IAI Accepts, published online ahead of print on 27 May 2014 Infect. Immun. doi:10.1128/IAI.01847-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Burkholderia pseudomallei capsular polysaccharide conjugates provide protection

2

against acute melioidosis

3 4

Andrew E. Scott1#, Mary N. Burtnick2, Margaret G. M. Stokes1, Adam O. Whelan1, E. Diane

5

Williamson1, Timothy P. Atkins1, Joann L. Prior1, Paul J. Brett2

6 7

1 Defence Science and Technology Laboratory, Porton Down, Salisbury, UK.

8

2 Department of Microbiology and Immunology, University of South Alabama, Mobile,

9

Alabama, USA

10 11

Correspondence

12

Fax: +44 01980 614307. Tel: +44 01980 613800. E-mail: [email protected]

13 14

Running title

15

Immunization against melioidosis

16 17 18 19 20 21 22 23 24 25

26

Abstract

27

Burkholderia pseudomallei, the etiologic agent of melioidosis, is a CDC Tier 1 Select Agent

28

that causes severe disease in both humans and animals. Diagnosis and treatment of

29

melioidosis can be challenging, and in the absence of optimal chemotherapeutic intervention

30

acute disease is frequently fatal. Melioidosis is an emerging infectious disease for which no

31

licensed vaccines currently exist. Due to the potential malicious use of B. pseudomallei, as

32

well as its impact on public health in endemic regions, there is significant interest in

33

developing vaccines for immunization against this disease. In the present study, type-A O-

34

polysaccharide (OPS) and manno-heptose capsular polysaccharide (CPS) antigens were

35

isolated from non-pathogenic, Select Agent-excluded strains of B. pseudomallei and

36

covalently linked to carrier proteins. Using these conjugates (OPS2B1 and CPS2B1) it was

37

demonstrated that, although high titer IgG responses against the OPS or CPS components of

38

the glycoconjugates could be raised in BALB/c mice, only those animals immunized with

39

CPS2B1 were protected against an intraperitoneal challenge of B. pseudomallei. Extending

40

upon these studies, it was also demonstrated that when the mice were immunized with a

41

combination of CPS2B1 and recombinant B. pseudomallei LolC, rather than with CPS2B1 or

42

LolC individually, they exhibited higher survival rates when challenged with a lethal dose of

43

B. pseudomallei. Collectively, these results suggest that CPS-based glycoconjugates are

44

promising candidates for the development of subunit vaccines for immunization against

45

melioidosis.

46 47

© Crown Copyright 2014. Published with the permission of the Defence Science and

48

Technology Laboratory on behalf of the controller of HMSO.

49 50

51

Introduction

52

Burkholderia pseudomallei is a Gram-negative, facultative intracellular pathogen that causes

53

melioidosis, a serious and often fatal disease in humans and animals (1). While the incidence

54

of melioidosis is particularly high in Southeast Asia and northern Australia, recent reports

55

have expanded the endemic region to include India and Brazil. Sporadic cases of melioidosis

56

have also been reported in various parts of the Caribbean, Central and South America, the

57

Middle East and Africa (2, 3). B. pseudomallei can be isolated from environmental niches

58

such as rice paddies, still or stagnant waters and moist soils which predominate in the tropics

59

(4), and it is believed that these habitats are the primary reservoirs from which susceptible

60

hosts acquire infections. Routes of infection include inhalation, ingestion and percutaneous

61

inoculation (5).

62

Melioidosis has a broad clinical spectrum and may manifest as acute localized

63

infections, acute pulmonary infections and fulminating septicemias or as chronic disease (6,

64

7). Remarkably, 20% of all community-acquired septicemias in northeast Thailand and 32%

65

of community-acquired bacteremic pneumonias in northern Australia are due to B.

66

pseudomallei infections (8, 9). Because of the non-specific presentation, the lack of rapid

67

diagnostic tests and the intrinsic resistance of B. pseudomallei to commonly used antibiotics,

68

diagnosis and treatment of melioidosis can be challenging. In the absence of optimal

69

chemotherapeutic intervention, mortality rates associated with acute human melioidosis

70

remain unacceptably high (40 - 50% in northeast Thailand and 19% in Australia (9, 10)). At

71

present, there are no human vaccines available for immunization against this emerging

72

infectious disease (for recent reviews see (11-13)). Due to the high risk of aerosol infection,

73

the severe course of disease and the potential for malicious use, B. pseudomallei is currently

74

classified as a CDC Tier 1 Select Agent.

