina-
phase III efficacy trial [14]. One clear advantage of viral vectors tion, use of RNA would eliminate this as an issue. Second, plasmid
over DNA vaccines is the efficiency with which the DNA payload DNA vaccines must be delivered into and transcribed within the
is introduced into host cells, due to the natural invasiveness of nucleus in order to transfect a cell, i.e. they must traverse two
the viral particle. Hence, the amount of plasmid DNA required membrane barriers (plasma and nuclear membranes). This could
for induction of immune responses is typically many orders of be particularly problematic in non-dividing cells, such as mature
magnitude greater than the amount of DNA contained in a viral myocytes, where the nuclear membrane remains intact. Several
vector vaccine. Two potential limitations of viral vectors, though, publications have demonstrated that microinjection of pDNA into
are related to safety and the inherent immunogenicity of the vec- the cytoplasm of non-dividing cells resulted in very low levels
tor itself. First, because viral vectors are usually originally derived of gene expression, but direct intra-nuclear injection of the same
from wild-type pathogenic viruses, there is at least a theoretical number of pDNA copies led to efficient transfection [32–34]. In con-
potential for reversion to a virulent state, just as there is for atten- trast, since RNA vaccines are translated directly in the cytoplasm,
uated live virus-based vaccines. However, extensive safety testing the need for delivery into the nucleus is obviated. Finally, the kinet-
of these vectors has demonstrated that this is likely not a major ics of antigen expression after RNA administration appears to peak
4416 J.B. Ulmer et al. / Vaccine 30 (2012) 4414–4418

Virus genome Sub-genomic promoter

m7G 5' nonstructural proteins Capsid, E2/E1 glycoproteins An

Sub-genomic promoter
Replicon vector
m7G 5' nonstructural proteins Antigen An

Fig. 1. Schematic illustration of an RNA replicon vaccine. Example of an RNA replicon vector derived from a positive-strand alphavirus genome. All replicons encode genes
(indicated here as nonstructural proteins) that drive their amplification within the cytoplasm of host cells. For use as vaccine vectors, replicons also encode antigen genes.
For alphavirus vectors, antigen genes are most commonly inserted in place of the capsid and glycoprotein genes, which are not needed for genome replication. In this way,
the vector amplifies its full-length genome when it is introduced into the cytoplasm, and then following genome amplification it initiates production of a sub-genomic mRNA
encoding the target antigen.

and decay rapidly, in contrast to DNA administration where antigen As an alternative delivery system, the gene gun has been used to
expression can persist for many weeks [35]. Hence, RNA vaccination directly introduce mRNA into the cytoplasm of cells [29]. Second,
better mimics antigen expression during an acute infection, which although RNA vaccines have a built-in adjuvant effect in the form
may be more conducive to induction of antigen-specific immune of TLR engagement, mRNA vaccine potency has been enhanced by
responses. coadministration of recombinant GM-CSF [19] or Flt-3 ligand [42],
The mechanisms of action for RNA vaccines have not been or RNA encoding GM-CSF [43]. Finally, several approaches have
fully elucidated, but likely involve some of the same processes been taken to improve the RNA molecule itself. Various modifi-
utilized by DNA vaccines for the expression and presentation of cations have been made to the 5 cap structure, the untranslated
encoded antigens leading to induction of immune responses. After regions, and codon usage in the translated region (for review,
injection, the RNA is exposed to RNases in the tissue [36], which see [44]), which have resulted in increased mRNA stability and
can degrade the vaccine and limit uptake of functional RNA by expression.
