You are on page 1of 34

 

 
Enhanced intranasal delivery of mRNA vaccine by overcoming the nasal
epithelial barrier via intra- and paracellular pathways

Man Li, Mengnan Zhao, Yao Fu, You Li, Tao Gong, Zhirong Zhang,
Xun Sun

PII: S0168-3659(16)30107-9
DOI: doi: 10.1016/j.jconrel.2016.02.043
Reference: COREL 8156

To appear in: Journal of Controlled Release

Received date: 28 October 2015


Revised date: 30 January 2016
Accepted date: 27 February 2016

Please cite this article as: Man Li, Mengnan Zhao, Yao Fu, You Li, Tao Gong, Zhirong
Zhang, Xun Sun, Enhanced intranasal delivery of mRNA vaccine by overcoming the
nasal epithelial barrier via intra- and paracellular pathways, Journal of Controlled Release
(2016), doi: 10.1016/j.jconrel.2016.02.043

This is a PDF file of an unedited manuscript that has been accepted for publication.
As a service to our customers we are providing this early version of the manuscript.
The manuscript will undergo copyediting, typesetting, and review of the resulting proof
before it is published in its final form. Please note that during the production process
errors may be discovered which could affect the content, and all legal disclaimers that
apply to the journal pertain.
ACCEPTED MANUSCRIPT

Enhanced intranasal delivery of mRNA vaccine by overcoming the nasal


epithelial barrier via intra- and paracellular pathways
Man Li, Mengnan Zhao, Yao Fu, You Li, Tao Gong, Zhirong Zhang, Xun Sun*

Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy,

T
IP
Sichuan University, Chengdu, 610041, People’s Republic of China

R
*Corresponding author: Xun Sun

SC
Key Laboratory of Drug Targeting, Ministry of Education, Sichuan University, No. 17.

NU
Section 3. Southern Renmin Road, Chengdu 610041, People’s Republic of China.

Tel: 86-28-8550 2037; Fax:86-28-85501615


MA
Email: xunsun22@gmail.com (X.Sun)
D
P TE
CE
AC

1
ACCEPTED MANUSCRIPT

Abstract

Facing the threat of highly variable virus infection, versatile vaccination systems are urgently

needed. Intranasal mRNA vaccination provides a flexible and convenient approach.

T
However, the nasal epithelium remains a major biological barrier to deliver antigens to nasal

IP
associated lymphoid tissue (NALT). To address this issue, a potent polymer-based intranasal

R
mRNA vaccination system for HIV-1 treatment was synthesized using cationic

SC
cyclodextrin-polyethylenimine 2k conjugate (CP 2k) complexed with anionic mRNA

encoding HIV gp120. The delivery vehicle containing CP 2k and mRNA overcame the

NU
epithelial barrier by reversibly opening the tight junctions, enhanced the paracellular delivery
MA
of mRNA and consequently minimized absorption of toxins in the nasal cavity. Together with

the excellent intracellular delivery and prolonged nasal residence time, strong system and

mucosal anti-HIV immune responses as well as cytokine productions were achieved with a
D
TE

balanced Th1/Th2/Th17 type. Our study provided the first proof of evidence that cationic

polymers can be used as safe and potent intranasal mRNA vaccine carriers to overcome the
P

nasal epithelial barrier. The safe and versatile polymeric delivery system represents a
CE

promising vaccination platform for infectious diseases.


AC

Graphic Abstract

2
ACCEPTED MANUSCRIPT

The self-assembled nanocomplex formulated with CP 2k could facilitate the delivery of


mRNA vaccine through intracellular and paracellular pathways, and consequently induce
both systemic and mucosal immune responses.

T
Key Words:

IP
mRNA vaccination, CD-PEI, nasal epithelial barrier, intracellular delivery, paracellular

R
delivery

SC
1. Introduction

NU
Vaccination has by far been the most effective approach to prevent the rapid spread of
MA
infectious diseases, such as human immunodeficiency virus infection and acquired immune

deficiency syndrome (HIV/AIDS) [1-3]. Great efforts have been contributed to the design of
D

HIV vaccines, and some showed promising results [4, 5]. Nonetheless, the development of
TE

potent vaccine still faces great challenges due to the high mutation rate and viral diversity of

infectious diseases. In immunotherapy, a universally applicable vaccine may no longer


P

provide sufficient protection due to the unique mutation frequently occurring in patients.
CE

Vaccination with mRNA offers a viable solution to this problem, which can be individually
AC

tailored and rapidly manufactured on demand [6]. Unlike plasmid DNA, mRNA can transfect

both dividing and non-dividing cells without entering the nucleus, resulting in higher gene

expression [7-10]. Techniques for effectively transfecting mRNA into cells have led to the

development of various mRNA-based vaccination systems [11-13]. For infectious diseases

with high mutation rates such as influenza, mRNA vaccine can be produced reliably in a

scalable process thus allowing quick response to emergence of pandemic strains and

consequently induce long-lived and protective immunity [14]. Therefore, mRNA vaccines are

of great significance in the prevention of variable virus infection [15]. Currently, mRNA

vaccines are injected intravenously [16], subcutaneously [17], or intradermally [18] in

3
ACCEPTED MANUSCRIPT

preclinical and clinical studies, which requires specially trained personnel. Also, the

injection-associated pain and acute infections render the administration of mRNA less

patient-friendly, which highlights the need to explore non-invasive routes of administration

T
for mRNA vaccine delivery.

IP
The U.S. Food and Drug Administration approved the first intranasal vaccine against

R
influenza (FluMist®) in 2003 [19]. As an alternative to systemic vaccine delivery, nasal

SC
immunization represents an effective and safe approach as compared to injection. The nasal

cavity is the portal of entry for many pathogens, and nasal-associated lymphoid tissue

NU
(NALT) is rich in antigen-presenting cells (APCs), T and B lymphocytes [20], making NALT
MA
a suitable site for antigen internalization and subsequent mucosal and systemic immune

responses [21]. Moreover, intranasal vaccination can induce immune response in distal
D

mucosal tissues such as in the vagina, because most mucosal lymphocytes are functionally
TE

connected [22]. This may be particularly helpful when vaccinating against sexually

transmitted pathogens such as HIV. Despite these advantages, intranasal mRNA vaccination
P

faces following obstacles: the presence of enzymes in the nasal cavity may degrade mRNA;
CE

cilia movement may accelerate the clearance of mRNA antigen and shorten its nasal
AC

residence time, resulting in poor antigen uptake. Although the nasal cavity contains APCs,

the nasal epithelium presents a major barrier to antigen delivery to these cells. Thus,

overcoming the nasal epithelial barrier is the key to achieve enhanced immune responses. To

ensure efficient delivery to APCs, multiple routes can be explored including (i) intracellular

route via transfection of epithelial cells [23], (ii) transcytotic route of delivery via M cells to

APCs such as dendritic cells and macrophages, and (iii) paracellular route of delivery via

tight junctions to reach underlying APCs [24].

Particulate delivery systems are proven efficient carriers for vaccine delivery [25], and

the most frequently used systems are lipoplex [26, 27] and liposomes [28-31]. While these

4
ACCEPTED MANUSCRIPT

lipid-based nano carriers can deliver mRNA efficiently, the epithelial barrier remains a

challenge to intranasal delivery of mRNA vaccines. Thus, overcoming the epithelial barrier

will likely lead to enhanced uptake of mRNA vaccine and stronger immune responses.

T
However, lipid-based carriers alone are unable to open the epithelial tight junctions to induce

IP
immune responses. To solve this problem, polymeric delivery systems offer a viable solution.

R
Polyethyleneimine (PEI) with a molecular weight of 25 kDa was proven to open the epithelial

SC
tight junctions [32], and has been shown as an efficient mRNA carrier in in vitro transfection

[33]. Nevertheless, PEI 25k-mRNA complexes showed poor colloidal stability and high

NU
cytotoxicity in mouse fibroblasts [33]. To overcome the epithelial barrier and enhance the
MA
mucus binding efficiency of PEI 25k, structural modification of low-molecular-weight PEI

with bioadhesive sugar moieties represents a feasible approach. Cyclodextrin, as a


D

biocompatible pharmaceutical excipient, has been selected to modify PEI 2k. Cyclodextrin
TE

was demonstrated to cause minimum damage to mucosal integrity and enhance drug

permeation via the nasal epithelium by transiently and reversibly opening tight junctions [34,
P

35]. Moreover, linking cyclodextrin to PEI lowered the charge density of polyamine
CE

backbone, and consequently reduced the cytotoxicity of cationic PEI polymer while
AC

maintained a large number of protonatable groups, which led to high gene-delivery efficiency

of DNA [36]. Based on these properties, we assume that cyclodextrin-PEI (CP) polymer may

serve as an efficient carrier for nasal mRNA vaccine delivery.

