Subscriber access provided by READING UNIV
Article
Comparison of the expression kinetics and immunostimulatory activity of replicating mRNA, non-replicating mRNA and pDNA after intradermal electroporation in pigs Bregje Leyman, Hanne Huysmans, Séan Mc Cafferty, Francis Combes, Eric Cox, Bert Devriendt, and Niek N. Sanders Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.7b00722 • Publication Date (Web): 03 Jan 2018 Downloaded from http://pubs.acs.org on January 4, 2018
Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.
Molecular Pharmaceutics is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.
Page 1 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
1 2
Comparison of the expression kinetics and immunostimulatory activity of replicating mRNA, non-replicating mRNA and pDNA after intradermal electroporation in pigs
3 4 5 6 7 8 9 10
Bregje Leyman1,4, Hanne Huysmans1,4, Séan Mc Cafferty1,2, Francis Combes1,2, Eric Cox3,
11
Bert Devriendt3, Niek N. Sanders1,2*
12 1
13
Ethology, Laboratory for Gene Therapy, Heidestraat 19, 9820 Merelbeke, Belgium
14
2
15 16 17 18
Ghent University, Faculty of Veterinary Medicine, Department of Nutrition, Genetics and
3
Cancer Research Institute (CRIG), Ghent University, Belgium
Ghent University, Faculty of Veterinary Medicine, Department of Virology, Parasitology and Immunology, Salisburylaan 133, 9820 Merelbeke, Belgium *
Corresponding author:
Email address:
[email protected] 19 20
Tel: +32 9 264 78 08
4
These authors contributed equally to this work
1 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 2 of 26
21
Abstract
22
Synthetic mRNA is becoming increasingly popular as an alternative to pDNA-based gene
23
therapy. Currently, multiple synthetic mRNA platforms have been developed. In this study we
24
investigated the expression kinetics and the changes in mRNA encoding cytokine and
25
chemokine levels following intradermal electroporation in pigs of pDNA, self-replicating
26
mRNA, modified- and unmodified mRNA. The self-replicating mRNA tended to induce the
27
highest protein expression, followed by pDNA, modified mRNA and unmodified mRNA.
28
Interestingly, the self-replicating mRNA was able to maintain its high expression levels
29
during at least 12 days. In contrast, the expression of pDNA and the non-replicating mRNAs
30
dropped after respectively one and two days. Six days after intradermal electroporation a
31
dose-dependent expression was observed for all vectors. Again, also at lower doses, the self-
32
replicating mRNA tended to show the highest expression. All the mRNA vectors, including
33
the modified mRNA, induced elevated levels of mRNA encoding cytokines and chemokines
34
in the porcine skin after intradermal electroporation, while no such response was noticed after
35
intradermal electroporation of the pDNA vector.
36 37
Keywords: Pigs, pDNA, mRNA-therapeutics, luciferase expression, innate immune response
38 39 40 41 42 43 2 ACS Paragon Plus Environment
Page 3 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
44
Introduction
45
Nucleic acid-encoded drugs based on plasmid DNA (pDNA) and messenger RNA (mRNA)
46
form a potential new drug class with multiple applications such as protein replacement
47
therapy and vaccination for cancer and infectious diseases1,2. One of the first reports on the
48
use of pDNA and mRNA to express proteins in vivo dates from the 1990s, when Wolff et al.
49
made the observation that intramuscular injection of mice with non-formulated pDNA or
50
mRNA coding for a luciferase reporter protein, resulted in the detection of this protein3.
51
Nevertheless, after this report the field of gene therapy has been dominated for many years by
52
a focus on pDNA and viral vectors. This conservative attitude was based on the belief that
53
mRNA is a very fragile and instable molecule4. However, after more than two decades of
54
research the goal of commercialization of pDNA based vaccines for human usage has not
55
been reached. The reason for this is not clear but might be a consequence of several
56
drawbacks of pDNA. For example, pDNA vectors have a low efficacy in non- or slow
57
dividing cells5, they are sensitive to epigenetic silencing6, they often contain an antibiotic
58
resistance gene and they result, after e.g. intramuscular injection, in a long-term uncontrolled
59
expression7,8. This long-term expression might be an advantage for applications such as
60
protein replacement therapy reducing the dosage frequency of the treatment, but may be a
61
disadvantage for other applications such as vaccination. Although viral vectors are more
62
effective, they are also afflicted with important disadvantages like a complex production
63
process and their ability to trigger immune responses against viral epitopes on the vector9-11.
64
Additionally, some viral vectors integrate in the genome of the host12. As an alternative to
65
pDNA and viral vectors, in vitro transcribed (IVT) mRNA (synthetic mRNA) is becoming
66
increasingly popular. In contrast to pDNA and viral vectors, mRNA-based therapeutics are
67
expressed very efficiently in dividing and non-dividing cells despite the lower stability as a
68
consequence of the ubiquitous RNases. Additionally, they also do not contain antibiotic 3 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 4 of 26
69
resistance genes, carry virtually no risk of genomic integration and oncogenic mutagenesis
70
and their expression is limited in time1,13-15. Multiple synthetic mRNA platforms, such as
71
unmodified mRNA, modified mRNA and self-replicating mRNA, are currently available.
72
After intracellular delivery of unmodified synthetic mRNAs, pattern recognition receptors
73
(PRRs), such as toll-like receptors (TLRs), NOD-like receptors and RIG-like receptors, sense
74
the mRNA and trigger an innate immune response that is characterized by the production of
75
cytokines and chemokines (e.g. INF-β, IL-6, IL-12, CCL-5 and CXCL-10)16,17. The
76
formulation of synthetic mRNA into particles increases the capacity of synthetic mRNA to
77
stimulate the innate immune system because these particles mostly end up in endosomes,
78
which contain the RNA sensing TLRs 3, 7 and 818. Notwithstanding that the innate
79
immunostimulatory effects of unmodified mRNA may serve as an intrinsic adjuvant, and
80
hence might be a major benefit for vaccination, it might hamper the mRNA translation
81
process and trigger mRNA degradation19. Therefore, researchers have tried to decrease the
82
capacity of synthetic mRNA to stimulate the innate immune system by incorporating
83
modified nucleotides such as pseudouridine (Ψ), N(1)-methyl-pseudouridine (m1Ψ), 5-
84
methylcytidine (m5C), N6-methyladenosine (m6A), 5-methyluridine (m5U), or 2-thiouridine
85
(s2U)16,19,20. Beside these two non-replicating mRNA platforms, also self-replicating mRNA
86
is gaining more and more attention. Self-replicating mRNAs are mostly based on positive-
87
stranded RNA viruses, like alphaviruses, and contain RNA-dependent RNA polymerase genes
88
responsible for amplifying the transgene which replaced the structural viral proteins1,21. Self-
89
replicating mRNAs are inherently immunostimulatory, as several single stranded and double
90
stranded RNA species are formed during the amplification process, which can activate PRRs
91
resulting in secretion of type I IFNs22. Despite the many studies that used mRNA for protein
92
replacement therapies and for vaccination against e.g. cancer, allergies, viral- and bacterial
93
infections, mRNA-based therapeutics are far from a commercial product1,17,23-28. Moreover,
4 ACS Paragon Plus Environment
Page 5 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
94
most of those studies are based on small animal models (mainly mice) that poorly mimic
95
humans. The evaluation of nucleic acid-encoded drugs in large animals (> 10 kg), such as e.g.