cells. In addition, the 2 -hydroyxl on the ribose sugar prevents the The feasibility of using RNA as the basis for a nucleic acid vaccine
mRNA adopting a stable double ␤-helix, due to steric hindrance, was initially regarded as questionable, due the inherent instabil-
and makes the macromolecule more prone to hydrolysis. Never- ity of mRNA in the presence of tissue fluids, the uncertainty of
theless, various cell types are capable of internalizing RNA by an developing reasonable manufacturing processes yielding a stable
active, saturable and specific process leading to local expression formulation, and the anticipated high cost of the product. Each of
of antigen [35]. Uptake is mediated by membrane domains rich these potential limitations is being addressed. Even naked mRNA
in caveolae and lipid rafts, and involves scavenger receptors [37]. is immunogenic in animals [19–21,25,45] and humans [46], indi-
Upon internalization, a portion of the RNA accumulates in the cyto- cating that RNA degradation in tissues after administration does
plasm where it is translated into protein. This in situ production not completely abrogate vaccine effectiveness. However, the effi-
of antigen provides a means to mimic pathogen infections and ciency of RNA delivery should be increased markedly through the
expression of tumor antigens leading to efficient presentation of use of enabling synthetic and viral delivery systems. For research
antigens by major histocompatibility complex (MHC) class I and II purposes, in vitro transcribed mRNA can be obtained from plas-
proteins, and induction of T cell responses in a manner analogous mid DNA containing a bacteriophage promoter (T7, SP6 or T3) and
to that provided by DNA vaccines and viral vectors. Alternatively, over the past 10 years many technical refinements to the com-
RNA vaccines can be constructed for the efficient production and mercial kits have resulted in dramatic improvements in quality
secretion (or cell-surface expression) of extracellular antigens to and yield [31,47]. More recently, RNA manufacturing by enzymatic
stimulate B cell responses and antigen-specific antibody produc- transcription of appropriate DNA templates now seems attainable
tion. The effectiveness of RNA vaccines may also be related to the at reasonable cost and large scale. Long-term storage of lyophilized
fact that RNA is known to be a potent stimulator of innate immu- RNA vaccines have previously been studied and RNAse-free RNA
nity. In vitro, mRNA has been shown to activate dendritic cells and vaccines were demonstrated to be no less stable than other con-
monocytes in a MyD88-dependent fashion involving signaling via ventional vaccines that require a cold chain to retain efficacy [48].
Toll-like receptors (TLR) [38,39]. In vivo, it was recently demon- These advancements have enabled the development of process for
strated that an mRNA vaccine caused the upregulation of various the GMP production of mRNA vaccines in quantities sufficient for
genes involved in chemotaxis and cell activation [40] as well as human clinical trials (for review, see [17]).
induction of TLR7-dependent CD4+ and CD8+ T cell responses, and While most of the published work has utilized mRNA as the
anti-tumor immunity [41]. Hence, the functionality of RNA vaccines vaccine, several publications have shown that RNA vaccines can
involves at least two components: (1) local expression of antigen also be derived from sub-genomic replicons that lack viral struc-
to facilitate presentation by MHC molecules and (2) engagement of tural proteins. Replicon RNA-based vaccines have been generated
pattern recognition receptors to stimulate innate immunity leading for a variety of RNA viruses including, Semliki Forest virus [21,25],
to potentiation of antigen-specific immune responses. Sindbis virus [20], poliovirus [49,50], tick-borne encephalitis virus
Many of the above-referenced studies have used naked mRNA [51,52], Kunjin virus [53], and bovine viral diarrhea [54]. RNA-based
as the vaccine (i.e., simply formulated in a buffer). While this vaccines have also been described in which the RNA vaccine is used
approach has been shown to elicit immune responses, the pres- to launch a live-attenuated virus infection. In this case, the inherent
ence of degradative enzymes in tissues likely limits the amount potency of the encoded live viral vaccine has permitted this type
of RNA that is available for internalization by cells in vivo. As a of RNA vaccine to elicit protective immunity at very low (ng) RNA
means to overcome this inherent drawback of using naked RNA, doses [51]. More commonly, experimental RNA-based vaccines are
work has focused on delivery systems, adjuvants, and engineering viral-particle delivered products engineered to express a heterol-
of the RNA molecule. First, to protect RNA from degradation and ogous antigen in place of the viral structural genes. These vaccines
enhance cellular uptake, encapsulation in liposomes [16,18,26] and are produced under special conditions (e.g., packaging cell lines)
complexation with cationic polymers [38,41] have proven effective. that permit production of single-round infectious particles carrying
J.B. Ulmer et al. / Vaccine 30 (2012) 4414–4418 4417

Table 1
Superior attributes of RNA vaccines.