Here, we report the synthesis of CP polymer and the fabrication of a CP polymer-based

intranasal delivery platform for mRNA vaccine. The HIV glycoprotein 120 (gp120) was

selected as a model antigen. CP 2k polymer and mRNA formed nanoscale complexes via

electrostatic interactions. We compared the ability of CD-PEI 2k (CP 2k) or PEI 25k to

deliver mRNA encoding gp120 and to induce gp120-specific immune responses. These

experiments were carried out in cultures of DC 2.4 cells, Madin-Darby canine kidney

5
ACCEPTED MANUSCRIPT

(MDCK) cells and Calu-3 cells to simulate the nasal epithelium. The in vivo interaction

between this delivery vehicle and the epithelial tight junctions, as well as immune responses

in mice intranasally immunized with mRNA complexes were investigated.

T
IP
2. Materials and methods

R
2.1 Materials and animals

SC
Branched polyethylenimine (PEI) with molecular weights of 2 or 25 kDa, β-cyclodextrin

(CD, molecular weight 1135), 1,1’-carbonyldiimidazole (CDI),

NU
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and chitosan (CS,

190-300 kDa, 75-85% deacetylated) were obtained from Sigma-Aldrich (St. Louis, MO,
MA
USA). Restriction enzymes were obtained from Fermentas (Thermo Fisher, USA). All other

reagents were of analytical purity.


D

BALB/c (female) mice 6-8 weeks old were obtained from Dashuo Biotechnology
TE

(Chengdu, China) and housed in a specific pathogen-free, light-cycled and


P

temperature-controlled facility. All experiments were approved by the Institutional Animal


CE

Care and Ethics Committee of Sichuan University.


AC

2.2 Synthesis of cyclodextrin-polyethylenimine conjugates

CP 2k was prepared as reported with some modifications [37]. Briefly, 12 ml of N,

N-dimethylformanide (DMF) containing β-cyclodextrin (0.84 g, 0.74 mmol) and CDI (1.60 g,

10.4 mmol) were stirred at room temperature for 1.5 h under nitrogen. The resulting CDI-CD

was purified by precipitation with cold diethyl ether and filtered to remove soluble impurities.

The product was dissolved in 10 ml of dimethylsulfoxide (DMSO) and stored at 4°C. Then

CDI-CD in 10 ml DMSO and 0.6 ml triethylamine (Et3N) were added dropwise with stirring

over 1.5 h to 18 ml of DMSO containing PEI 2k (9.0 g). Then the reaction was allowed to

6
ACCEPTED MANUSCRIPT

stand another 5 h. The resulting solution was dialyzed against distilled water for 3 days and

lyophilized overnight.

T
2.3 In vitro transcription of mRNA

IP
The DNA sequence of HIV gp120 (GenBank DQ667594.1) was synthesized by Sangon

R
(Shanghai, China); a Kozak consensus sequence was inserted, as were Hind III and Bam HI

SC
restriction sites at the ends. The sequence was then cloned into the pVAX1 vector (Life

Technologies, USA), which contains a T7 promoter and a poly(A) tail. A plasmid expressing

NU
the luciferase reporter gene was constructed by inserting the luciferase coding sequence into

the pVAX1 backbone. The recombinant plasmid was amplified and purified using an
MA
endo-free plasmid kit (Omega, USA) and linearized using Xba I. Synthetic mRNA was

prepared by in vitro transcription using the T7 high yield RNA synthesis kit and an
D

m7G(5')ppp(5')G RNA cap structure analog (New England Biolabs, MA, USA) according to
TE

the manufacturer’s instructions. The mRNA product was precipitated with phenol/chloroform
P

and re-suspended in RNase-free water. The concentration of mRNA was determined by


CE

measuring the absorbance at 260 nm. After incubating the purified transcription products at
AC

70°C for 10 min, mRNA purity and size were analyzed by running an aliquot together with

RiboRuler High Range RNA Ladder (Fermentas, Thermo Fisher, USA) on agarose gels and

staining with SYBR green II dye. Labeling of mRNA with Cy3 or fluorescein was performed

using the Label IT nucleic acid labeling kit (Mirus Bio, WI, USA) according to the

manufacturer’s protocol.

2.4 Formulation and characterization of CP 2k/mRNA

Cationic CP 2k was dissolved in sterile, distilled RNase-free water to a concentration of

1 mg/ml. The CP 2k/mRNA delivery vehicle was formed by adding pre-diluted CP 2k to

7
ACCEPTED MANUSCRIPT

pre-diluted mRNA at N/P ratios of 8, 16 or 24 (calculated as the molar ratio of nitrogen in

PEI portion of CP 2k/phosphate in RNA). The mixture was vortexed for 10 sec, then

incubated at room temperature for 20 min to allow particles to form. Size distribution and

T
zeta potential of the delivery vehicle were measured by photon correlation spectroscopy using

IP
a Zetasizer Nano ZS90 (Malvern Instruments, UK). Samples were diluted to 1 ml using

R
RNase-free distilled water and equilibrated for 30 sec before measuring at a fixed angle.

SC
Measurements were made in triplicate.

CP 2k/mRNA morphology was analyzed by transmission electron microscopy (TEM,

NU
H-600, Hitachi, Japan). Samples were loaded on a copper grid and stained with
MA
phosphotungstic acid (1%) for 20 sec before observation.

The ability of CP 2k to bind RNA was checked using agarose gel electrophoresis. CP

2k/mRNA complexes were prepared as described above with 1 μg mRNA, mixed with 5 ×
D
TE

RNA loading buffer (Tiangen, Shanghai, China), and electrophoresed on a 4% agarose gel.

The gel was run at 120 V for 30 min and then visualized and photographed using the
P

ChemiDoc™ XRS system (Bio-Rad, Hercules, CA).


CE

To assess the ability of CP 2k to protect the complexed mRNA from RNase degradation,
AC

RNase (Sigma, USA) was incubated with naked gp120 mRNA (10 μg) or the equivalent

amount of mRNA complexed with CP 2k at the optimal ratio N/P 16. At the indicated time

points, EDTA was added to a final concentration of 10%, and the mixture was denatured at

65°C for 5 min. Then mRNA was released from the CP 2k/mRNA by adding 30 U of heparin,

and the released mRNA was isolated by phenol/chloroform precipitation. Levels of

gp120-mRNA were measured as Ct values in quantitative Real-Time PCR (qRT-PCR) using

the SsoFastTM EvaGreen Supermix on an iCycler iQTM 5 system (Bio-Rad, USA).

2.5 Cell culture

8
ACCEPTED MANUSCRIPT

Madin-Darby canine kidney (MDCK) cells, Calu-3 human lung adenocarcinoma cells

and DC 2.4 murine dendritic cells were obtained from the cell bank at the Chinese Academy

of Science (Shanghai, China). Cells were maintained in Dulbecco’s Modified Eagle Medium

T
High Glucose containing L-glutamine, or in RPMI 1640 medium (Hyclone, Life Technology,

IP
USA) supplemented with 10% fetal bovine serum (FBS; Gibco, USA), and 1%

R
penicillin/streptomycin.

SC
2.6 In vitro transfection

NU
DC 2.4, Calu-3 and MDCK cells were transfected with mRNA encoding luciferase
MA
(mLuc), which was produced by in vitro transcription as described above. CP 2k/mLuc

containing 1 μg luciferase mRNA was prepared at N/P ratios of 8, 16 or 24 (calculated as the


D

molar ratio of nitrogen in PEI portion of CP 2k to phosphate in DNA). In parallel, PEI


TE

25k/mLuc was prepared at N/P ratios of 8 or 16. Cells were seeded in 24-well plates at a

density of 1 × 105 cells/well and incubated for 24 h before transfection. Cells were incubated
P

in serum-free medium for 4 h after transfection, the cell culture medium was replaced with
CE

0.5 ml of fresh complete culture medium, and the cells were incubated another 20 h at 37°C
AC

in a 5% CO2 incubator. Luciferase expression was measured using a luciferase assay system

(Promega, WI, USA) following the manufacturer’s instructions. The total protein

concentration in transfected cells was determined using the BCA protein assay kit (Pierce,

Thermo Fisher, MA, USA).

2.7 Endosome escape assay

DC 2.4 cells were seeded on coverslips in 6-well culture plates for 24 h, then treated

with CP 2k/fluorescein-labeled mRNA (2 μg/well). At 2 or 6 h, cells were washed three times

with phosphate-buffered saline (PBS) containing 0.015% (w/v) heparin and stained with

9
ACCEPTED MANUSCRIPT

LysoTracker (Beyotime, China) for 1 h at 37°C. Then samples were washed with PBS and

observed using a confocal laser scanning microscope (FV1000, Olympus, USA).

T
2.8 Nasal residence time

IP
CP 2k or PEI 25k were complexed with Cy3-labeled mRNA, and administered

R
intranasally to mice under anesthesia with intraperitoneal injection of 1% (w/v) sodium

SC
pentobarbital (5 μg Cy3-mRNA per animal). In parallel, some animals were given naked

Cy3-mRNA. All mice were scanned at 0, 0.5, 1, 2 and 3 h after administration using an

NU
IVIS® Spectrum imaging system (Caliper Life Sciences, Hopkinton, MA, USA). Absolute
MA
fluorescence in the nasal cavity was quantified over time using an excitation wavelength of

548 nm and an emission wavelength of 562 nm. Relative fluorescence was expressed as the
D

percentage of the initial fluorescence in the nasal cavity.


