CVI Accepted Manuscript Posted Online 21 January 2015 Clin. Vaccine Immunol. doi:10.1128/CVI.00714-14 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

1

Vaccine Self-assembling Immune Matrix is a new delivery platform that enhances immune

2

responses to rHBsAg in mice

3 4

Rafaella F. Q. Grenfell*a,b, Lisa M. Shollenberger*a, E. Farah Samlia, Donald A. Harna#

5 6

Department of Infectious Diseases, College of Veterinary Medicine and The Center for Tropical

7

and Emerging Global Diseases, University of Georgia, Athens, GA, USAa. Schistosomiasis

8

Laboratory, Rene Rachou Research Center, Oswaldo Cruz Foundation, Belo Horizonte, Minas

9

Gerais, Brazilb.

10 11

Running Title: Adjuvanticity of Vaccine Self-assembling Immune Matrix

12 13

#Address correspondence to Donald A. Harn, [email protected].

14

*R.F.Q.G. and L.M.S. contributed equally to this work.

1 of 23

15

ABSTRACT

16 17

Vaccination remains the most effective public health tool to prevent infectious diseases. Many

18

vaccines are marginally effective and need enhancement for immune compromised, elderly and

19

very young populations. To enhance immunogenicity, we exploited the biphasic property of the

20

(RADA)4 synthetic oligopeptide to create VacSIM™ (Vaccine Self-assembling Immune

21

Matrix), a new delivery method. VacSIM™ solution can easily be mixed with antigens,

22

organisms and adjuvants for injection. Post-injection, the peptides self-assemble into hydrated

23

nanofiber gel-matrices, forming a depot with antigens and adjuvants in the aqueous phase. We

24

believe the depot provides slow release of immunogens, leading to increased activation of

25

antigen presenting cells that then drive enhanced immunogenicity. Using recombinant hepatitis B

26

surface antigen (rHBsAg) as a model immunogen, we compared VacSIM™ delivery to delivery

27

in alum or complete Freund’s adjuvant (CFA). Delivery of the rHBsAg antigen to mice via

28

VacSIM™ without adjuvant elicited higher specific IgG responses than when rHBsAg was

29

delivered in alum or CFA. Evaluating IgG subtypes shows a mixed Th1/Th2 type response

30

following immunization with VacSIM™, which was driven further toward Th1 with addition of

31

CpG as adjuvant. Increased specific IgG endpoint titers were observed in both C57BL/6 and

32

BALB/c mice, representative of Th1 and Th2 environments, respectively. Restimulation of

33

splenocytes suggests VacSIM™ does not cause an immediate pro-inflammatory response in the 2 of 23

34

host. Overall, these results suggest that VacSIM™, as a new delivery method, has potential to

35

enhance immunogenicity and efficacy of numerous vaccines.

36 37

INTRODUCTION

38 39

Vaccines remain the single greatest public health tool to combat infectious diseases. Vaccine

40

formulation and delivery are key to the ability of vaccines to induce the desired immune

41

responses. One goal of vaccine delivery is to present vaccine antigens in a manner that enhances

42

antigen presenting cell (APC) activation, leading to antigen/organism uptake and processing of

43

vaccine antigen(s). Delivery methods or adjuvants that safely enhance vaccine

44

immunogenicity/efficacy are desirable for vaccines that are marginally effective, or for vaccines

45

administered to low-responders, or immune compromised populations. Additional goals are to

46

reduce the number of doses required to induce effective, vaccine responses and/or to reduce the

47

amount of vaccine/dose, especially when a single dose of vaccine is administered, as with annual

48

influenza vaccine. Lastly, in pandemics, a vaccine that produces high titers after a single

49

administration would be beneficial. Recent advances in the understanding of how innate

50

mechanisms influence adaptive immunity have led to more rational design in the development of

51

new vaccine adjuvants and delivery systems.

52 3 of 23

53

Aluminum salts were the first adjuvants licensed for human vaccines in the 1920s. The licensure

54

of non-aluminum salt adjuvants took an additional 80 years (1). One reason for this long gap is

55

that the principles of adjuvant activity were largely unknown, thus the development of adjuvants

56

was empirical, Moreover, many adjuvants, including Freund’s adjuvant, were reactogenic and

57

not acceptable for licensure (2). Recent methods to improve vaccine delivery have taken several

58

approaches, including, virosomes (3-5), vector-based (6-8), liposome based (9-11) and more

59

traditional formulation with adjuvants (12-17). Each of these methods has some drawbacks,

60

either in terms of reactogenicity, regulatory issues, product stability or time required for

61

formulation, however each of these methods focuses on presenting the vaccine as a particulate.