Parameter Vaccine type

Live Subunit Viral vector DNA mRNA Replicon RNA

Synthetic − − − +/− + +
Generic manufacturing − − − + + +
Safety +/− + +/− + + +
Antibody induction + + + +/− + +
CTL induction + − + + + +
In vivo expression + − + + + +
Control of expression − − + + + +
Absence of eukaryotic contaminants − +/− − + + +
In vivo self amplification + − + − − +
Potency in humans + + +/− +/− +/− TBD

RNAs encoding the vaccine antigens [55–59]. In this way, transient, neutralizing antibodies. However, the magnitude of the responses
high levels of antigen production can be achieved without the use of in these recent trials was similar to those previously observed
a “live”, spreading viral infection. Replicons derived from different for other types of nucleic acid vaccines. Therefore, the RNA vac-
RNA viruses differ with regard to levels and duration of heterolo- cine approach holds promise as an effective means of eliciting
gous gene expression allowing the generation of a versatile toolbox functional, protective immune responses in humans, but success
for vaccine or gene therapy applications [60]. An illustration of an will likely require enabling delivery technologies. Next generation
RNA vaccine based on an alphavirus replicon is depicted in Fig. 1. replicon RNA vaccines will be formulated with synthetic delivery
The RNA amplification process in the cytoplasm produces multiple technologies and will aspire to combine the effectiveness of live
copies of antigen-encoding mRNA and creates dsRNA intermedi- attenuated vaccines, an equal or better safety profile than plasmid
ates, which are known to be potent stimulators of innate immunity DNA vaccines, and completely synthetic methods of manufacture.
[61]. Thus, on a mass basis, replicon RNA vaccines have the potential Such a vaccine would possess the desired attributes of an ideal
to be more effective than corresponding mRNA vaccines. Indeed, a vaccine.
direct comparison of the two types of RNA vaccines demonstrated
significantly higher and more persistent expression levels in vivo
after replicon RNA administration [20]. References
These replicon vaccines have been administered as naked RNA
[1] Rappuoli R, Mandl CW, Black S, De Gregorio E. Vaccines for the twenty-first
packaged in viral particles, or delivered by electroporation in situ century society. Nature Reviews Immunology 2011;11(12):865–72.
[52,62,63] Viral particle delivery of replicons has the advantage of [2] Ulmer JB, Donnelly JJ, Parker SE, Rhodes GH, Felgner PL, Dwarki VJ, et al. Het-
erologous protection against influenza by injection of DNA encoding a viral
efficiency, as previously described for viral vectors, but complicates
protein. Science 1993;259(5102):1745–9.
manufacturing, introduces theoretical safety considerations, and [3] Ferraro B, Morrow MP, Hutnick NA, Shin TH, Lucke CE, Weiner DB. Clinical
has the potential limitation of anti-vector immunity. Hence, facil- applications of DNA vaccines: current progress. Clinical Infectious Diseases
itated delivery of RNA replicons using synthetic systems, such as 2011;53(3):296–302.
[4] Lara AR, Ramirez OT. Plasmid DNA production for therapeutic applications.
those evaluated for mRNA or DNA vaccines may increase potency Methods in Molecular Biology 2012;824:271–303.
without the added complications commonly seen with viral vec- [5] Davis BS, Chang GJ, Cropp B, Roehrig JT, Martin DA, Mitchell CJ, et al. West
tors. Nile virus recombinant DNA vaccine protects mouse and horse from virus
challenge and expresses in vitro a noninfectious recombinant antigen that
can be used in enzyme-linked immunosorbent assays. Journal of Virology
2001;75(9):4040–7.
3. Prospects
[6] Garver KA, LaPatra SE, Kurath G. Efficacy of an infectious hematopoietic necrosis
(IHN) virus DNA vaccine in Chinook Oncorhynchus tshawytscha and sockeye O.
RNA vaccines, particularly self-amplifying replicons, have the nerka salmon. Diseases of Aquatic Organisms 2005;64(1):13–22.