TE

2.9 In vivo uptake


P

CP 2k or PEI 25k containing 5 μg Cy3-labeled mRNA were administered intranasally to


CE

mice under anesthesia with intraperitoneal injection of 1% (w/v) sodium pentobarbital. As a


AC

control, some animals were given naked Cy3-mRNA. At 6 h after administration, mice were

sacrificed by neck dislocation, and nasal epithelial cells were isolated as described [38]. Cells

were digested by incubating for 2 h at 37°C with type IV collagen. Mouse palates were

excised using a No. 22 scalpel blade, the palates were gripped behind the incisor teeth with

fine forceps and gently pulled toward the molar teeth. The NALT was teased gently into the

medium to release cells. After washing nasal epithelial cells and NALT cells with PBS, the

cells were centrifuged, re-suspended in PBS, and analyzed by flow cytometry to measure

uptake of Cy3-mRNA.

10
ACCEPTED MANUSCRIPT

2.10 In vivo toxicity

To investigate whether long-term intranasal administration of the polymer/mRNA

complex can cause absorption of toxins present in the nasal cavity, toxicity was analyzed in

T
vivo using a procedure similar to that described [39]. Female BALB/c mice weighing

IP
18.0-22.0 g were randomly divided into five groups (five mice/group). Mice received

R
once-daily intranasal doses of CP 2k/mRNA or PEI 25k/mRNA (10 μg mRNA, optimal ratio

SC
N/P 16) or double-distilled water, followed by LPS (5 mg/kg) on days 1, 3, 5 and 7. Negative

control animals received no treatment. Positive control animals received one intraperitoneal

NU
injection of LPS (5 mg/kg). Blood samples were collected via retro-orbital puncture on day 8.
MA
Livers were removed and sectioned for histology analysis.
D

2.11 Influence on tight junctions in an in vitro epithelial cell model


TE

The effect of different polymer/mRNA complexes on tight junction opening were

analyzed on in vitro MDCK cell model. Briefly, 2 × 105 MDCK cells were seeded into the
P

apical chamber of 24-well transwell inserts (3 μm Transwell® inserts, Corning, USA) and
CE

incubated in a 5% CO2 incubator at 37°C with refreshing of apical and basolateral media
AC

every 2 days. The transepithelial electrical resistance (TEER) was monitored using Millipore

ERS Ohm meter (Millipore, USA). The tight junction formation was identified when the

TEER reached 200 Ω·cm2. Complete medium in both apical and basolateral chambers were

replaced with serum free media for 30 min before experiment. The mRNA complexes were

prepared with 1 μg luciferase encoding RNA at the optimal ratio with CP 2k or PEI 25k and

added into the apical chamber with serum free media. Chitosan (CS, 0.02 mg/ml) was used as

positive control. After 1h of incubation, both apical and basolateral chambers were washed

with Hank’s balanced salt solution (HBSS) and cells were cultured with complete culture

media for another 11 h. For quantitative analysis, cells were collected at 1, 6 and 12 h; total

11
ACCEPTED MANUSCRIPT

RNA was extracted and levels of ZO-1 mRNA were assayed by qRT-PCR. Besides, the

TEER were monitored at prearranged time points to evaluate the recovery of epthelial tight

junctions (n = 3).

T
IP
2.12 Animal immunization

R
Female BALB/c mice aged 6-8weeks were randomized into 5 groups (5 animals per

SC
group), and immunized twice with mRNA encoding HIV gp120 at an interval of 2 weeks.

Mice were anesthetized with 1% (w/v) sodium pentobarbital and given intranasal doses of CP

NU
2k/mRNA, PEI 25k/mRNA or naked mRNA (10 μg mRNA/mouse). As negative controls,
MA
mice received no treatment or unrelated mRNA encoding luciferase complexed with CP 2k,

which excludes effects of RNA-mediated immune stimulation. Mice were held upright for
D

some time to ensure maximal dosing and to prevent swallowing. At 10 days after the second
TE

immunization, all mice were sacrificed by neck dislocation, and the immune response was

analyzed.
P
CE

2.13 Serum and mucosal antibody response


AC

ELISA was used to measure serum levels of antigen-specific antibodies (IgG total, IgG1

and IgG2a), as well as levels of secreted IgA antibodies in nasal and vaginal mucosal washes.

Polystyrene 96-well plates were coated overnight at 4°C with 100 μl/well of 1 μg/ml HIV

gp120 Con_B (Immune Technology, Suzhou, China). Plates were washed with PBS

containing 0.1% Tween-20 (PBST), then incubated with 100 μl of serial dilutions of serum or

mucosal washes; dilutions for assay of IgG, IgG1 and IgG2 arranged from 1:8 to 1:512.

Plates were washed, incubated with 100 μl of horseradish peroxidase-conjugated anti-mouse

IgG antibody (diluted 1:10000 for IgG or 1:5000 for IgG1 and IgG2a) or IgA antibody

(diluted 1:5000). TMB substrate (BD PharMingen, CA, USA) was added and the reaction

12
ACCEPTED MANUSCRIPT

was stopped with 2M H2SO4. Optical density was measured at 450 nm using a microplate

reader (VarioSkan, Thermo Fisher Scientific, MA, USA).

T
2.14 Cytotoxicity T lymphocyte (CTL) assay

IP
An in vivo CTL killing assay was used to measure the cytotoxicity of antigen-specific

R
CD8+ T cells [40]. Splenocytes were isolated from naïve mice. Red blood cells were lysed

SC
using ACK buffer, and the cell suspension was divided equally into two aliquots. One was

incubated for 2 h at 37°C with 2 μg/ml HIV gp120 peptide and the other with medium only.

NU
Then the peptide-pulsed cells were stained for 10 min at 37°C with 2 μM carboxyfluorescein
MA
succinimidyl ester (CFSE; Invitrogen, CA, USA), while the non-pulsed cells were stained

with 0.2 μM CFSE. CFSE labeling was then quenched by addition of FBS to a final

concentration of 20% (v/v). The two aliquots (1 × 107 cells/100 μl) were mixed equally, and
D
TE

the cell suspension was injected into the tail vein of untreated (control) or immunized mice.

After 18 h, splenocytes from recipient mice were prepared and analyzed by flow cytometry.
P

The percentage of specific lysis was calculated as follows:


CE

  ratio of CFSE low / CFSE high cells re cov ered from naive mice  
Specific Lysis  %   100  1   low high 
  ratio of CFSE / CFSE cells re cov ered from immunized mice  
AC

2.15 Intracellular cytokine staining

Ten days after second immunization, mice were sacrificed by neck dislocation.

Splenocytes were harvested by homogenizing through a 70-μm cell strainer (BD Falcon) in

10% RPMI 1640 medium, and red blood cells were lysed using ACK lysis buffer (0.15 M

NH4Cl, 10.0 mM KHCO3, 0.1 mM EDTA, pH 7.4). Splenocytes were washed, cultured in

RPMI 1640 medium supplemented with 10% FBS, 2-mercaptoethanol and 2 μg/ml HIV

gp120 peptide sodium from the consensus B V3 region

(TRPNNNTRKSIHIGPGRAFYTTGEIIGDIRQAH; Kaijie, Chengdu, China). After 1-h

13
ACCEPTED MANUSCRIPT

incubation, brefeldin A (eBioscience, CA, USA) was added to each sample to a final

concentration of 1 μg/ml and incubated another 5 h at 37°C. Cells were washed, then

incubated at 4°C with flourescein isothiocyanate (FITC)-conjugated anti-mouse CD8a

T
antibody or FITC-conjugated anti-mouse CD4 antibody (eBioscience). Cells were washed

IP
again, fixed, and permeabilized with Fixation and Permeabilization Buffers (eBioscience).

R
Cells were centrifuged, then incubated in permeabilization buffer containing either

SC
polyethyleneimine (PE)-conjugated anti-mouse IFN-γ antibody or PE-conjugated anti-mouse

IL-4antibody. Cells were washed, diluted in PBS containing 1% BSA, and analyzed by

NU
two-color flow cytometry (Beckman Coulter, CA, USA). Percentages of cells positive for
MA
CD8+/IFN-γ+ and CD4+/IL-4+were determined using Kaluza software (Beckman Coulter, CA,

USA).
D
TE

2.16 Cytokine production by ELISA

IFN-γ and IL-4 production in splenocytes culture supernatants and NALT supernatants
P

were measured by ELISA (BD PharMingen, CA, USA) according to the manufacturer’s
CE

protocol. The splenocytes isolated from immunized mice were cultured in RPMI 1640
AC

medium with supplementary 10% FBS in 96-well plates (1 × 105 cells/well) with the presence

of 2 μl/ml HIV gp120 peptide and incubated for 72 hours. Then the supernatants were

collected by centrifugation. Each sample was examined in duplicate.