62 63

To enhance vaccine immunogenicity over that seen when conventional alum-based delivery

64

methods are used, we focused on identifying ways to deliver vaccines such that vaccine antigens

65

are released over time. This led to our development of a new vaccine delivery method we call

66

VacSIM™ (Vaccine Self-Assembling Immune Matrix). VacSIM™ is based on the unique

67

properties of the (RADA)4 synthetic oligopeptide and other biopolymers (18-22). The (RADA)4

68

synthetic oligopeptide was created by Zhang (22), and commercialized by 3-D Matrix Inc. for

69

cell scaffolding, and is currently in clinical trials for wound healing (PuraStat®), tissue repair

70

(23), and dental implant scaffolding (PuraMatrix) (24), and as such, has already undergone third

71

party reactogenicity and toxicity testing (25). VacSIM™ is composed solely of the (RADA)4 4 of 23

72

synthetic oligopeptide. Thus, it is biocompatible and biodegradable. Ex vivo, a 1.0% VacSIM™

73

solution is in liquid phase, allowing the flexibility to mix virtually any antigen, organism and

74

adjuvant. Upon injection into tissue, VacSIM™ self-assembles into hydrated nanofibers (26),

75

forming a gel matrix depot, which entraps and concentrates vaccine components in the aqueous

76

phase at the injection site (27). We hypothesize that the vaccine depot allows slow egress of

77

antigen out of the gel pores, leading to persistent release of antigen, which is considered to be

78

important in the development of robust adaptive immunity and memory responses (28, 29). We

79

theorize that slow release of antigen and possible cellular infiltration of the VacSIM™ gel-depot,

80

increases activation and maturation of antigen presenting cells that then drive more robust

81

adaptive responses. Further, it is possible that the gel depot protects vaccine components from

82

degradation.

83 84

VacSIM™ is different from first and second generation hydrogels, such as alginates and

85

methacrylates, as well as microneedle and layer-by-layer technology, as all of these require

86

polymerization ex vivo (30-35). Further, it is different from other self-assembling peptide (SAP)

87

technologies, such as the Q11 peptide, where the antigenic motif must be directly conjugated to

88

the SAP (36-38). In contrast, VacSIM™ is prepared by simply mixing SAP and antigens prior to

89

administration.

90 5 of 23

91

The biphasic property of VacSIM™, coupled with the inert nature of the resultant vaccine gel

92

depot, provides novel technology that can be translated for use in a multitude of vaccines. In this

93

study, we tested the ability of VacSIM™ to enhance specific immune responses to the

94

recombinant hepatitis B surface antigen.

95 96

MATERIALS & METHODS

97 98

Immunizations. Five to seven week old female BALB/c or C57BL/6 mice were purchased from

99

Harlan Laboratories, housed in specific pathogen-free conditions and allowed to acclimate for

100

one week prior to manipulation. All animal work was performed in accordance with all

101

applicable policies and approved by the institutional animal care and use committee. Mice were

102

immunized subcutaneously with recombinant Hepatitis B surface antigen (rHBsAg) adw subtype

103

(5 g, Fitzgerald Industries, Inc. Massachusetts, USA) ± CpG (50 g ODN 1826, InvivoGen,

104

Inc. California, USA), via alhydrogel (250 g, Inject Alum, Thermo Fisher Scientific, Inc.,

105

Pittsburgh, PA, USA) ± CpG, by VacSIM™ ± CpG, in Freund's (13 l, Sigma-Aldrich Co.

106

Missouri, USA) or sham-immunized with VacSIM™ or phosphate buffered saline (PBS), at a

107

maximum volume of 200 l per injection site. Three or four weeks later, as indicated, mice were

108

administered a second, identical immunization.

109 6 of 23

110

Antibody Responses. For the kinetic experiments, blood samples were collected from all

111

immunized and control mice weekly, beginning week -1 prior to primary immunization.

112

rHBsAg-specific antibodies in sera were analyzed by ELISA. Briefly, plates were coated with 4

113

g/ml rHBsAg before singly diluted sera (1:800 for IgA, IgG2a and 1:1200 for IgM, IgG, IgG1)

114

was added. Antibodies specific for rHBsAg were detected by HRP-conjugated secondary

115

antibodies (1:2000 α-IgA, 1:2500 α-IgM, and 1:2500 α-IgG from Santa Cruz; 1:1000 α-IgG1 and

116

1:1000 IgG2a from Invitrogen). Plates were developed with SureBlue TMB 1-component

117

substrate (KPL) and stopped by addition of 2N sulfuric acid. To evaluate the IgG1/IgG2a ratio,

118

we multiplied the absorbance values by the dilution factors of the sera to normalize prior to

119

dividing. For non-kinetic studies, all methods remain the same except that mice were bled prior

120

to immunization, and post each subsequent immunization. Each sample of serum was diluted

121

serially for endpoint titer antibody analysis and samples with undetectable antibody were

122

assigned a value below the detection limit.