[7] Grosenbaugh DA, Leard AT, Bergman PJ, Klein MK, Meleo K, Susaneck S, et al.
potential to capture the advantages of both DNA vaccines and
Safety and efficacy of a xenogeneic DNA vaccine encoding for human tyrosinase
viral delivery while overcoming the drawbacks of each technology as adjunctive treatment for oral malignant melanoma in dogs following sur-
(see Table 1). The prospect of RNA vaccines being a more effec- gical excision of the primary tumor. American Journal of Veterinary Research
tive approach than other types of nucleic acid vaccines has led to 2011;72(12):1631–8.
[8] Draghia-Akli R, Li X, Schwartz RJ. Enhanced growth by ectopic expression
their advancement into human clinical trials. So far, mRNA vac- of growth hormone releasing hormone using an injectable myogenic vector.
cines have been administered to cancer patients in several trials as Nature Biotechnology 1997;15(12):1285–9.
active immunotherapeutic immunization protocols, supported by [9] Sardesai NY, Weiner DB. Electroporation delivery of DNA vaccines: prospects
for success. Current Opinion in Immunology 2011;23(3):421–9.
preclinical proof of concept in animal tumor models (for review, see [10] Lori F, Weiner DB, Calarota SA, Kelly LM, Lisziewicz J. Cytokine-adjuvanted
[31]). In the first trial, mRNA encoding genes cloned from metastatic HIV-DNA vaccination strategies. Springer Seminars in Immunopathology
melanoma tumors were used as an autologous immunization strat- 2006;28(3):231–8.
[11] Belisle SE, Yin J, Shedlock DJ, Dai A, Yan J, Hirao L, et al. Long-term program-
egy [46]. Subsequent trials used combinations of known tumor ming of antigen-specific immunity from gene expression signatures in the
antigens, such as MUC1, CEA, telomerase, MAGE-1, tyrosinase, in PBMC of rhesus macaques immunized with an SIV DNA vaccine. PLoS One
metastatic melanoma [64] and renal cell carcinoma [65] patients. 2011;6(6):e19681.
[12] Hirao LA, Hokey DA, Morrow MP, Jure-Kunkel MN, Weiner DB. Immune modu-
In these exploratory clinical trials, the mRNA vaccines elicited
lation through 4-1BB enhances SIV vaccine protection in non-human primates
antigen-specific immune responses (both antibodies and T cells), against SIVmac251 challenge. PLoS One 2011;6(9):e24250.
demonstrating proof of concept that mRNA vaccines are active in [13] Rollier CS, Reyes-Sandoval A, Cottingham MG, Ewer K, Hill AV. Viral vectors
as vaccine platforms: deployment in sight. Current Opinion in Immunology
humans. Clinical trials have also been performed with RNA replicon
2011;23(3):377–82.
vaccines packaged in viral particles. A bivalent vaccine consisting [14] Vaccari M, Poonam P, Franchini G. Phase III HIV vaccine trial in Thailand:
of replicons encoding cytomegalovirus (CMV) gB and pp65/IE1 pro- a step toward a protective vaccine for HIV. Expert Review of Vaccines
teins was shown to be well tolerated and immunogenic in healthy 2010;9(9):997–1005.
[15] Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, et al. Direct
CMV seronegative volunteers [66]. All 40 individuals generated gene transfer into mouse muscle in vivo. Science 1990;247(4949 Pt 1):
polyfunctional CD4+ and CD8+ T cell responses, as well as virus 1465–8.
4418 J.B. Ulmer et al. / Vaccine 30 (2012) 4414–4418

[16] Martinon F, Krishnan S, Lenzen G, Magne R, Gomard E, Guillet JG, et al. Induction [43] Hess PR, Boczkowski D, Nair SK, Snyder D, Gilboa E. Vaccination with mRNAs
of virus-specific cytotoxic T lymphocytes in vivo by liposome-entrapped mRNA. encoding tumor-associated antigens and granulocyte-macrophage colony-
European Journal of Immunology 1993;23(7):1719–22. stimulating factor efficiently primes CTL responses, but is insufficient to
[17] Pascolo S. Messenger RNA-based vaccines. Expert Opinion on Biological Ther- overcome tolerance to a model tumor/self antigen. Cancer Immunology and
apy 2004;4(8):1285–94. Immunotherapy 2006;55(6):672–83.