To analyze the cytokine production in NALT culture supernatant, the palate of

immunized mice were isolated. After washing with RPMI 1640 medium supplemented with

10% FBS, the palate was incubated with 200 μl RPMI 1640 medium in 96 well plate. After

48 hours’ incubation, the supernatant was collected to determine the content of IFN-γ and

IL-4 produced by NALT cells.

14
ACCEPTED MANUSCRIPT

2.17 Cytokine productions by qRT-PCR

Quantitative RT-PCR (qRT-PCR) was used to assay production of Th1 cytokines (IFN-γ

and IL-2), Th2 cytokines (IL-4 and IL-10), Th17 cytokine (IL-17) and type I IFN cytokines

T
(IFN-α4 and IFN-β1) by splenocytes from immunized mice. Total RNA was extracted from

IP
spleens using an RNA isolation kit (TianGen, China), it was reverse-transcribed using the

R
TIANscript RT kit, and cytokine mRNA levels were analyzed using SsoFast TM EvaGreen

SC
Supermix on an iCycler iQTM 5 system (Bio-Rad, USA). β-actin served as the internal

control.

NU
MA
2.18 Statistical analysis

Experiments were performed at least in triplicate unless otherwise noted. The data are

shown as the mean ± SD. Statistical difference was analyzed by Student’s t test and one-way
D
TE

ANOVA with Bonferroni post hoc test. All tests are accepted as statistically significant when

the p value is less than 0.05.


P
CE

3. Results
AC

3.1 Synthesis and characterization of CP 2k polymer

β-cyclodextrin-PEI 2k (CP 2k) was synthesized (Figure 1A) with a molar ratio of

cyclodextrin:PEI of 0.36:1, and the structure was verified by 1HNMR. Signals from ethylene

protons in PEI (–CH2CH2NH–) appeared at 2.4–3.0 ppm. Signals assigned to the hydrogens

C1 and C2-C6 of β-cyclodextrin were observed at 5.1 and 3.0-4.0 ppm, respectively

(Supporting Information Figure S1). The molecular weight of CP 2k was 80 kDa based on gel

permeation chromatography.

15
ACCEPTED MANUSCRIPT

T
R IP
SC
NU
Figure 1. Synthesis of CP 2k and formation of CP 2k/mRNA. (A) Synthesis of CP 2k. (B)
Formation of CP 2k/mRNA. CP 2k and mRNA were dissolved in sterile, distilled RNase-free
water. Particles were formed by electrostatic interaction.
MA
3.2 Formulation and characterization of the CP 2k/mRNA
D

HIV gp120 mRNA was transcribed and identified (Figure 2A). CP 2k and HIV gp120
TE

mRNA were complexed by electrostatic interaction (Figure 1B). CP 2k completely

condensed mRNA even at an N/P of 8, where N/P refers to the molar ratio of nitrogen in PEI
P
CE

to phosphate in DNA. This complete condensation was observed as slowed mobility on

agarose gels in the absence of any free mRNA band (Figure 2B). The optimal ratio of CP 2k
AC

to mRNA was found to be N/P 16, based on an in vitro transfection assay. The average size

of CP 2k/mRNA was 117.3 ± 3.44 nm at N/P 16; and they showed spherical morphology

with a ζ-potential of 26.4 ± 2.8 mV (Figure 2C and Table 1). The ability of CP 2k to protect

the mRNA from RNases was measured using qRT-PCR. Even after 4 h incubation with

RNase, a low Ct value was detected for CP 2k/mRNA , indicating a relatively high gp120

mRNA level (Figure 2D). These results suggest that the delivery vehicle could protect mRNA

from degradation in the nasal cavity.

16
ACCEPTED MANUSCRIPT

T
R IP
SC
NU
MA
Figure 2.Characterization of the CP 2k/mRNA . Particles were formed by adding mRNA
solution to a CP 2k solution at N/P ratios 8, 16 and 24 (calculated as the molar ratio of
nitrogen in PEI portion of CP 2k/phosphate in DNA) with gentle mixing. (A)
D

Characterization of the 1600-nt (nucleotides) mRNA by gel electrophoresis. (B) Gel


TE

retardation assay to detect condensation of mRNA into CP 2k at different N/P ratios. (C) Size
distribution and morphology of CP 2k/mRNA formed at an N/P ratio of 16. The optimal ratio
was determined by in vitro transcription, and morphology was examined by transmission
P

electron microscopy. Scale bar, 200 nm. (D) Ability of CP 2k to protect mRNA from
nucleases. Heparin was used to release mRNA from complexes, and mRNA levels were
CE

determined using qRT-PCR targeting the gp120 coding region (n = 3).


AC

Table 1. Size and ζ potential of CP 2k/mRNA complex at different N/P ratios (n=3)
Size (nm) PDI ζ potential (mV)
CP2k:mRNA (N/P=8) 137.2 ± 5.43 0.203 ± 0.031 13.1 ± 2.3
CP2k:mRNA (N/P=16) 117.3 ± 3.44 0.161 ± 0.028 26.4 ± 2.8
CP2k:mRNA (N/P=24) 112.4 ± 3.56 0.189 ± 0.025 27 ± 3.1

3.3 Enhanced mRNA transfection by CP 2k/mRNA delivery vehicle

To assess the transfection efficiency of the CP 2k/mRNA complex, mRNA encoding

luciferase (mLuc) was used, and complexes were transfected into DC 2.4 murine dendritic

cells; MDCK epithelial cells, which form tight junctions [41]; and Calu-3 bronchial epithelial

cells [42]. The CP 2k/mLuc complex efficiently transfected all three cell lines, and the

17
ACCEPTED MANUSCRIPT

highest luciferase expression was achieved at N/P 16. Under these conditions, luciferase

expression was significantly higher than that observed with PEI 25k/mLuc in all three cell

lines (p < 0.01; Figure 3A and 3B; Supporting Information Figure S2). In addition, CP

T
2k/mLuc (N/P 16) was less cytotoxic than PEI 25k/mLuc to DC 2.4 and MDCK cells, based

IP
on MTT assays (Supporting Information Figure S3).

R
After internalization, CP 2k/mRNA was expected to escape from the endo/lysosome to

SC
release mRNA cargo into the cytoplasm. To examine this process, we used confocal laser

scanning microscopy to track fluorescein-labeled mRNA (green fluorescence) complexed

NU
with CP 2k. At the same time, we localized endo/lysosomes using LysoTracker (red
MA
fluorescence). Co-localization of mRNA and endo/lysosomes was detected as yellow

fluorescence. After 2 h incubation following transfection into DC 2.4 cells, most


D

fluorescein-mRNA co-localized with endo/lysosomes. After 6 h incubation, more green


TE

fluorescence was observed in the cytoplasm than in the endo/lysosome (Figure 3C), which

indicates that CP 2k/mRNA escaped from the endo/lysosome after internalization thus
P

facilitating the expression of mRNA in the cytoplasm. The successful endosomal escape of
CE

our delivery vehicle may reflect the potent buffering capacity of PEI, which allows mRNA to
AC

escape via the “proton sponge” mechanism [43] by increasing ionic concentration and

eventully rupturing the endosome membrane.

18
ACCEPTED MANUSCRIPT

T
R IP
SC
NU
MA
D
P TE
CE

Figure 3. CP 2k facilitates in vitro gene expression and endosomal escape of mRNA vaccine.
At 24 h after transfection, expression of the luciferase reporter gene was analyzed in (A) DC
2.4 cells, (B) MDCK cells (n = 3, **p < 0.01). (C) Release of CP 2k from endosomes in DC
2.4 cells was analyzed using confocal laser scanning microscopy. DC 2.4 cells were
AC

incubated for the indicated times with fluorescein-mRNA (green) complexed with CP 2k
(CP2k/m) and then stained with LysoTracker (red). Co-localization of fluorescein-mRNA
and endosome/lysosomes appears as yellow fluorescence. Scale bar, 10 μm.

3.4 Prolonged nasal residence time and efficient in vivo delivery of mRNA by CP 2k

Given the promising intracellular results, we attempted to explore the in vivo behavior of

the delivery vehicle. Prolonged nasal retention time may increase the probability of in vivo

uptake. CP 2k containing Cy3-labeled mRNA encoding gp120 were administered intranasally

to BALB/c mice. In vivo imaging was used to determine whether condensing mRNA with CP

could prolong retention time in the nasal cavity (Figure 4A). Nasal residence time was only 2

19
ACCEPTED MANUSCRIPT

h for naked mRNA, whereas it was 3 h for CP 2k/mRNA, at which time residual fluorescence

was still 23.8 ± 4.52% of the initial level. In contrast, residual fluorescence for PEI

25k/mRNA was 17.7 ± 6.24% at 2 h and it was barely detectable at 3 h. These results suggest

T
that the cationic CP 2k polymer shows stronger mucosal adhesion than PEI 25k, leading to

IP
longer nasal retention time of the mRNA, which further increases uptake by NALT and by

R
nasal epithelial cells. Mean fluorescence due to Cy3 was 8.98 ± 0.31 in NALT and 10.34 ±

SC
1.34 in nasal epithelial cells, indicating significantly greater uptake for CP 2k/mRNA than for

either PEI 25k/mRNA or naked mRNA (p < 0.05, Figure 4B).