75

Several studies have demonstrated that B. pseudomallei expresses a number of factors

76

that are required for virulence in animal models of infection. Included amongst these are the

77

Bsa type III secretion system, the VirAG two-component regulatory system and the cluster 1

78

type VI secretion system (for recent reviews see (5, 14)). Additionally, studies in our

79

laboratories and others have shown that the O-polysaccharide (OPS) component of B.

80

pseudomallei lipopolysaccharides (LPS) and the manno-heptose capsular polysaccharide

81

(CPS) are both virulence determinants (15-17) and protective antigens (18). Consequently,

82

these carbohydrate moieties have become important components of the various subunit

83

vaccines that are currently being developed for immunization against melioidosis (19, 20).

84

Unlike other Gram-negative pathogens, B. pseudomallei isolates appear to express

85

only a limited repertoire of OPS (21) and CPS antigens (16, 22). At present, the significance

86

of these observations with regards to virulence and evasion of host immune responses

87

remains to be fully determined. Nevertheless, these attributes bode well from a vaccine

88

development standpoint. Relevant to the current study, a number of studies have shown that

89

monoclonal and polyclonal antibodies specific for OPS and CPS can be used to passively

90

immunize mice against lethal challenges of B. pseudomallei (18, 20, 23-26). Such findings

91

confirm the protective capacity of these surface exposed antigens and support the rationale

92

for exploring the use of OPS and CPS antigens to actively immunize against melioidosis.

93

Immunologically, antigens can be classified as either T cell-dependent (TD) or T cell-

94

independent Type 1 or Type 2 (TI-1 or TI-2). Carbohydrates such as capsular antigens and O-

95

polysaccharides are generally considered to be TI-2 antigens (27). Typically, high molecular

96

weight TI-2 antigens such as capsular polysaccharides are immunogenic due to their ability to

97

crosslink multiple surface immunoglobulin molecules present on antigen-specific B cells

98

(28), but without the involvement of T helper (Th) cells, TI-2 antigens induce poor

99

immunological memory, and only limited affinity maturation and isotype switching (29).

100

Additionally, without dosing at frequent intervals antibody levels often decline. Efforts to

101

overcome the poor immunogenicity of many clinically relevant polysaccharides have led to

102

the development of glycoconjugate vaccines (30, 31), a number of which are currently

103

licensed for human use (32). Covalent linkage of polysaccharides to carrier proteins promotes

104

Th cell involvement which improves immunological memory (33) and increases isotype

105

switching. The affinities of the antibodies elicited by glycoconjugates may also be higher

106

than those produced by polysaccharides alone (29).

107

In the present study, B. pseudomallei OPS- and CPS-based glycoconjugates were

108

constructed and evaluated their immunogenic potential and protective capacities using a

109

murine model of melioidosis. Collectively, the results suggest that CPS-based

110

glycoconjugates are promising candidates for the development of subunit vaccines for

111

immunization against melioidosis.

112 113

Materials and methods

114

Strain and growth conditions

115

The bacteria used in this study were B. pseudomallei strains K96243 (34), RR2808 (ΔpurM,

116

ΔwcbB) (35) and RR2683 (ΔpurM, ΔrmlD) (19) and E. coli BL21 Star(DE3)(pLysS)

117

(pCRT7/NT-TOPO-LolC) (36). B. pseudomallei strains RR2808 and RR2683 are derivatives

118

of the adenine auxotroph Bp82 (37), a Select Agent excluded ΔpurM derivative of strain

119

1026b. Strain RR2808 (CPS mutant; source of OPS) and RR2683 (OPS mutant; source of

120

CPS) enable the production of highly purified polysaccharides without the requirement for

121

BSL3 facilities. B. pseudomallei K96243 and E. coli BL21 were cultured in L-broth and on

122

L-agar at 37°C. B. pseudomallei RR2808 and RR2683 were cultured in L-broth and on L-

123

agar supplemented with thiamine (5μg/mL) and adenine (100μg/mL) at 37°C. Bacterial

124

stocks were maintained at –80°C as 20% glycerol suspensions. Wild type B. pseudomallei

125

K96243 was handled at Advisory Committee for Dangerous Pathogens (ACDP) containment

126

level 3.

127 128

For animal challenges, B. pseudomallei K96243 was inoculated from a glycerol stock into

129

100ml L-broth and incubated for 24 hours at 37°C with shaking (180 rpm). The OD590 was

130

adjusted to 0.4 (corresponding to approximately 4 x 108 CFU / ml) and diluted in L-broth to

131

the correct concentration for challenge.

132 133

OPS and CPS purification

134

Broth in 2 L baffled Erlenmeyer flasks was inoculated with B. pseudomallei RR2808 or

135

RR2683 and incubated overnight at 37°C with vigorous shaking. Cell pellets were obtained

136

by centrifugation and extracted using a modified hot aqueous-phenol procedure (38). Purified

137

OPS and CPS antigens were then obtained essentially as previously described (19, 39).