96
pigs that are physiologically more closely related to humans than mice, is uncommon24,29.
97
Additionally, a side-by-side comparison of the expression efficacy and immunogenicity of
98
pDNA and the abovementioned different mRNA platforms has not been performed. In this
99
study the expression kinetics and the capacity to stimulate the innate immune system of either
100
(1) pDNA, (2) self-replicating mRNA, (3) modified- or (4) unmodified mRNA was compared
101
in a porcine model. Because of our interest in mRNA vaccination, we focused on intradermal
102
electroporation as the skin is extremely immuno-competent and easily accessible.
103
Additionally, porcine and human skin show several anatomical, immunological and
104
physiological similarities30. The data of this study might be very useful for the further
105
development of synthetic mRNA-based therapeutics, and especially the development of
106
mRNA-based vaccines.
107
Materials and methods
108
Mice and pigs
109
Seven week old female Balb/c mice (Janvier, France) were housed in individually ventilated
110
cages at 25 °C under natural day-night rhythm with ad libitum access to feed and water and
111
enriched with mouse houses and nesting material. Belgian landrace pigs of 12 weeks old were
112
housed together at 25 °C under natural day-night rhythm with ad libitum access to feed and
113
water. Experiments were started after an acclimatization period of at least 1 week. At the start
114
of the experiments the pigs weighed about 35-40 kg. All in vivo experiments were approved
115
by the ethical committee of the Faculty of Veterinary Medicine, Ghent University (EC
116
2013/57, 2015/77 and 2015/156).
117
Plasmids
5 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 6 of 26
118
The pGL4.13 [luc2/SV40] plasmid (pDNA) was purchased from Promega (Wisconsin, USA)
119
and contains the luc2 reporter gene and the SV40 early enhancer/promotor and a synthetic β-
120
lactamase (Ampr) coding region. This plasmid contains 293 CpG motifs (i.e. 63 CpG per 1000
121
base pair (bp); the sequence can be found at GenBank accession number AY738225.1).
122
pTK160 (11519 bp.) and pTK305 (4112 bp.) plasmids were used to produce the self-
123
replicating or (un)modified mRNAs by in vitro transcription (IVT). pTK305 and pTK160
124
contain, besides the luciferase gene, a bacteriophage T7 polymerase promoter, 5′ and 3′
125
untranslated regions (UTRs) and consensus recognition sequences for the I-SceI
126
endonuclease. pTK160 also contains nonstructural proteins (nSP1-4) of the Venezuelan
127
equine encephalitis virus (VEEV), that form the replicase complex. All constructs contain the
128
same Fluc2 gene (protein ID = AAV52875.1) which has been codon optimized for
129
mammalian expression. Conformity of the luciferase transcripts is essential for proper
130
comparison and subsequent selection of the most suited construct for future applications.
131
mRNA in vitro transcription
132
The unmodified and modified mRNAs were produced by IVT of the I-SceI-linearized
133
pTK305 plasmid using a MEGAscript T7 transcription Kit (Invitrogen, Massachusetts, USA)
134
with unmodified nucleotides or the m1Ψ modified nucleotides (Tebu-bio, Belgium) replacing
135
all the non-modified equivalents. The self-replicating mRNA was produced from the I-SceI-
136
linearized pTK160 plasmid using the same kit and unmodified nucleotides. Next, the mRNAs
137
were purified and capped using vaccinia virus capping enzymes and 2’-O-Methyltransferase
138
(Cellscript, Wisconsin, USA) to create cap1 and were then again purified using the RNeasy®
139
Mini kit (Qiagen, Germany). The self-replicating mRNAs have a poly(A) tail of 40
140
adenosines. The non-replicating mRNAs also have a poly(A) tail of 40 adenosines after IVT,
141
but for these mRNAs the poly(A) tail was extended by poly(A)tailing with the A-plusTM
142
Poly(A) polymerase Tailing Kit (Cellscript) to approximately 200 adenosines, followed by 6 ACS Paragon Plus Environment
Page 7 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
143
purification. All mRNA’s were quantified using a Nanodrop spectrophotometer (Thermo
144
Fisher Scientific, Massachusetts, USA) and the purity was determined by formamide agarose
145
gel electrophoresis as described previously31. The biological activity of each new batch of
146
mRNA was compared with that of previous batches. To this extent 5 µg of mRNA (dissolved
147
in 50 µl phosphate buffer saline (Ca2+ and Mg2+ free Dulbecco's Phosphate-Buffered Saline,
148
PBS, Ambion, Massachusetts, USA) was intradermally (ID) injected in mice (n = 3).
149
Injections were immediately followed by needle electroporation (AgilePulse, BTX Harvard
150
Apparatus, Massachusetts, USA) of the injection spot as described previously9 and the
151
bioluminescence was measured after 24 h, using an IVIS Lumina II (PerkinElmer, Belgium)
152
and the photon flux (photons/s) in the region of interest was calculated using the Living
153
IMAGE Software 4.3.1. Bioluminescence of untreated skin was measured and used as
154
background. The molecular weight of all final products was calculated by loading the
155
sequences of the mRNAs and pDNA into OligoCalc: an online oligonucleotide properties
156
calculator of Kibbe WA.