NU
MA
D
P TE
CE

Figure 4. CP 2k facilitates in vivo delivery of mRNA vaccine and prolongs nasal residence
time. (A) Relative fluorescence intensity of Cy3-mRNA complexed with CP 2k (CP
AC

2k/mRNA) or PEI 25k (PEI 25k/mRNA) in the nasal cavity was determined as the percentage
of initial fluorescence (n = 4). (B) In vivo uptake efficiency of Cy3-mRNA complexed with
CP 2k (CP2k/m) or PEI 25k (PEI25k/m) was determined by flow cytometry at 6 h after
intranasal administration (n = 4, *p < 0.05).

3.5 Efficient delivery of mRNA vaccine through the epithelial barrier with higher safety

One potential disadvantage of using polymers to deliver mRNA is that they may

facilitate bioabsorption of toxins that may be present in the nasal cavity. To examine this

problem in CP 2k, we co-administered CP 2k/mRNA, PEI 25k/mRNA (encoding HIV gp120)

or double-distilled water and lipopolysaccharide (LPS) into mice every other day over 7 days.

LPS absorption into the nasal cavity should lead to hepatotoxicity. Serum levels of alanine

20
ACCEPTED MANUSCRIPT

transaminase (ALT) were higher in animals given PEI 25k/mRNA + LPS than in those given

CP 2k/mRNA + LPS or double distilled water + LPS (p < 0.05, Figure 5A). In contrast,

serum levels of ALT and aspartate transaminase (AST) were similar between animals treated

T
with CP 2k/mRNA + LPS or with double-distilled water + LPS. Liver sections were analyzed

IP
for histological changes. Groups treated with CP 2k/mRNA or double-distilled water showed

R
no histological differences from untreated animals, while animals treated with PEI

SC
25k/mRNA exhibited obvious cell swelling (Figure 5B, arrows). As a positive control,

animals were injected intraperitoneally with LPS, and they showed severe liver damage,

NU
inflammatory infiltration and necrosis (Figure 5B, arrows). These results suggest that
MA
intranasally administered CP 2k/mRNA does not facilitate nasal absorption of LPS, whereas

PEI 25k/mRNA permits it enough to lead to moderate liver damage.


D

Next we sought to understand why PEI 25k, but not CP 2k, promoted LPS absorption.
TE

We examined the effects of CP 2k/mRNA and PEI 25k/mRNA on epithelial tight junction

integrity in an in vitro epithelial MDCK cell model. We focused on tight junction protein
P

ZO-1, which plays a vital role in maintaining the defense function of epithelium [44].
CE

Treating MDCK cells with either type of delivery vehicle significantly lowered ZO-1 protein
AC

expression 6 h later, based on qRT-PCR and immunofluorescence (Figure 5C). Surprisingly,

by 12 h after treatment with CP 2k/mRNA, the level of ZO-1 protein recovered and normal

tight junction morphology was restored. The ZO-1 protein remained disassembled even at 12

h after treatment with PEI 25k/mRNA. Measurements of TEER during 12 h of incubation

were consistent with these observations (Supporting Information Figure S4). These results

suggest that while PEI 25k irreversibly alters tight junction integrity, CP 2k reversibly opens

tight junctions, thereby promoting intranasal delivery through paracellular pathways while

still maintaining the integrity of the epithelial barrier.

21
ACCEPTED MANUSCRIPT

T
R IP
SC
NU
Figure 5. In vivo toxicity and paracellular delivery of polymer/mRNA . (A) Serum levels of
MA
ALT and AST were determined in mice intranasally given CP 2k/gp120-mRNA (CP2k/m),
PEI 25k/gp120-mRNA (PEI25k/m) or double-distilled H2O together with LPS (5 mg/kg)
every other day for 7 days (mean ± SD, n = 5, *p < 0.05). (B) Liver sections of mice treated
with polymer/mRNA delivery vehicles and LPS. Arrows indicate pathological changes such
D

as cell swelling or necrosis (n = 5, 200 ×). (C) ZO-1 protein expression in an in vitro
epithelial cell model was analyzed by qRT-PCR after treatment with CP 2k/mRNA (CP2k/m)
TE

or PEI 25k/mRNA (PEI25k/m). Results were expressed as normalized fold expression (n =


3).
P

3.6 Enhanced humoral and cellular immune responses induced by CP 2k/mRNA


CE

To test whether the higher transfection efficiency, longer nasal residence time and
AC

superior epithelial permeation profile of CP 2k/mRNA translated into stronger immune

responses, mice were intranasally vaccinated twice 2 weeks apart. Titers of HIV

gp120-specific IgG and two IgG subtypes IgG1 and IgG2a were assayed in BALB/c mice 10

days after the second immunization in order to evaluate the humoral immune response. CP

2k/mRNA elicited significantly more IgG production than either PEI 25k/mRNA or naked

mRNA (p < 0.01, Figure 6A). Naked mRNA and luciferase mRNA complexed with CP 2k

(CP 2k/mLuc) induced barely detectable levels of gp120-specific antibodies. CP 2k/mRNA

also induced significantly higher titers of IgG1 and IgG2a than did naked mRNA (p < 0.05,

Figure 6B and 6C). Since mucosal vaccines administered at one site have been shown to elicit

22
ACCEPTED MANUSCRIPT

an immune response in distal mucosal tissues via movement of effector cells between

mucosal tissues [27], we also assessed the immune response in both the nasal and vaginal

compartments (Figure 6D). The immune response in mucosa was measured by assaying

T
levels of secreted sIgA. CP 2k/mRNA induced greater sIgA secretion than PEI 25k/mRNA in

IP
vaginal washes (p < 0.01), while naked mRNA led to a relatively low level of sIgA secretion.

R
These results suggest that condensation processcan endow the mRNA with the ability to elicit

SC
sIgA secretion in distal mucosa, which suggests its potential use in vaccines against sexually

transmitted pathogens such as HIV. Animals treated with a CP 2k containing luciferase

NU
mRNA (CP 2k/mLuc) showed barely detectable sIgA levels in either the nasal or vaginal
MA
washes. This suggests that the observed sIgA induction was gp120-specific.
D
P TE
CE
AC

Figure 6. Humoral immune response induced by mRNA vaccination analyzed by ELISA.


BALB/c mice were intranasally immunized twice with 10 μg gp120-mRNA in naked form or
complexed with CP 2k or PEI 25k. Mice treated with unrelated CP 2k/mLuc were set as
negative control. At 10 days after the second immunization, samples of serum and of lavage
from nasal and vaginal cavities were obtained. (A) Serum titers of anti-gp120-specific IgG.
Serial two-fold dilutions of serum were analyzed. (B) Serum titers of IgG1 and IgG2a
subtypes. (C) Ratio of IgG1/IgG2a titers in different animal groups. (D) Levels of secreted

23
ACCEPTED MANUSCRIPT

IgA in nasal wash and vaginal wash. To obtain nasal washes, mice were washed with 1 ml of
PBS containing 1% BSA. The vaginal cavity of mice was washed twice with 150 μl of PBS
containing 1% BSA. Nasal washes were measured in the ELISA without further dilution,
while vaginal washes were diluted two-fold before ELISA (mean ± SD, n = 5, *p < 0.05, **p
< 0.01, n.s, non-significant)

T
Besides the humoral immune response, the induced cellular immune response was

IP
evaluated in mice by measuring gp120-specific cytotoxic T lymphocyte activity (CTL), as

R
well as CD8+ and CD4+ T cell responses. CTL activity was expressed as the percentage of

SC
gp120-specific cytotoxic lysis in vaccinated mice relative to that in untreated mice.

NU
Splenocytes were stained with carboxyfluorescein succinimidyl ester (CFSE) at 2 μM

(CFSEhigh) or 0.2 μM (CFSElow). The percentage was calculated from the ratio of the number
MA
of cells in the CFSEhigh population after pulsing with gp120 peptide to the number of cells in

the non-pulsed CFSElow population. CTL activity was 22.8 ± 1.31% after two intranasal
D

immunizations with CP 2k/mRNA, significantly greater than 14.4 ± 1.46% after


TE

immunization with PEI 25k/mRNA (p < 0.01, Figure 7A). CTL activity in these groups was
P

significantly higher than in the group vaccinated with naked mRNA (p < 0.05). Similar
CE

results were obtained when intracellular cytokine staining (ICS) was used to measure the

levels of gp120-specific CD8+ and CD4+ T cell responses. CP 2k/mRNA induced similar
AC

levels of CD8+ and CD4+ T cell responses, which were significantly higher than those

induced by PEI 25k/mRNA (p < 0.05, Figure 7B).