[18] Hoerr I, Obst R, Rammensee HG, Jung G. In vivo application of RNA leads to [44] Pascolo S. Vaccination with messenger RNA (mRNA). Handbook of Experimen-
induction of specific cytotoxic T lymphocytes and antibodies. European Journal tal Pharmacology 2008;183:221–35.
of Immunology 2000;30(1):1–7. [45] Conry RM, LoBuglio AF, Loechel F, Moore SE, Sumerel LA, Barlow DL, et al. A car-
[19] Carralot JP, Probst J, Hoerr I, Scheel B, Teufel R, Jung G, et al. Polarization of cinoembryonic antigen polynucleotide vaccine for human clinical use. Cancer
immunity induced by direct injection of naked sequence-stabilized mRNA vac- Gene Therapy 1995;2(1):33–8.
cines. Cellular and Molecular Life Sciences 2004;61(18):2418–24. [46] Weide B, Carralot JP, Reese A, Scheel B, Eigentler TK, Hoerr I, et al. Results of the
[20] Johanning FW, Conry RM, LoBuglio AF, Wright M, Sumerel LA, Pike MJ, first phase I/II clinical vaccination trial with direct injection of mRNA. Journal
et al. A Sindbis virus mRNA polynucleotide vector achieves prolonged and of Immunotherapy 2008;31(2):180–8.
high level heterologous gene expression in vivo. Nucleic Acids Research [47] Yamamoto A, Kormann M, Rosenecker J, Rudolph C. Current prospects for
1995;23(9):1495–501. mRNA gene delivery. European Journal of Pharmaceutics and Biopharmaceutics
[21] Zhou X, Berglund P, Rhodes G, Parker SE, Jondal M, Liljestrom P. Self-replicating 2009;71(3):484–9.
Semliki Forest virus RNA as recombinant vaccine. Vaccine 1994;12(16):1510–4. [48] Jones KL, Drane D, Gowans EJ. Long-term storage of DNA-free RNA for use in
[22] Granstein RD, Ding W, Ozawa H. Induction of anti-tumor immunity with epi- vaccine studies. Biotechniques 2007;43(5):675–81.
dermal cells pulsed with tumor-derived RNA or intradermal administration of [49] Jackson CA, Messinger J, Palmer MT, Peduzzi JD, Morrow CD. Gene expression
RNA. Journal of Investigative Dermatology 2000;114(4):632–6. in the muscle and central nervous system following intramuscular inocu-
[23] Conry RM, LoBuglio AF, Wright M, Sumerel L, Pike MJ, Johanning F, et al. Charac- lation of encapsidated or naked poliovirus replicons. Virology 2003;314(1):
terization of a messenger RNA polynucleotide vaccine vector. Cancer Research 45–61.
1995;55(7):1397–400. [50] Vignuzzi M, Gerbaud S, van der Werf S, Escriou N. Naked RNA immu-
[24] Kreiter S, Selmi A, Diken M, Koslowski M, Britten CM, Huber C, et al. Intranodal nization with replicons derived from poliovirus and Semliki Forest virus
vaccination with naked antigen-encoding RNA elicits potent prophylactic and genomes for the generation of a cytotoxic T cell response against the
therapeutic antitumoral immunity. Cancer Research 2010;70(22):9031–40. influenza A virus nucleoprotein. Journal of General Virology 2001;82(Pt 7):
[25] Ying H, Zaks TZ, Wang RF, Irvine KR, Kammula US, Marincola FM, et al. 1737–47.
Cancer therapy using a self-replicating RNA vaccine. Nature Medicine [51] Mandl CW, Aberle JH, Aberle SW, Holzmann H, Allison SL, Heinz FX. In vitro-
1999;5(7):823–7. synthesized infectious RNA as an attenuated live vaccine in a flavivirus model.