157
Optimization of ex vivo quantification of firefly luciferase expression
158
In vivo imaging of firefly luciferase expression in pigs is not possible, due to the size of the
159
animals. Therefore, we decided to measure the bioluminescence signal ex vivo after excision
160
of the injection spots. However, the storage time and temperature of the excised samples may
161
affect the bioluminescence signal. Therefore, we first determined the optimal storage
162
condition of the biopsies in mice. In more detail, Balb/c mice were ID injected with pDNA
163
coding for luciferase (20 µg in 50 µl PBS), followed by electroporation, as described before9.
164
Forty-eight hours after the injection 8 mm diameter skin punch biopsies (Miltex, The
165
Netherlands) were taken from the injection sites. All mice were humanely euthanized before
166
skin biopsies were taken. Half of the biopsies (n = 4) were immediately immersed in 24-well
167
plates containing 600 µl ice cold D-luciferin solution (15 mg D-luciferin (Gold 7 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 8 of 26
168
Biotechnology, Missouri, USA) per ml PBS). The other half was immediately immersed in
169
600 µl warm (37 °C) D-luciferin solution. After 15 min incubation on ice or at 37°C, the
170
bioluminescence signal was measured. Additionally, we also followed the evolution of the
171
bioluminescence signal in four excised spots during 90 min. This was done in a similar
172
manner as described above using an incubation on ice as this condition gave the best results
173
(see supplementary Figure 1 for results). These data showed that skin biopsies should be
174
immediately transferred to an ice-cold D-luciferin solution which is placed on ice.
175
Additionally, the bioluminescence signal should be measured as soon as possible after biopsy
176
using a fixed time point (in this study we used 18 min) after biopsy.
177
Analysis of pDNA and mRNA expression in pigs: kinetics and dose dependency
178
One week after arrival of the pigs, five rectangular (1 cm x 2 cm)-sites were shaved and
179
marked using tattoo ink (MS Schippers, Belgium) on the back of each pig, for later
180
identification of the injection sites. Subsequently, the expression kinetics of pDNA,
181
unmodified mRNA, modified mRNA and self-replicating mRNA was studied after
182
intradermal injection of 20 µg of the different vectors (dissolved in 50 µl PBS) in the marked
183
spots. Control spots were injected with PBS. Injection spots were immediately electroporated
184
using 4 mm gap needle array electrodes (needle length 5 mm) using the AgilePulse-BTX
185
Harvard Apparatus-program-protocol (two pulses of 450 V with a pulse duration of 0.050 ms
186
and a pulse interval of 0.2 ms; and subsequently after 50 ms eight pulses of 110 V with a
187
pulse duration of 10 ms and a pulse interval of 20 ms). One-, two-, six- and twelve days
188
following administration, the luciferase expression of the different mRNA vectors and the
189
pDNA was determined by taking a skin punch biopsy of the injection spots. Ex vivo
190
bioluminescent imaging of the biopsies occurred as optimized in mice. For each construct
191
(including control) and time point we had five injection spots. Additionally, we also evaluated
192
the dose dependency of the luciferase expression. For that purpose, pigs were again shaved 8 ACS Paragon Plus Environment
Page 9 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
193
and marked. The marked spots were subsequently ID electroporated with 1, 5 or 20 µg of
194
each of the luciferase expression mRNA vectors and pDNA. After 1 and 6 days all animals
195
were humanely euthanized, skin biopsies were collected, and ex vivo bioluminescent imaging
196
occurred as optimized previously in mice. For each dose and time point we included 4
197
injection spots.
198
Analysis of the local innate immune response after intradermal electroporation of pDNA
199
and mRNA vectors in pigs
200
To study the effect on the innate immune system of each of the luciferase expression vectors,
201
pigs were ID injected with 20 µg of (1) pDNA, (2) self-replicating mRNA, (3) modified
202
mRNA, (4) unmodified mRNA and (5) PBS as a control. All injections were immediately
203
followed by electroporation as described above. Twenty-four hours later, pigs were humanely
204
sacrificed, and skin biopsies were taken from the injection sites, and immediately frozen in
205
RNAlater (Sigma-Aldrich, Germany) and stored at -20 °C until further analysis. RNA was
206
isolated from the biopsies following the RNAzol® RT protocol (Sigma-Aldrich). RNA
207
concentrations were assessed at 260 nm by a Nanodrop spectrophotometer (Thermo Fisher
208
Scientific) and the purity of the RNA samples was checked using an Experion RNA StdSens
209
Analysis kit (Bio-rad Laboratories, California, USA). Reverse transcription was carried out
210
using the RT² First Strand kit (Qiagen) according to the manufacturer’s instructions. Purity
211
and quantity were assessed again after reverse transcription. Prior to gene expression analysis
212
using the commercially available RT² profiler PCR Array (PASS-122Z pig antiviral response,
213
Qiagen), the cDNA template was mixed with RT² SYBR Green mastermix (Qiagen) and
214
diluted with RNase-free water according to the RT² profiler PCR Array Handbook from
215
Qiagen. Thermo-cycling was performed in 384-well plates and parameters were set as
216
described in the manufacturer’s protocol.
217
Statistical analysis 9 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 10 of 26
218
Statistical analysis was performed using GraphPad Prism 6 (GraphPad Software, California,
219
USA). Differences between groups were analysed by means of a non-parametric Kruskal-
220
Wallis test and a post-hoc test following Dunn using the Benjamini-Hochberg correction for
221
multiple testing. The qPCR results were analyzed using the CFX manager software v3.0 (Bio-
222
Rad Laboratories) by a Gene Study with normalized expression (∆∆Cq) based on the mean
223
efficiency corrected CT-value and after inter-run calibration. A P-value of p < 0.05 was
224
considered significant.