24
ACCEPTED MANUSCRIPT

T
R IP
SC
Figure 7. Cellular immune response induced by CP 2k/gp120-mRNA, PEI
25k/gp120-mRNA, naked mRNA, unrelated CP 2k/mLuc. Untreated mice were set as blank

NU
control. At 10 days after the second immunization, splenocytes were obtained. Cytotoxic T
lymphocyte activity (CTL) and Intracellular cytokine staining (ICS) were analyzed by flow
cytometry. (A) The percentage of CD+8 T cell lysis that was gp120-specific was calculated
MA
(mean ± SD, n = 5, *p < 0.05, **p < 0.01). (B) CD+8 T cells producing IFN-γ and CD4+ T
cells producing IL-4 were analyzed by ICS. Mean percentages of double-positive cells were
calculated as the percentage of cytokine-positive cells to total cells (n = 5, *p < 0.05).
D

3.7 CP 2k/mRNA induced a Th1/Th2/Th17 immune response and reduced mRNA


TE

immunogenicity
P

To assess the bias of induced immune responses, we analyzed the cytokine productions
CE

of polymer-based mRNA vaccine. Th1 cytokine IFN-γ and Th2 cytokine IL-4 in the

splenocytes culture supernatant and NALT culture supernatant were determined by ELISA.
AC

Moreover, qRT-PCR was used to assay levels of Th1 cytokines (IFN-γ and IL-2), Th2

cytokines (IL-4 and IL-10) and Th17 cytokine (IL-17) in splenocytes of immunized mice.

Traditional DNA vaccine was reported to induce a balanced Th1/Th2 immunity [4]. With our

mRNA vaccination regimen, CP 2k/mRNA led to higher production of Th1 and Th2

cytokines than either PEI 25k/mRNA or naked mRNA. CP 2k/mRNA also induced quite high

levels of Th17 cytokine, indicating that CP 2k/mRNA evoked a Th1/Th2/Th17 immune

response (Figure 8A-8E). Similar results were observed when levels of IFN-γ and IL-4 in

splenocytes culture supernatant were measured by enzyme-linked immunosorbent assay

(Figure 8A and 8B). IL-4 is involved in the maturation of B cells, and the secretion of IFN-γ

25
ACCEPTED MANUSCRIPT

plays a critical role in the elimination of HIV infected cells. Thus, the augmentation of IL-4

and IFN-γ, as well as other cytokines, provided strong evidence regarding the potency of this

mRNA vaccine delivery system.

T
Naked mRNA is known to activate TLR3/7 pathways [45, 46], and this adjuvanticity

IP
explains why naked mRNA encoding HIV gp120 induced cytokine production at the site of

R
intranasal administration of our mRNA vaccines. This induction was reflected in the

SC
up-regulation of co-stimulatory molecules (Supporting Information Figure S5), as well as the

high cytokine levels in NALT culture supernatant (Figure 8C and 8D). Naked mRNA has

NU
also been shown to induce innate immunity by activating the TLR3 signaling pathway,
MA
leading to secretion of type I interferon [47]. This poses a problem for mRNA vaccines

because TLR3 activation can lower transgene expression, hampering the induction of an
D

antigen-specific immune response [17]. In our experiments, CP 2k/mRNA led to lower type I
TE

interferon production than naked mRNA (Figure 8E). This suggests that antigen expression

of the mRNA may be higher when CP 2k polymer is used. In addition, either CP 2k/mRNA
P

or PEI 25k/mRNA led to 1- to 2-fold higher production of type I interferon than what was
CE

measured in untreated control mice. This suggests that the mRNA still triggers a slight innate
AC

immune response after condensation into polyplexes, which is desirable given that the innate

response provides initial defenses in pathogen invasion. These results indicate that CP

polymer-based polyplexes may achieve a balance between antigen-specific immune response

and innate immunity.

26
ACCEPTED MANUSCRIPT

T
R IP
SC
NU
MA
D
P TE
CE
AC

Figure 8. The cytokines produced by gp120-mRNA vaccine. Th1 cytokines (IFN-γ, IL-12)
and Th2 cytokines (IL-4, IL-10) were determined by ELISA and qRT-PCR. Ten days after
second immunization, the splenocytes of immunized mice were isolated and seeded into
96-well plates at a density of 1 × 105 cells/well. The IFN-γ (A) and IL-4 (B) in splenocytes
culture supernatant were analyzed (n = 5, *p < 0.05, **p < 0.01). The NALT of mice were
isolated and cultured with 200 μl RPMI 1640 medium for 48 h. The IFN-γ (C) and IL-4 (D)
in NALT culture supernatant were analyzed (n = 5, *p < 0.05). (E) Quantitative RT-PCR
measurement of cytokine production in splenocytes from vaccinated mice. Levels of
transcripts encoding Th1 cytokines (IFN-γ and IL-2), Th2 cytokines (IL-4 and IL-10) and
Th17 cytokine (IL-17) were determined using specific primers. In addition, levels of
transcripts encoding the type I interferons IFN-α4 and IFN-β1 were analyzed. Results were
expressed as normalized fold expression relative to the level in non-vaccinated animals (n =
5).

27
ACCEPTED MANUSCRIPT

4. Discussion

Here we explored the application of CP polymer in the field of intranasal mRNA

T
vaccination in an effort to overcome the nasal epithelial barrier and induce enhanced systemic

IP
and mucosal immunity. We provide in vivo evidence that CP polymer/mRNA administered

R
intranasally can efficiently deliver vaccine to distal mucosal sites, which is important for

SC
vaccines against mucosally transmitted diseases. These delivery vehicle do not appear to

suffer the short nasal residence time or clearance by cilia that can significantly reduce

NU
delivery of mRNA vaccines, and consequently enchanced the vaccine delivery across the
MA
nasal epithelial barrier [48]. Combining the high mucosal affinity of cyclodextrin[49] with

the good adjuvanticity of the cationic PEI polymer, stronger immune reponses were elicited.
D

[50].
TE

Our results suggest that CP polymer efficiently deliver their mRNA cargo by two

mechanisms. One is intracellular delivery into nasal epithelial cells: first these cells take up
P

the mRNA and translate the encoded antigen; then the transfected cells are recognized and
CE

lysed by natural killer cells, releasing the antigen; and finally the antigen is taken up by
AC

dendritic cells and cross-presented to T and B cells [23]. This intracellular mechanism was

elucidated by Frank Wegmann and colleagues, who showed that antigen complexed with PEI

25k could be taken up by DC cells and epithelial cells [50]. We also found that both nasal

epithelium and NALT took up larger amounts of mRNA delivered with CP 2k than with PEI

25k (Figure 4B).

Next, CP polymer were found to efficiently deliver their mRNA cargo into the

underlying NALT by a paracellular pathway in which the CP 2k polymer disassembles the

ZO-1 protein (Figure 5C), and reversibly opened tight junctions and lowering TEER

(Supporting Information Figure S4). This may allow efficient delivery of the mRNA to the

28
ACCEPTED MANUSCRIPT

underlying dendritic cells, which then express the antigen and consequently induce immune

responses. In contrast, we also provided evidence that PEI 25k irreversibly opened tight

junctions, and their integrity remained compromised at 12 h after treatment with PEI

T
25k/mRNA. As a result, co-administration of this delivery vehicle and LPS led to obvious

IP
liver damage (Figure 5A and 5B). The observation that CP 2k reversibly opens tight junctions

R
probably helps explain why it is much less toxic than PEI 25k.

SC
The mucoadhesive properties of polymers are critical to the efficiency of the carrier

system for an intranasal vaccine. In our experiments, the presence of cyclodextrin rendered

NU
CP polymer highly mucoadhesive which likely results in longer nasal residence time than PEI
MA
25k (Figure 4A). Moreover, good colloidal stability and mucus-binding ability are vital for

intranasal drug delivery systems [51]. The longer nasal residence time of our mRNA vaccine
D

may also reduce the risk that antigen is delivered to the lung or the central nervous system,
TE

reducing clearance and increasing antigen uptake. In terms of clinical application of

polymeric vaccine delivery systems, safety and efficacy are two major factors to be
P

considered. In our study, CP 2k polymer, synthesized by conjugating low-molecular weight


CE

PEI 2k with β-cyclodextrin, exhibited minimum cytotoxicity and kept the intactness of
AC

epithelium after intranasal vaccination, showing good biocompatibility. Currently, PEI-based

DNA delivery system has entered clinical trial for treatment of bladder cancer. Therefore, CP

2k may hold the promise for clinical application. Taken together, our results indicate that CP

2k circumvented the nasal epithelial barrier by facilitating intra- and paracellular delivery of

mRNA and prolonging the nasal residence time, and consequently elicited stronger mucosal

and systemic T cell and B cell responses.