123 124

ELISpots. Splenocytes were obtained 3weeks post-prime or 2 weeks post-boost, then stimulated

125

for 20 hours to evaluate HBsAg-specific cell-mediated responses using IFN ELISpot, according

126

to the manufacturer’s instructions (BD Biosciences, San Francisco, CA, USA). Briefly, single

127

cell suspensions (3 and 1.5 × 105 cells per well) were cultured at 37°C with 5% CO2 for 20 hours

128

in complete medium [RPMI-1640 (Hyclone, Thermo Scientific, Utah, USA) supplemented with 7 of 23

129

10% FBS, 100 U/ml penicillin, 100 μg/ml streptomycin, antimycotic, non-essential amino acids

130

and β-mercaptoethanol (Sigma-Aldrich, St. Louis, MO, USA). Cells were stimulated with 1

131

μg/ml Concanavalin A (ConA), 20 µM HBs specific peptide [S28-39, IPQSLDSWWTSL

132

(synthesized at greater than 95% purity [Biosynthesis Inc., Lewisville, TX, USA and dissolved in

133

DMSO prior to dilution in culture media]) or left unstimulated. ELISpot plates were developed

134

with AEC substrate and spot forming units (SFU) were counted using an Immunospot Analyzer

135

(C.T.L.). The SFU value was expressed as mean of triplicate cultures per mouse.

136 137

Flow cytometry. Splenocytes collected at 3 weeks post prime and 2 weeks post boost were

138

stimulated for 3 days with 5 µM HBs S28-39 peptide and 40 U/ml rIL-2 (Peprotech) for

139

assessment of molecular specificity. Briefly, HBs peptide was bound with H2-Ld DimerX

140

reagent (BD) at 37oC overnight and incubated with secondary antibody (A85-1-PE, BD) and

141

isotype control (mIgG1λ, BD). Two million re-stimulated splenocytes were stained with

142

DimerX-HB reagent, α-CD8 Pacific Blue antibody (BD) and viability dye (Live/Dead Near

143

Infrared fixable dye, Invitrogen). Live cells were acquired using a BD LSRII flow cytometer and

144

analyzed with FACS Diva software (BD).

145 146

Cytokine responses. Splenocytes were isolated two weeks post boost and tested for rHBsAg-

147

specific cytokine production. Single cell suspensions (1.5 × 106/ml) were cultured at 37°C with 8 of 23

148

5% CO2 in complete medium and stimulated with 1 μg/ml ConA, 5 μg/ml rHBsAg or left

149

unstimulated. Levels of TNF-α and IFN were quantified after 24 and 48 hours culture, IL-5

150

after 48 hours, IL-4 at 48 and 72 hrs and IL-10 after 72 hours, each in triplicate, by ELISA,

151

according to manufacturer’s instructions (BD).

152 153

Statistical analyses. Except for the kinetic studies in Figure 1A, all other data are represented by

154

box and whisker plots, ranging from the minimum value to the maximum, with the mean

155

displayed as a plus (+) sign. Statistical analyses (one- or two-way ANOVAs with Bonferroni

156

post tests) were performed using Prism 5 (GraphPad, California, USA). Differences were

157

considered statistically significant when p values were ≤ 0.05 and are denoted by asterisks.

158 159

RESULTS

160 161

Using recombinant hepatitis B surface antigen (rHBsAg) as a model immunogen, we compared

162

the VacSIM™ delivery to rHBsAg in alum or Freund’s, +/- CPG as a model adjuvant for use

163

with VacSIM™ and alum.

164 165

VacSIM™ enhances and sustains specific humoral immunity

166 9 of 23

167

To assess humoral immunity, total rHBsAg-specific IgA, IgM and IgG titers were quantified at

168

various times post immunization and are presented in Figure 1A. Mice immunized with

169

VacSIM™-delivered rHBsAg had higher specific IgM levels than CFA-delivered antigen

170

(Figure 1B, upper panel, p

Vaccine self-assembling immune matrix is a new delivery platform that enhances immune responses to recombinant HBsAg in mice.

Vaccination remains the most effective public health tool to prevent infectious diseases. Many vaccines are marginally effective and need enhancement ...
1MB Sizes 0 Downloads 9 Views