225
Results
226
The transfection capacity of self-replicating mRNA outperforms pDNA and non-
227
replicating mRNA in the porcine skin
228
The luciferase expression of the different mRNA vectors was slightly below that of pDNA
229
one day after intradermal electroporation in pigs (Figure1). Interestingly, at day 1 and 2 the
230
luciferase expression of the different mRNA vectors was similar and not significantly
231
different from each other. While the average luciferase expression of modified and
232
unmodified mRNA showed a 2.5 and 5-fold decrease at day 6 and 12 respectively, the
233
average luciferase signal of the self-replicating mRNA showed, compared to the expression
234
on day 1 and 2, a slight increase at day 6 and 12 (Figure 1). The pDNA luciferase expression
235
levels tended to outperform the mRNA luciferase expression levels one day after injection,
236
however, the average pDNA signal dropped below the signal of all mRNAs on day 2, and was
237
inferior to the signal of the self-replicating mRNA until the end of the experiment. After day 2
238
the expression of the pDNA stayed constant till day 6 and subsequently showed a further brief
239
drop at day 12 (Figure 1). Note that no significant differences have been noticed and only
240
trends could be observed. We also calculated the area under the curve as a measure of the
241
total amount of luciferase produced during the 12-day follow-up period (Figure 2). Although
242
no significant differences could be observed, Figure 2 shows that self-replicating mRNA 10 ACS Paragon Plus Environment
Page 11 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
243
tends to perform better than pDNA, unmodified as well as modified non-replicating mRNA
244
after intradermal electroporation in pigs.
245
246 247
Figure 1. Quantification of the mean bioluminescence signal after intradermal electroporation
248
of the mRNA vectors and pDNA in pigs. Skin biopsies of the injection spots were taken 1
249
day, 2 days, 6 days and 12 days after intradermal electroporation of 20 µg of either self-
250
replicating mRNA (3.83 x 1012 molecules), unmodified mRNA (1.49 x 1013 molecules),
251
modified mRNA (1.48 x 1013 molecules), or pDNA (4.20 x 1012 molecules mole). The value
252
between brackets is the copy number, i.e. the number of molecules present in 20 µg of the
253
vectors. The mean value of the total flux (p/s) is given along with the standard error of the
254
mean (SEM). No significant (p > 0.05) differences were observed. Each data point is the
255
mean of five biopsies obtained from five different pigs. The dotted line represents the
256
background signal (Bkg).
11 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 12 of 26
257 258
Figure 2. The area under the curve (AUC) of the bioluminescence signal versus time graphs
259
was calculated, along with the SEM, over a period of 12 days after intradermal
260
electroporation of 20 µg of either self-replicating mRNA (3.83 x 1012 molecules), unmodified
261
mRNA (1.49 x 1013 molecules), modified mRNA (1.48 x 1013 molecules), or pDNA (4.20 x
262
1012 molecules). For these calculations, the bioluminescence signal of the negative controls
263
was subtracted from all data points. The value between brackets is the copy number, i.e. the
264
number of molecules present in 20 µg of the vectors. Each bar is the mean of five biopsies
265
obtained from five different pigs (n=5). No significant differences could be detected.
266
Effect of the vector dose on the luciferase expression
267
The expression level of different pDNA and mRNA doses (1 µg, 5 µg, 20 µg) were compared
268
1 and 6 days after intradermal electroporation (Figure 3). One day after injection, a real dose-
269
dependent effect on the luciferase expression was not noticed for the non-replicating mRNAs
270
and the pDNA. Nevertheless, the expression of pDNA tends to show an increase when 20 µg
271
was used. Surprisingly, one day after administration, the self-replicating mRNA showed the
272
highest expression at the lowest dose (1 µg), while the two other doses (5 and 20 µg) showed
273
a circa three-fold lower expression (Figure 3). Six days after intradermal electroporation the
12 ACS Paragon Plus Environment
Page 13 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
274
expression levels of all vectors and doses showed a huge drop, except the expression level of
275
the self-replicating mRNA, which showed a slight increase when a dose of 20 µg was
276
administered (Figure 3). Additionally, a more clear dose-dependent expression was observed
277
for all mRNA vectors and pDNA six days after intradermal electroporation but no significant
278
differences could be observed.
279
280 281
Figure 3. Dose-dependent expression after intradermal electroporation in pigs of 1 µg, 5 µg
282
and 20 µg of the mRNA vectors or pDNA. The bioluminescence signal in the punch biopsies
283
was measured one day and six days after injection. The average value of the total flux (p/s) in
284
the skin biopsies from four pigs (1 µg and 5 µg) or five pigs (20 µg) is given along with its
285
SEM. The dotted line represents the background signal (Bkg). At each dose and time point
286
the expression levels of the different vectors were not significantly different. Note that in the
287
x-axis also the copy number of each vector is shown in mole.
288
Electroporation of synthetic mRNA but not pDNA induces transcriptional changes
289
associated with an innate immune response in the porcine skin 13 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 14 of 26
290
Next we analyzed the capacity of the synthetic mRNAs and pDNA to provoke changes in the
291
transcriptome of cytokines and chemokines involved in the pig antiviral immune response
292
after intradermal electroporation in pigs. Elevated expression of mRNAs coding for these
293
cytokines and chemokines might give us information about the immune stimulatory properties
294
of the different constructs. Depending on this potential, the constructs can be used for
295
different applications. A strong self-adjuvant effect can be advantageous for mRNA based
296
vaccines. For protein-replacement therapies, however, stimulation of the innate immune
297
system should be kept to a minimum17. Twenty-four hours after intradermal electroporation of
298
20 µg of either (1) pDNA, (2) unmodified mRNA, (3) modified mRNA and, (4) self-
299
replicating mRNA the innate immune response was characterized by analyzing the expression
300
level of 84 cytokines and chemokines in the injections spots (Figure 4). Intradermal
301
electroporation of pDNA did, compared to control (i.e. PBS plus electroporation), not
302
significantly affect the mRNA expression level of any of the tested genes. In contrast, self-
303
replicating mRNA, 1mψ-modified mRNA and unmodified mRNA, induced a significant (p
0.05) than the induced
310
upregulation after electroporation of modified mRNA and self-replicating mRNA.
311
14 ACS Paragon Plus Environment
Page 15 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
312 313
Figure 4. Gene regulation of 84 genes involved in innate immunity after intradermal
314
electroporation of 20 µg of the different vectors in pigs. Twenty-four hours after
315
administration of the vectors total RNA was isolated from the excised injection spots and the
316
expression of the different innate immune marker genes was measured relative to control (i.e.
317
intradermal electroporation of PBS). The heat map represents normalized mean expression
318
values depicted as log 2-fold change relative to control. Reported values were obtained from
319
three biopsies taken from three different pigs. Values indicated by a star* indicate a
320
significant difference (p < 0.05) compared to control (n = 3).