5. Conclusion

Here, a simple and versatile intranasal mRNA vaccine delivery system was developed

29
ACCEPTED MANUSCRIPT

based on a cationic polymer consisting of β-cyclodextrin and low-molecular-weight PEI 2k,

which carries the high mucosal affinity of cyclodextrin and the good adjuvanticity of the

cationic PEI polymer. Our study provided the first in vivo evidence that CP polymer

T
administered intranasally efficiently delivered an mRNA vaccine across the nasal epithelial

IP
barrier and induced potent immune responses against HIV-1 gp120 model antigen without

R
causing absorption of toxins present in the nasal cavity. By reversibly opening tight junctions

SC
in the nasal epithelium, CP 2k not only facilitated paracellular antigen delivery but also

reduced absorption of toxins present in the nasal cavity. Intranasal inoculation of mice with

NU
CP 2k/mRNA led to strong mucosal and systemic immune responses in a balanced
MA
Th1/Th2/Th17 profile. Using CP 2k or PEI 25k, condensation of mRNA into particles also

reduced the ability of mRNA to stimulate an innate immune response, likely translating into
D

higher expression of the antigen encoded in the mRNA and therefore higher antigen-specific
TE

immunity against gp120. Thus, this cationic CP polymer represents an excellent platform

material for intranasal mRNA vaccination.


P
CE

Acknowledgement
AC

We acknowledge the financial support of the National Natural Science Foundation of China

(No.81173011, 81422044) and the National Science & Technology Major Project of China

(No. 2012ZX09304004).

REFERENCES
[1] W. Fischer, S. Perkins, J. Theiler, T. Bhattacharya, K. Yusim, R. Funkhouser, C. Kuiken, B. Haynes, N.L.
Letvin, B.D. Walker, B.H. Hahn, B.T. Korber, Polyvalent vaccines for optimal coverage of potential T-cell
epitopes in global HIV-1 variants, Nature medicine, 13 (2007) 100-106.
[2] H.X. Liao, R. Lynch, T. Zhou, F. Gao, S.M. Alam, S.D. Boyd, A.Z. Fire, K.M. Roskin, C.A. Schramm, Z.
Zhang, J. Zhu, L. Shapiro, J.C. Mullikin, S. Gnanakaran, P. Hraber, K. Wiehe, G. Kelsoe, G. Yang, S.M. Xia,
D.C. Montefiori, R. Parks, K.E. Lloyd, R.M. Scearce, K.A. Soderberg, M. Cohen, G. Kamanga, M.K. Louder,
L.M. Tran, Y. Chen, F. Cai, S. Chen, S. Moquin, X. Du, M.G. Joyce, S. Srivatsan, B. Zhang, A. Zheng, G.M.
Shaw, B.H. Hahn, T.B. Kepler, B.T. Korber, P.D. Kwong, J.R. Mascola, B.F. Haynes, Co-evolution of a
broadly neutralizing HIV-1 antibody and founder virus, Nature, 496 (2013) 469-476.

30
ACCEPTED MANUSCRIPT

[3] J. Huang, B.H. Kang, M. Pancera, J.H. Lee, T. Tong, Y. Feng, H. Imamichi, I.S. Georgiev, G.Y. Chuang, A.
Druz, N.A. Doria-Rose, L. Laub, K. Sliepen, M.J. van Gils, A.T. de la Pena, R. Derking, P.J. Klasse, S.A.
Migueles, R.T. Bailer, M. Alam, P. Pugach, B.F. Haynes, R.T. Wyatt, R.W. Sanders, J.M. Binley, A.B. Ward,
J.R. Mascola, P.D. Kwong, M. Connors, Broad and potent HIV-1 neutralization by a human antibody that binds
the gp41-gp120 interface, Nature, 515 (2014) 138-142.
[4] L. Xu, Y. Liu, Z. Chen, W. Li, Y. Liu, L. Wang, Y. Liu, X. Wu, Y. Ji, Y. Zhao, L. Ma, Y. Shao, C. Chen,
Surface-Engineered Gold Nanorods: Promising DNA Vaccine Adjuvant for HIV-1 Treatment, Nano Letters, 12

T
(2012) 2003-2012.
[5] Y. Tian, H. Wang, Y. Liu, L. Mao, W. Chen, Z. Zhu, W. Liu, W. Zheng, Y. Zhao, D. Kong, Z. Yang, W.

IP
Zhang, Y. Shao, X. Jiang, A Peptide-Based Nanofibrous Hydrogel as a Promising DNA Nanovector for
Optimizing the Efficacy of HIV Vaccine, Nano Letters, 14 (2014) 1439-1445.
[6] S. Kreiter, M. Vormehr, N. van de Roemer, M. Diken, M. Lower, J. Diekmann, S. Boegel, B. Schrors, F.

R
Vascotto, J.C. Castle, A.D. Tadmor, S.P. Schoenberger, C. Huber, O. Tureci, U. Sahin, Mutant MHC class II
epitopes drive therapeutic immune responses to cancer, Nature, 520 (2015) 692-696.

SC
[7] S. Mizutani, R.J. Colonno, In vitro synthesis of an infectious RNA from cDNA clones of human rhinovirus
type 14, Journal of virology, 56 (1985) 628-632.
[8] M. Mockey, C. Gonçalves, F.P. Dupuy, F.M. Lemoine, C. Pichon, P. Midoux, mRNA transfection of
dendritic cells: synergistic effect of ARCA mRNA capping with Poly (A) chains in cis and in trans for a high

NU
protein expression level, Biochemical and biophysical research communications, 340 (2006) 1062-1068.
[9] A. Yamamoto, M. Kormann, J. Rosenecker, C. Rudolph, Current prospects for mRNA gene delivery,
European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur
Pharmazeutische Verfahrenstechnik e.V, 71 (2009) 484-489.
MA
[10] Q. Hu, M. Wu, C. Fang, C. Cheng, M. Zhao, W. Fang, P.K. Chu, Y. Ping, G. Tang, Engineering
Nanoparticle-Coated Bacteria as Oral DNA Vaccines for Cancer Immunotherapy, Nano Letters, 15 (2015)
2732-2739.
[11] S. Pascolo, Messenger RNA-based vaccines, Expert opinion on biological therapy, 4 (2004) 1285-1294.
[12] O. Andries, M. De Filette, J. Rejman, S.C. De Smedt, J. Demeester, M. Van Poucke, L. Peelman, C.
D

Peleman, T. Lahoutte, N.N. Sanders, Comparison of the gene transfer efficiency of mRNA/GL67 and
pDNA/GL67 complexes in respiratory cells, Mol Pharm, 9 (2012) 2136-2145.
TE

[13] G. Tavernier, O. Andries, J. Demeester, N.N. Sanders, S.C. De Smedt, J. Rejman, mRNA as gene
therapeutic: how to control protein expression, Journal of controlled release : official journal of the Controlled
Release Society, 150 (2011) 238-247.
[14] B. Petsch, M. Schnee, A.B. Vogel, E. Lange, B. Hoffmann, D. Voss, T. Schlake, A. Thess, K.J. Kallen, L.
P

Stitz, T. Kramps, Protective efficacy of in vitro synthesized, specific mRNA vaccines against influenza A virus
infection, Nature biotechnology, 30 (2012) 1210-1216.
CE

[15] K. Deng, M. Pertea, A. Rongvaux, L. Wang, C.M. Durand, G. Ghiaur, J. Lai, H.L. McHugh, H. Hao, H.
Zhang, J.B. Margolick, C. Gurer, A.J. Murphy, D.M. Valenzuela, G.D. Yancopoulos, S.G. Deeks, T. Strowig, P.
Kumar, J.D. Siliciano, S.L. Salzberg, R.A. Flavell, L. Shan, R.F. Siliciano, Broad CTL response is required to
AC

clear latent HIV-1 due to dominance of escape mutations, Nature, 517 (2015) 381-385.
[16] M. Mockey, E. Bourseau, V. Chandrashekhar, A. Chaudhuri, S. Lafosse, E. Le Cam, V.F. Quesniaux, B.
Ryffel, C. Pichon, P. Midoux, mRNA-based cancer vaccine: prevention of B16 melanoma progression and
metastasis by systemic injection of MART1 mRNA histidylated lipopolyplexes, Cancer gene therapy, 14 (2007)
802-814.
[17] C. Pollard, J. Rejman, W. De Haes, B. Verrier, E. Van Gulck, T. Naessens, S. De Smedt, P. Bogaert, J.
Grooten, G. Vanham, S. De Koker, Type I IFN counteracts the induction of antigen-specific immune responses
by lipid-based delivery of mRNA vaccines, Molecular therapy : the journal of the American Society of Gene
Therapy, 21 (2013) 251-259.
[18] H. Kubler, B. Scheel, U. Gnad-Vogt, K. Miller, W. Schultze-Seemann, F. Vom Dorp, G. Parmiani, C.
Hampel, S. Wedel, L. Trojan, D. Jocham, T. Maurer, G. Rippin, M. Fotin-Mleczek, F. von der Mulbe, J. Probst,
I. Hoerr, K.J. Kallen, T. Lander, A. Stenzl, Self-adjuvanted mRNA vaccination in advanced prostate cancer
patients: a first-in-man phase I/IIa study, Journal for immunotherapy of cancer, 3 (2015) 26.
[19] N.J. Carter, M.P. Curran, Live Attenuated Influenza Vaccine (FluMist®; Fluenz™), Drugs, 71 (2011)
1591-1622.
[20] J. Bienenstock, M.R. McDermott, Bronchus‐and nasal‐associated lymphoid tissues, Immunological
reviews, 206 (2005) 22-31.
[21] J. Holmgren, C. Czerkinsky, Mucosal immunity and vaccines, Nature medicine, 11 (2005) S45-53.
[22] P. Brandtzaeg, Function of mucosa-associated lymphoid tissue in antibody formation, Immunological
investigations, 39 (2010) 303-355.
[23] A. Gebril, M. Alsaadi, R. Acevedo, A.B. Mullen, V.A. Ferro, Optimizing efficacy of mucosal vaccines,
(2012).