[26] Zhou WZ, Hoon DS, Huang SK, Fujii S, Hashimoto K, Morishita R, et al. RNA Nature Medicine 1998;4(12):1438–40.
melanoma vaccine: induction of antitumor immunity by human glycoprotein [52] Kofler RM, Aberle JH, Aberle SW, Allison SL, Heinz FX, Mandl CW. Mimicking
100 mRNA immunization. Human Gene Therapy 1999;10(16):2719–24. live flavivirus immunization with a noninfectious RNA vaccine. Proceed-
[27] Roesler E, Weiss R, Weinberger EE, Fruehwirth A, Stoecklinger A, Mostbock ings of the National Academy of Sciences of the United States of America
S, et al. Immunize and disappear-safety-optimized mRNA vaccination with 2004;101(7):1951–6.
a panel of 29 allergens. The Journal of Allergy and Clinical Immunology [53] Harvey TJ, Anraku I, Linedale R, Harrich D, Mackenzie J, Suhrbier A, et al. Kunjin
2009;124(5), 1070-7 e1–11. virus replicon vectors for human immunodeficiency virus vaccine develop-
[28] Weiss R, Scheiblhofer S, Roesler E, Weinberger E, Thalhamer J. mRNA vaccina- ment. Journal of Virology 2003;77(14):7796–803.
tion as a safe approach for specific protection from type I allergy. Expert Review [54] Edlich B, Hogdal LJ, Rehermann B, Behrens SE. Dendritic cells transfected with
of Vaccines 2012;11(1):55–67. Her2 antigen-encoding RNA replicons cross-prime CD8 T cells and protect mice
[29] Qiu P, Ziegelhoffer P, Sun J, Yang NS. Gene gun delivery of mRNA in situ results against tumor challenge. Vaccine 2010;28(49):7764–73.
in efficient transgene expression and genetic immunization. Gene Therapy [55] Li HO, Zhu YF, Asakawa M, Kuma H, Hirata T, Ueda Y, et al. A cytoplasmic RNA
1996;3(3):262–8. vector derived from nontransmissible Sendai virus with efficient gene transfer
[30] Aberle JH, Aberle SW, Kofler RM, Mandl CW. Humoral and cellular immune and expression. Journal of Virology 2000;74(14):6564–9.
response to RNA immunization with flavivirus replicons derived from tick- [56] Kalhoro NH, Veits J, Rautenschlein S, Zimmer G. A recombinant vesicular stom-
borne encephalitis virus. Journal of Virology 2005;79(24):15107–13. atitis virus replicon vaccine protects chickens from highly pathogenic avian
[31] Kreiter S, Diken M, Selmi A, Tureci O, Sahin U. Tumor vaccination using mes- influenza virus (H7N1). Vaccine 2009;27(8):1174–83.
senger RNA: prospects of a future therapy. Current Opinion in Immunology [57] Widman DG, Frolov I, Mason PW. Third-generation flavivirus vaccines based
2011;23(3):399–406. on single-cycle, encapsidation-defective viruses. Advances in Virus Research
[32] Mirzayans R, Aubin RA, Paterson MC. Differential expression and stability of 2008;72:77–126.
foreign genes introduced into human fibroblasts by nuclear versus cytoplasmic [58] Perri S, Greer CE, Thudium K, Doe B, Legg H, Liu H, et al. An alphavirus repli-
microinjection. Mutation Research 1992;281(2):115–22. con particle chimera derived from venezuelan equine encephalitis and sindbis
[33] Thorburn AM, Alberts AS. Efficient expression of miniprep plasmid DNA after viruses is a potent gene-based vaccine delivery vector. Journal of Virology
needle micro-injection into somatic cells. Biotechniques 1993;14(3):356–8. 2003;77(19):10394–403.