321
15 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 16 of 26
322 323
Figure 5. Innate immune marker genes that were commonly upregulated by all three mRNA
324
vectors 24 h after intradermal electroporation in pigs. The graph shows the normalized mean
325
expression values of the indicated genes relative to control (i.e. intradermal electroporation of
326
PBS). Each bar represents mean ± standard deviation of three biopsies from three different
327
pigs. There were no significant differences between the different mRNA vectors.
328
Discussion
329
In this study, we compared the expression kinetics and intrinsic effect on the innate immunity
330
of three mRNA-based platforms and a state-of-the-art pDNA vector in pigs. Messenger RNA
331
based therapeutics have the potential to form a new class of drugs with a broad range of
332
possible applications. Especially in the field of vaccination, mRNA is very popular because of
333
its safety and its high transfection efficiency in non-dividing cells15. Additionally, mRNA
334
vaccines have a great flexibility with respect to production and application, enabling a fast
335
development of e.g. prophylactic vaccines against emerging diseases. Different mRNA
336
platforms have been developed allowing researchers to select the most suited mRNA platform
337
for their application32. Most preclinical studies with mRNA therapeutics have been performed
338
in mice. For pDNA therapeutics it is well-known that they have a much lower efficiency in 16 ACS Paragon Plus Environment
Page 17 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
339
larger animals and humans than in mice, which makes translation of data obtained in mice to
340
humans difficult10,11,33,34. In this work, we evaluated different mRNA platforms in pigs. The
341
mRNAs were administered via intradermal injection. This route was chosen because of our
342
interest in mRNA vaccination. To prevent carrier related effects on the efficiency and
343
immunogenicity of the vectors we used electroporation to increase the intracellular delivery of
344
the mRNAs and pDNA.
345
In this study we found that the expression after intradermal electroporation of pDNA reaches
346
a maximum after one day and is followed by a sharp drop in expression at day 2. This drop
347
was unexpected as after intradermal electroporation of the same pDNA in mice we observed
348
that the pDNA reaches its maximal expression after about two days and stays on a high level
349
up to six days after administration (Supplementary Figure 2). This drop in pDNA expression
350
in pigs after day 1 might be attributed to epigenetic silencing. It is also surprising that one
351
day after administration pDNA and not the non-replicating mRNAs shows the highest
352
expression. Indeed, after delivery of pDNA in the cytosol it has the longest path to fulfil
353
before it becomes translated: it has to migrate to the nucleus where it gets transcribed into
354
mRNA and subsequently the mRNA must be transported to the cytosol where it is translated
355
into proteins.
356
The non-replicating mRNAs also reach their maximum one day after injection and
357
subsequently show a steady decrease in expression until day six. In mice similar drops in
358
expression that start 4 to 24h after carrier mediated delivery of non-replicating mRNAs have
359
been reported35-37. The group of Joachim Rädler determined that the intracellular half-life of
360
mRNA after intracellular delivery is about 11h. Taking into account that the half-life of firefly
361
luciferase is only a few hours
362
delivery nicely fits with this degradation kinetics of mRNA. An early drop in expression as
363
seen with the non-replicating mRNAs does occur after administration of the self-replicating
38,39
, the reported luciferase expression kinetics after mRNA
17 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 18 of 26
364
mRNA. The capacity of the self-replicating mRNA to produce many subgenomic mRNA
365
copies in the cell enables this vector to maintain its protein production level during at least 12
366
days in the skin of pigs. However, this effect is only obtained when the highest dose (20 µg) is
367
used. In mice, only 1 µg of the same self-amplifying mRNA is needed to obtain such
368
sustained protein production, which indicates that mice are easier transfected with self-
369
replicating mRNA than pigs (Supplementary Figure 3). The self-replicating mRNA needs
370
some time before the subgenomic mRNA is abundantly produced in the second cycle of
371
replication. This causes a maximum expression around six days after injection as expected
372
taking into account previously reported expression profiles of replicating mRNAs in mice40.
373
Furthermore, when interpreting these data one must also take into account that, based on the
374
molecular weight, the number of self-replicating mRNA molecules per mass is about 4 times
375
lower than the number of non-replicating mRNA molecules and comparable to the number of
376
pDNA molecules. Taking into account this difference in copy number and the difference in
377
luciferase expression between self-amplifying mRNA, non-replicating mRNA and pDNA at
378
later time points (day 6 and 12) (Figure 1), we can conclude that the self-amplifying mRNA is
379
about 100, 40 and 4 times more effective on day 6 and 50, 30 and 5 times more effective on
380
day 12 than unmodified mRNA, modified mRNA and pDNA, respectively. We also need to
381
take into account that none of the differences between the expression kinetics of the different
382
vectors appeared to be significant. This is the result of the small number of pigs (4 or 5 per
383
experiment) in combination with high variability of the ex vivo bioluminescence signal. The
384
mean values and their standard errors of the individual data points shown in Figures 1, 2, 3
385
and means and the standard deviations of Figure 5 are displayed in Supplementary Table 1.
386
Therefore, we can only draw conclusions about predictive trends in expression kinetics.
387
Next, we compared the capacity of the synthetic mRNAs and pDNA to stimulate the innate
388
immune system by measuring transcriptional changes in innate immune responsive genes one 18 ACS Paragon Plus Environment
Page 19 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
389
day after intradermal electroporation in pigs. An innate immune alarm might be
390
advantageous, as it can serve as a self-adjuvant, for mRNA based vaccines. However, for
391
protein-replacement therapies such innate immune response might diminish the protein
392
expression efficiency and promote an immune response against the therapeutic protein36. The
393
RT2 profiler PCR array demonstrated that self-amplifying, unmodified and modified mRNA,
394
but not pDNA, triggered an innate immune response after intradermal electroporation in pigs.
395
This is in accordance with previous studies in large animals and humans, showing poor
396
immunogenicity of DNA vaccines compared to their immunogenicity in small
397
animals10,11,33,34. Our results indicate a significantly higher expression of genes involved in
398
signaling downstream of toll-like receptors (IRF7, CCL5/RANTES, CD40), NOD-like
399
Receptors (CASP1, OAS2), RIG-1-Like receptors (DDX58/RIG-1, DHX58, IFIH1, ISG15,
400
IRF7) and type 1 Interferon signaling (MX1) for all tested mRNAs compared to control (i.e.