31
ACCEPTED MANUSCRIPT

[24] J. Brooking, S.S. Davis, L. Illum, Transport of Nanoparticles Across the Rat Nasal Mucosa, Journal of drug
targeting, 9 (2001) 267-279.
[25] Q. Liu, X. Chen, J. Jia, W. Zhang, T. Yang, L. Wang, G. Ma, pH-Responsive Poly(d,l-lactic-co-glycolic
acid) Nanoparticles with Rapid Antigen Release Behavior Promote Immune Response, ACS Nano, 9 (2015)
4925-4938.
[26] M.L. Read, S. Singh, Z. Ahmed, M. Stevenson, S.S. Briggs, D. Oupicky, L.B. Barrett, R. Spice, M. Kendall,
M. Berry, A versatile reducible polycation-based system for efficient delivery of a broad range of nucleic acids,

T
Nucleic acids research, 33 (2005) e86-e86.
[27] X. Su, J. Fricke, D.G. Kavanagh, D.J. Irvine, In vitro and in vivo mRNA delivery using lipid-enveloped

IP
pH-responsive polymer nanoparticles, Mol Pharm, 8 (2011) 774-787.
[28] F.T. Zohra, E.H. Chowdhury, T. Akaike, High performance mRNA transfection through carbonate
apatite–cationic liposome conjugates, Biomaterials, 30 (2009) 4006-4013.

R
[29] W. De Haes, G. Van Mol, C.l. Merlin, S.C. De Smedt, G. Vanham, J. Rejman, Internalization of mRNA
lipoplexes by dendritic cells, Molecular Pharmaceutics, 9 (2012) 2942-2949.

SC
[30] P. Midoux, C. Pichon, Lipid-based mRNA vaccine delivery systems, Expert Review of Vaccines, 14 (2015)
221-234.
[31] K.K. Phua, H.F. Staats, K.W. Leong, S.K. Nair, Intranasal mRNA nanoparticle vaccination induces
prophylactic and therapeutic anti-tumor immunity, Scientific reports, 4 (2014) 5128.

NU
[32] G. Ranaldi, I. Marigliano, I. Vespignani, G. Perozzi, Y. Sambuy, The effect of chitosan and other
polycations on tight junction permeability in the human intestinal Caco-2 cell line1, The Journal of Nutritional
Biochemistry, 13 (2002) 157-167.
[33] H. Debus, P. Baumhof, J. Probst, T. Kissel, Delivery of messenger RNA using poly(ethylene
MA
imine)–poly(ethylene glycol)-copolymer blends for polyplex formation: Biophysical characterization and in
vitro transfection properties, Journal of Controlled Release, 148 (2010) 334-343.
[34] F. Merkus, J. Verhoef, E. Marttin, S. Romeijn, P. Van der Kuy, W. Hermens, N. Schipper, Cyclodextrins in
nasal drug delivery, Advanced drug delivery reviews, 36 (1999) 41-57.
[35] R.U. Agu, M. Jorissen, T. Willems, G. Van den Mooter, R. Kinget, N. Verbeke, P. Augustijns, Safety
D

assessment of selected cyclodextrins—effect on ciliary activity using a human cell suspension culture model
exhibiting in vitro ciliogenesis, International journal of pharmaceutics, 193 (2000) 219-226.
TE

[36] S.H. Pun, N.C. Bellocq, A. Liu, G. Jensen, T. Machemer, E. Quijano, T. Schluep, S. Wen, H. Engler, J.
Heidel, Cyclodextrin-modified polyethylenimine polymers for gene delivery, Bioconjugate chemistry, 15 (2004)
831-840.
[37] G. Tang, H. Guo, F. Alexis, X. Wang, S. Zeng, T. Lim, J. Ding, Y. Yang, S. Wang, Low molecular weight
P

polyethylenimines linked by β‐cyclodextrin for gene transfer into the nervous system, The journal of gene
CE

medicine, 8 (2006) 736-744.


[38] H. Asanuma, A.H. Thompson, T. Iwasaki, Y. Sato, Y. Inaba, C. Aizawa, T. Kurata, S.-i. Tamura, Isolation
and characterization of mouse nasal-associated lymphoid tissue, Journal of immunological methods, 202 (1997)
123-131.
AC

[39] K. Sonaje, K.J. Lin, M.T. Tseng, S.P. Wey, F.Y. Su, E.Y. Chuang, C.W. Hsu, C.T. Chen, H.W. Sung,
Effects of chitosan-nanoparticle-mediated tight junction opening on the oral absorption of endotoxins,
Biomaterials, 32 (2011) 8712-8721.
[40] Y. Jiang, M. Li, Z. Zhang, T. Gong, X. Sun, Enhancement of nasal HIV vaccination with adenoviral
vector-based nanocomplexes using mucoadhesive and DC-targeting adjuvants, Pharm Res, 31 (2014)
2748-2761.
[41] C. Witschi, R.J. Mrsny, In vitro evaluation of microparticles and polymer gels for use as nasal platforms for
protein delivery, Pharmaceutical research, 16 (1999) 382-390.
[42] K.A. Foster, M.L. Avery, M. Yazdanian, K.L. Audus, Characterization of the Calu-3 cell line as a tool to
screen pulmonary drug delivery, International journal of pharmaceutics, 208 (2000) 1-11.
[43] I. Richard, M. Thibault, G. De Crescenzo, M.D. Buschmann, M. Lavertu, Ionization behavior of chitosan
and chitosan–DNA polyplexes indicate that chitosan has a similar capability to induce a proton-sponge effect as
PEI, Biomacromolecules, 14 (2013) 1732-1740.
[44] K. Ohtake, T. Maeno, H. Ueda, M. Ogihara, H. Natsume, Y. Morimoto, Poly-l-Arginine Enhances
Paracellular Permeability via Serine/Threonine Phosphorylation of ZO-1 and Tyrosine Dephosphorylation of
Occludin in Rabbit Nasal Epithelium, Pharmaceutical research, 20 (2003) 1838-1845.
[45] K. Karikó, H. Ni, J. Capodici, M. Lamphier, D. Weissman, mRNA is an endogenous ligand for Toll-like
receptor 3, Journal of Biological Chemistry, 279 (2004) 12542-12550.
[46] M. Fotin-Mleczek, K.M. Duchardt, C. Lorenz, R. Pfeiffer, S. Ojkic-Zrna, J. Probst, K.-J. Kallen, Messenger
RNA-based vaccines with dual activity induce balanced TLR-7 dependent adaptive immune responses and
provide antitumor activity, Journal of Immunotherapy, 34 (2011) 1-15.

32
ACCEPTED MANUSCRIPT

[47] O. Andries, M. De Filette, S.C. De Smedt, J. Demeester, M. Van Poucke, L. Peelman, N.N. Sanders, Innate
immune response and programmed cell death following carrier-mediated delivery of unmodified mRNA to
respiratory cells, Journal of controlled release : official journal of the Controlled Release Society, 167 (2013)
157-166.
[48] M.I. Ugwoke, R.U. Agu, N. Verbeke, R. Kinget, Nasal mucoadhesive drug delivery: background,
applications, trends and future perspectives, Advanced drug delivery reviews, 57 (2005) 1640-1665.
[49] H. Bshara, R. Osman, S. Mansour, A.E.-H.A. El-Shamy, Chitosan and cyclodextrin in intranasal

T
microemulsion for improved brain buspirone hydrochloride pharmacokinetics in rats, Carbohydrate polymers,
99 (2014) 297-305.

IP
[50] F. Wegmann, K.H. Gartlan, A.M. Harandi, S.A. Brinckmann, M. Coccia, W.R. Hillson, W.L. Kok, S. Cole,
L.P. Ho, T. Lambe, M. Puthia, C. Svanborg, E.M. Scherer, G. Krashias, A. Williams, J.N. Blattman, P.D.
Greenberg, R.A. Flavell, A.E. Moghaddam, N.C. Sheppard, Q.J. Sattentau, Polyethyleneimine is a potent

R
mucosal adjuvant for viral glycoprotein antigens, Nature biotechnology, 30 (2012) 883-888.
[51] Y. Wu, W. Wei, M. Zhou, Y. Wang, J. Wu, G. Ma, Z. Su, Thermal-sensitive hydrogel as adjuvant-free

SC
vaccine delivery system for H5N1 intranasal immunization, Biomaterials, 33 (2012) 2351-2360.

NU
MA
D
P TE
CE
AC

33

You might also like