[34] Zabner J, Fasbender AJ, Moninger T, Poellinger KA, Welsh MJ. Cellular and [59] Caley IJ, Betts MR, Irlbeck DM, Davis NL, Swanstrom R, Frelinger JA,
molecular barriers to gene transfer by a cationic lipid. Journal of Biological et al. Humoral, mucosal, and cellular immunity in response to a human
Chemistry 1995;270(32):18997–9007. immunodeficiency virus type 1 immunogen expressed by a Venezuelan
[35] Probst J, Weide B, Scheel B, Pichler BJ, Hoerr I, Rammensee HG, et al. Spon- equine encephalitis virus vaccine vector. Journal of Virology 1997;71(4):
taneous cellular uptake of exogenous messenger RNA in vivo is nucleic acid- 3031–8.
specific, saturable and ion dependent. Gene Therapy 2007;14(15):1175–80. [60] Gehrke R, Heinz FX, Davis NL, Mandl CW. Heterologous gene expression by
[36] Probst J, Brechtel S, Scheel B, Hoerr I, Jung G, Rammensee HG, et al. Charac- infectious and replicon vectors derived from tick-borne encephalitis virus and
terization of the ribonuclease activity on the skin surface. Genetics Vaccines direct comparison of this flavivirus system with an alphavirus replicon. Journal
Therapy 2006;4:4. of General Virology 2005;86(Pt 4):1045–53.
[37] Lorenz C, Fotin-Mleczek M, Roth G, Becker C, Dam TC, Verdurmen WP, [61] Wang Y, Swiecki M, McCartney SA, Colonna M. dsRNA sensors and plasmacy-
et al. Protein expression from exogenous mRNA: uptake by receptor- toid dendritic cells in host defense and autoimmunity. Immunological Review
mediated endocytosis and trafficking via the lysosomal pathway. RNA Biology 2011;243(1):74–90.
2011;8(4):627–36. [62] Piggott JM, Sheahan BJ, Soden DM, O‘Sullivan GC, Atkins GJ. Electroporation
[38] Scheel B, Teufel R, Probst J, Carralot JP, Geginat J, Radsak M, et al. Toll-like of RNA stimulates immunity to an encoded reporter gene in mice. Molecular
receptor-dependent activation of several human blood cell types by protamine- Medicine Report 2009;2(5):753–6.
condensed mRNA. European Journal of Immunology 2005;35(5):1557–66. [63] Johansson DX, Ljungberg K, Kakoulidou M, Liljestrom P. Intradermal electro-
[39] Kariko K, Ni H, Capodici J, Lamphier M, Weissman D. mRNA is an poration of naked replicon RNA elicits strong immune responses. PLoS One
endogenous ligand for Toll-like receptor 3. Journal of Biological Chemistry 2012;7(1):e29732.
2004;279(13):12542–50. [64] Weide B, Pascolo S, Scheel B, Derhovanessian E, Pflugfelder A, Eigentler TK,
[40] Fotin Mleczek M, Zanzinger K, Heidenreich R, Lorenz C, Thess A, Duchardt KM, et al. Direct injection of protamine-protected mRNA: results of a phase 1/2
et al. Highly potent mRNA based cancer vaccines represent an attractive plat- vaccination trial in metastatic melanoma patients. Journal of Immunotherapy
form for combination therapies supporting an improved therapeutic effect. The 2009;32(5):498–507.
Journal of Gene Medicine; in press. [65] Rittig SM, Haentschel M, Weimer KJ, Heine A, Muller MR, Brugger W, et al.
[41] Fotin-Mleczek M, Duchardt KM, Lorenz C, Pfeiffer R, Ojkic-Zrna S, Probst J, Intradermal vaccinations with RNA coding for TAA generate CD8+ and CD4+
et al. Messenger RNA-based vaccines with dual activity induce balanced TLR-7 immune responses and induce clinical benefit in vaccinated patients. Molecular
dependent adaptive immune responses and provide antitumor activity. Journal Therapy 2011;19(5):990–9.
of Immunotherapy 2011;34(1):1–15. [66] Bernstein DI, Reap EA, Katen K, Watson A, Smith K, Norberg P, et al.
[42] Kreiter S, Diken M, Selmi A, Diekmann J, Attig S, Husemann Y, et al. FLT3 lig- Randomized, double-blind, phase 1 trial of an alphavirus replicon vac-
and enhances the cancer therapeutic potency of naked RNA vaccines. Cancer cine for cytomegalovirus in CMV seronegative adult volunteers. Vaccine
Research 2011;71(19):6132–42. 2009;28(2):484–93.

You might also like