401
PBS plus electroporation). Signaling through these different pathways results in the activation
402
of e.g. type 1 interferon (IFN), tumor necrosis factor (TNF), interleukin-6 (IL-6) and caspase
403
1 activation. This creates a pro-inflammatory microenvironment that can ameliorate the
404
efficiency of mRNA vaccines1. Ishii et al. demonstrated that TANK-binding kinase-1 (TBK1)
405
plays an important role in the innate immune response after intramuscular electroporation of
406
pDNA vaccines in mice41. However, in our study intradermal electroporation of pDNA in pigs
407
causes a downregulation of TBK1. Also others found that stimulation of the IRF3 pathway
408
can result in a temporal decrease in TBK142. It has been described in the past that the
409
incorporation of modified nucleosides such as m1Ψ in mRNA can decrease the activation of
410
the innate immune system43. Therefore, we were surprised that also the m1Ψ modified mRNA
411
induced an upregulation of innate immune responsive genes19. Nevertheless, unmodified
412
mRNA induced the highest upregulation of the innate immune marker genes (Figure 4).
413
Interestingly, self-amplifying mRNA increased the innate immune responsive genes to a
19 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 20 of 26
414
lesser extent than unmodified mRNA. One would expect the opposite as many uncompleted
415
mRNA strands are present during the self-amplification process. However, our self-
416
amplifying mRNA is based on the genome of a positive-strand RNA virus, which are known
417
to replicate in organelle-like structures, which prevents an exorbitant innate immune
418
response44. The upregulation of genes that signal downstream of toll-like receptors and the
419
weak upregulation of TLR3, 7 and 8 (Figure 4) was remarkable as one may expect that
420
mRNA does not encounter these endosomal TLRs after electroporation16. However, in recent
421
work it has been proposed that electroporation can also induce endocytosis (electro-
422
endocytosis)45. Therefore, it is possible that part of the electro-transferred mRNA uses the
423
classical endosomal trafficking pathway, and is recognized by TLRs residing in endosomes45.
424
In line with this reasoning, we would also expect an upregulation of TLR9 after
425
electroporation of pDNA as TLR9 recognizes unmethylated CpG motifs which are abundantly
426
present in the pGL4.13 plasmid (63 per 1000 bp). However, our data show that
427
electroporation of this pDNA did not cause a significant upregulation of TLR9 mRNA in pigs.
428
It has been reported by Hochrein et al. and Guzylack-Pirou et al. that pigs show a lower
429
response to CpG containing oligodeoxynucleotides compared to mice46,47. This may explain
430
the non-significant upregulation of TLR9 mRNA and the absence of a significant induction of
431
innate immune responsive genes after intradermal electroporation of pDNA in pigs. However,
432
another explanation of the lack of an innate immune response after intradermal
433
electroporation of pDNA in pigs in our study can also be due to the low dose (20µg pDNA
434
per 40kg, i.e. 0.5µg/kg). Indeed, a recent study in non-human primates showed that
435
intradermal electroporation of a HIV pDNA vaccine at a much higher dose of 22µg/kg
436
resulted in a significant, although moderate, induction of IL-1248
437
Finally, the production costs of therapeutic mRNAs will also be of importance for their
438
implementation in future applications. Production of mRNA on research scale is rather 20 ACS Paragon Plus Environment
Page 21 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
439
expensive but the key to reduction of the production cost is upscaling of the production
440
process to an industrial level. The mandatory Good Manufacturing Practice (GMP)
441
regulations require product-specific validation. For mRNA however, the same production
442
process can be used for many different mRNA vaccines, bypassing the need for new
443
validation. This ensures production costs that are competitive with those of protein, peptide,
444
DNA, cell or recombinant pathogen based vaccines14. For vaccination purposes, an adequate
445
immune response is required to induce a memory immune response. Nucleotide modifications
446
have been shown to reduce this immune response by diminishing binding to TLRs, making
447
the modified mRNA maybe less suited for vaccination and more suited for example protein
448
replacement therapy. These modified nucleotides are more expensive compared to the
449
conventional ones. In our modified mRNA, the uridine was completely replaced by N1-
450
methylpseudouridine. Kormann et al., however, showed that a replacement of 25% is
451
sufficient to provoke a decrease of mRNA binding to pattern recognition receptors, what
452
would also lead to reduced production costs17.
453 454
Conclusions
455
Finally, to our knowledge, this is the first study in pigs where the intradermal translational
456
kinetics and the effect on the innate immune system of different synthetic mRNA-platforms
457
and pDNA were compared. In conclusion, our results show that mRNA modifications do not
458
completely prevent recognition by innate immune receptors in pigs, compared to previous
459
studies in mice16. This is probably due to the fact that we used silica columns and not a HPLC
460
based method (to remove double stranded mRNAs) for the purification of our mRNA.
461
Furthermore, synthetic mRNA, and specifically self-replicating mRNA, is a very promising
462
and a cost-effective DNA-alternative for the development of genetic therapeutics for use in
463
farm animals or humans14. In the future, mRNA based therapeutics may replace the current 21 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 22 of 26
464
commercial available DNA-based therapeutics in veterinary medicine like Oncept® (a DNA
465
cancer vaccine for dogs, Merial), Apex®-IHN (a DNA vaccine for fish, Vical), Clynav® (a
466
DNA vaccine for fish, Elanco) and LifeTide® SW 5 (a DNA vector for pigs encoding growth
467
hormone releasing hormone, VGX™ Animal Health)49.
468 469
Acknowledgements
470
We are grateful to Prof. dr. Frank Pasmans of the Department of Bacteriology, Pathology and
471
Avian Diseases, Faculty of Veterinary Medicine, UGent, among other things for the use of the
472
CFX384 Bio-rad-cycler. Furthermore, the technical assistance of Sofie De Bruyckere
473
(Department of Bacteriology, Pathology and Avian Diseases, Faculty of Veterinary Medicine,
474
UGent) and Rudy Cooman (Department of Virology, Parasitology and Immunology, Faculty
475
of Veterinary Medicine, UGent) is greatly appreciated. The plasmids pTK160 and pTK305
476
were kind gifts from Tasuku Kitada and Ron Weiss (Massachusetts Institute of Technology,
477
USA). This work was supported by the concerted research action (GOA) fund of Ghent
478
University: Project Code BOF15/GOA/013.
479
Supporting Information
480
Supplementary figure 1: (A) Effect of storage temperature of skin biopsies on the ex vivo
481
bioluminescence signal. (B) Time-dependent evolution of the bioluminescence signal in the
482
skin biopsies.
483
Supplementary Figure 2: Luciferase expression kinetics after intradermal electroporation of
484
10 µg of pDNA in mice.
485
Supplementary Figure 3: Luciferase expression kinetics after intradermal electroporation of 1
486
µg of self-replicating mRNA in mice.
22 ACS Paragon Plus Environment
Page 23 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
487
Supplementary Table 1: Overview of the variance of the data displayed in Figures 1, 2, 3 and
488
5. Variance is displayed as standard error of the mean (SEM) of the bioluminescence signal
489
(p/s) for Figures 1-3 and standard deviation of the bioluminescence signal for Figure 5.
490
References
491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531
1 2
3 4 5 6 7
8
9
10
11
12
13 14
15
Sahin, U., Kariko, K. & Tureci, O. mRNA-based therapeutics--developing a new class of drugs. Nat Rev Drug Discov 13, 759-780, doi:10.1038/nrd4278 (2014). Steinle, H., Behring, A., Schlensak, C., Peter Wendel, H. & Avci-Adali, M. Application of in vitro transcribed messenger RNA for cellular engineering and reprogramming: Progress and challenges. Stem Cells, doi:10.1002/stem.2402 (2016). Wolff, J. A. et al. Direct gene transfer into mouse muscle in vivo. Science 247, 1465-1468 (1990). Pascolo, S. The messenger's great message for vaccination. Expert Rev Vaccines 14, 153-156, doi:10.1586/14760584.2015.1000871 (2015). Dupuis, M. et al. Distribution of DNA vaccines determines their immunogenicity after intramuscular injection in mice. J Immunol 165, 2850-2858 (2000). Krishnan, M. et al. Effects of epigenetic modulation on reporter gene expression: implications for stem cell imaging. FASEB J 20, 106-108, doi:10.1096/fj.05-4551fje (2006). Geall, A. J. et al. Nonviral delivery of self-amplifying RNA vaccines. Proceedings of the National Academy of Sciences of the United States of America 109, 14604-14609, doi:10.1073/pnas.1209367109 (2012). Deering, R. P., Kommareddy, S., Ulmer, J. B., Brito, L. A. & Geall, A. J. Nucleic acid vaccines: prospects for non-viral delivery of mRNA vaccines. Expert Opin Drug Deliv 11, 885-899, doi:10.1517/17425247.2014.901308 (2014). Denies, S., Cicchelero, L., Van Audenhove, I. & Sanders, N. N. Combination of interleukin-12 gene therapy, metronomic cyclophosphamide and DNA cancer vaccination directs all arms of the immune system towards tumor eradication. J Control Release 187, 175-182, doi:10.1016/j.jconrel.2014.05.045 (2014). van Rooij, E. M. et al. Effect of vaccination route and composition of DNA vaccine on the induction of protective immunity against pseudorabies infection in pigs. Vet Immunol Immunopathol 66, 113-126 (1998). MacGregor, R. R. et al. First human trial of a DNA-based vaccine for treatment of human immunodeficiency virus type 1 infection: safety and host response. J Infect Dis 178, 92-100 (1998). Hacein-Bey-Abina, S. et al. A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. N Engl J Med 348, 255-256, doi:10.1056/NEJM200301163480314 (2003). Zabner, J., Fasbender, A. J., Moninger, T., Poellinger, K. A. & Welsh, M. J. Cellular and molecular barriers to gene transfer by a cationic lipid. J Biol Chem 270, 18997-19007 (1995). Kallen, K. J. & Thess, A. A development that may evolve into a revolution in medicine: mRNA as the basis for novel, nucleotide-based vaccines and drugs. Ther Adv Vaccines 2, 10-31, doi:10.1177/2051013613508729 (2014). Van Tendeloo, V. F. et al. Highly efficient gene delivery by mRNA electroporation in human hematopoietic cells: superiority to lipofection and passive pulsing of mRNA and to electroporation of plasmid cDNA for tumor antigen loading of dendritic cells. Blood 98, 49-56 (2001).
23 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581
16
17 18
19
20
21 22
23
24
25 26
27
28 29 30 31 32 33
34
Page 24 of 26
Andries, O. et al. Innate immune response and programmed cell death following carriermediated delivery of unmodified mRNA to respiratory cells. Journal of controlled release : official journal of the Controlled Release Society 167, 157-166, doi:10.1016/j.jconrel.2013.01.033 (2013). Kormann, M. S. et al. Expression of therapeutic proteins after delivery of chemically modified mRNA in mice. Nat Biotechnol 29, 154-157, doi:10.1038/nbt.1733 (2011). Nguyen, D. N. et al. Lipid-derived nanoparticles for immunostimulatory RNA adjuvant delivery. Proceedings of the National Academy of Sciences of the United States of America 109, E797-803, doi:10.1073/pnas.1121423109 (2012). Anderson, B. R. et al. Incorporation of pseudouridine into mRNA enhances translation by diminishing PKR activation. Nucleic acids research 38, 5884-5892, doi:10.1093/nar/gkq347 (2010). Kariko, K., Buckstein, M., Ni, H. & Weissman, D. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity 23, 165-175, doi:10.1016/j.immuni.2005.06.008 (2005). Lundstrom, K. Alphaviruses in gene therapy. Viruses 7, 2321-2333, doi:10.3390/v7052321 (2015). Pushko, P. et al. Replicon-helper systems from attenuated Venezuelan equine encephalitis virus: expression of heterologous genes in vitro and immunization against heterologous pathogens in vivo. Virology 239, 389-401, doi:10.1006/viro.1997.8878 (1997). Brazzoli, M. et al. Induction of Broad-Based Immunity and Protective Efficacy by Selfamplifying mRNA Vaccines Encoding Influenza Virus Hemagglutinin. J Virol 90, 332-344, doi:10.1128/JVI.01786-15 (2016). Bogers, W. M. et al. Potent immune responses in rhesus macaques induced by nonviral delivery of a self-amplifying RNA vaccine expressing HIV type 1 envelope with a cationic nanoemulsion. J Infect Dis 211, 947-955, doi:10.1093/infdis/jiu522 (2015). Li, J. et al. Messenger RNA vaccine based on recombinant MS2 virus-like particles against prostate cancer. Int J Cancer 134, 1683-1694, doi:10.1002/ijc.28482 (2014). Steitz, J., Britten, C. M., Wolfel, T. & Tuting, T. Effective induction of anti-melanoma immunity following genetic vaccination with synthetic mRNA coding for the fusion protein EGFP.TRP2. Cancer Immunol Immunother 55, 246-253, doi:10.1007/s00262-005-0042-5 (2006). Weiss, R., Scheiblhofer, S., Roesler, E., Weinberger, E. & Thalhamer, J. mRNA vaccination as a safe approach for specific protection from type I allergy. Expert Rev Vaccines 11, 55-67, doi:10.1586/erv.11.168 (2012). Tavernier, G. et al. mRNA as gene therapeutic: how to control protein expression. J Control Release 150, 238-247, doi:10.1016/j.jconrel.2010.10.020 (2011). Brito, L. A. et al. A cationic nanoemulsion for the delivery of next-generation RNA vaccines. Mol Ther 22, 2118-2129, doi:10.1038/mt.2014.133 (2014). Debeer, S. et al. Comparative histology and immunohistochemistry of porcine versus human skin. Eur J Dermatol 23, 456-466, doi:10.1684/ejd.2013.2060 (2013). Andries, O. et al. Comparison of the gene transfer efficiency of mRNA/GL67 and pDNA/GL67 complexes in respiratory cells. Mol Pharm 9, 2136-2145, doi:10.1021/mp200604h (2012). Schlake, T., Thess, A., Fotin-Mleczek, M. & Kallen, K. J. Developing mRNA-vaccine technologies. RNA Biol 9, 1319-1330, doi:10.4161/rna.22269 (2012). Verfaillie, T. et al. Priming of piglets against enterotoxigenic E. coli F4 fimbriae by immunisation with FAEG DNA. Vaccine 22, 1640-1647, doi:10.1016/j.vaccine.2003.09.045 (2004). Hirao, L. A. et al. Intradermal/subcutaneous immunization by electroporation improves plasmid vaccine delivery and potency in pigs and rhesus macaques. Vaccine 26, 440-448, doi:10.1016/j.vaccine.2007.10.041 (2008).
24 ACS Paragon Plus Environment
Page 25 of 26 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620
Molecular Pharmaceutics
35
36
37 38
39
40 41 42
43
44 45 46 47
48
49
Kariko, K., Muramatsu, H., Keller, J. M. & Weissman, D. Increased erythropoiesis in mice injected with submicrogram quantities of pseudouridine-containing mRNA encoding erythropoietin. Mol Ther 20, 948-953, doi:10.1038/mt.2012.7 (2012). Pardi, N. et al. Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. J Control Release 217, 345-351, doi:10.1016/j.jconrel.2015.08.007 (2015). Oberli, M. A. et al. Lipid Nanoparticle Assisted mRNA Delivery for Potent Cancer Immunotherapy. Nano Lett 17, 1326-1335, doi:10.1021/acs.nanolett.6b03329 (2017). Ignowski, J. M. & Schaffer, D. V. Kinetic analysis and modeling of firefly luciferase as a quantitative reporter gene in live mammalian cells. Biotechnol Bioeng 86, 827-834, doi:10.1002/bit.20059 (2004). Leclerc, G. M., Boockfor, F. R., Faught, W. J. & Frawley, L. S. Development of a destabilized firefly luciferase enzyme for measurement of gene expression. Biotechniques 29, 590-591, 594-596, 598 passim (2000). Ljungberg, K. & Liljestrom, P. Self-replicating alphavirus RNA vaccines. Expert Rev Vaccines 14, 177-194, doi:10.1586/14760584.2015.965690 (2015). Ishii, K. J. et al. TANK-binding kinase-1 delineates innate and adaptive immune responses to DNA vaccines. Nature 451, 725-729, doi:10.1038/nature06537 (2008). Zhao, Y. X., Tian, B., Edeh, C. B. & Brasier, A. R. Quantitation of the Dynamic Profiles of the Innate Immune Response Using Multiplex Selected Reaction Monitoring-Mass Spectrometry. Mol Cell Proteomics 12, 1513-1529, doi:10.1074/mcp.M112.023465 (2013). Kariko, K. et al. Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. Mol Ther 16, 1833-1840, doi:10.1038/mt.2008.200 (2008). Romero-Brey, I. & Bartenschlager, R. Membranous replication factories induced by plusstrand RNA viruses. Viruses 6, 2826-2857, doi:10.3390/v6072826 (2014). Rosazza, C., Meglic, S. H., Zumbusch, A., Rols, M. P. & Miklavcic, D. Gene Electrotransfer: A Mechanistic Perspective. Curr Gene Ther 16, 98-129 (2016). Hochrein, H. & Wagner, H. Of men, mice and pigs: looking at their plasmacytoid dendritic cells [corrected]. Immunology 112, 26-27, doi:10.1111/j.1365-2567.2004.01878.x (2004). Guzylack-Piriou, L., Balmelli, C., McCullough, K. C. & Summerfield, A. Type-A CpG oligonucleotides activate exclusively porcine natural interferon-producing cells to secrete interferon-alpha, tumour necrosis factor-alpha and interleukin-12. Immunology 112, 28-37, doi:10.1111/j.1365-2567.2004.01856.x (2004). Todorova, B. et al. Electroporation as a vaccine delivery system and a natural adjuvant to intradermal administration of plasmid DNA in macaques. Sci Rep 7, 4122, doi:10.1038/s41598-017-04547-2 (2017). Myhr, A. I. DNA Vaccines: Regulatory Considerations and Safety Aspects. Curr Issues Mol Biol 22, 79-88, doi:10.21775/cimb.022.079 (2016).
621
25 ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
622
Abstract graphics
623
For table of contents use only.
624 625
Title: Comparison of the expression kinetics and immunostimulatory activity of replicating mRNA, non-replicating mRNA and pDNA after intradermal electroporation in pigs.
626 627
Authors: Bregje Leyman, Hanne Huysmans, Séan Mc Cafferty, Francis Combes, Eric Cox, Bert Devriendt, Niek N. Sanders
Page 26 of 26
628
629
26 ACS Paragon Plus Environment