138 139

Glycoconjugate synthesis

140

The OPS2B1 and CPS2B1 glycoconjugates used in this study were synthesized using

141

reductive amination chemistry as previously described (19, 39). Briefly, purified OPS or CPS

142

samples were solubilized at 5 mg/ml in PBS and added to a small amber vials. To each ml of

143

the solutions was added 6 mg (~30 mM) of sodium meta-periodate (NaIO4; Pierce). Once the

144

crystals had dissolved by gentle agitation, the reaction mixtures were gently stirred at room

145

temperature for 40 minutes. To remove any excess oxidizing agent, the reaction mixtures

146

were applied to a Zeba Desalt Spin Columns (Pierce) equilibrated with PBS and the eluates

147

collected. To facilitate conjugation of the OPS or CPS antigens to cationized bovine serum

148

albumin (cBSA; Pierce), the activated polysaccharides were added to small amber vials. To

149

each ml of the OPS or CPS solutions was added 0.5 ml of the carrier proteins (5 mg/ml in

150

PBS). After mixing by gentle agitation, 10μl aliquots of a sodium cyanoborohydride stock (1

151

M NaBH3CN in 10 mM NaOH) were added to each ml of the conjugation mixtures and the

152

reactions were gently stirred at room temperature for 4 days. Following this, 10μl aliquots of

153

a sodium borohydride stock (1M NaBH4 in 10mM NaOH) were added to each ml of the

154

conjugation mixtures and the reactions were stirred for 40 minutes. The conjugate reactions

155

were then brought to 5ml with ultrapure water, dialyzed against distilled water and then

156

lyophilized. The resulting preparations were re-suspended in ultrapure water as 1mg/ml

157

stocks and stored at -20°C until required for use. BCA assays were used to quantitate the

158

protein concentrations of the glycoconjugate stocks (the remaining masses of which were

159

assumed to be polysaccharide).

160 161

LolC protein preparation

162

LolC protein (encoded by BPSL2277 in B. pseudomallei K96243) was purified from E. coli

163

BL21 Star(DE3)(pLysS) (pCRT7/NT-TOPO-LolC) as previously described (36). Briefly,

164

cells were grown to mid-log phase at 37°C, induced by the addition of 1mM IPTG and cooled

165

to 20°C for overnight incubation. Following cell disruption by sonication, LolC was loaded

166

onto HisTrap FF columns (GE Healthcare) and eluted in buffer (40mM Tris-Cl, 750 mM

167

NaCl, 1% glucose, 5% glycerol, pH 7.5) using imidazole in steps to 500mM. The recovered

168

material was dialysed against PBS and protein purity assessed using SDS-PAGE and

169

concentration determined using a BCA assay.

170 171

SDS-PAGE and Western immunoblotting

172

Glycoconjugate samples were analysed using SDS-PAGE and Western immunoblotting as

173

previously described (19, 39). Briefly, samples solubilized in 1X SDS-PAGE sample buffer

174

and heated to 100°C for 5 minutes prior to electrophoresis on 12% Precise gels (Pierce).

175

Proteins were visualized via staining with Coomassie Blue R-250. For Western immunoblot

176

analyses, the glycoconjugate samples and controls were separated on the same 12% gels and

177

electrophoretically transferred to nitrocellulose membranes, before hybridization to either B.

178

pseudomallei OPS-specific mAb (Pp-PS-W) or a CPS-specific mAb (3C5).

179 180

Animal studies

181

All investigations involving animals were carried out according to the requirements of the

182

UK Animal (Scientific Procedures) Act 1986. Studies were performed using 6 – 8 week old

183

female BALB/c mice (Charles River) randomly allocated into cages of five upon arrival.

184

Mice were held under a 12 hour light/dark cycle with free access to food and water and were

185

implanted with a microchip to allow tracking of individual mice. After challenge with B.

186

pseudomallei, animals were handled under ACDP containment level 3 conditions within a

187

rigid-wall half-suit isolator. Mice were checked at least twice daily following challenge and

188

clinical signs for each mouse recorded. Upon reaching pre-determined humane end-points,

189

mice were culled via cervical dislocation.

190

Mice were immunized subcutaneously (s.c.) on days 0, 21 and 35 with the various

191

immunogens. All immunogens were formulated in Alhydrogel 2% (500 μg/mouse; Brenntag)

192

plus ODN 2006 (CpG; 20 μg/mouse; InvivoGen). The mice received 5µg per dose of OPS or

193

CPS as a conjugate, 5µg per dose of unconjugated OPS or CPS and 10µg per dose of cBSA

194

or LolC. Challenges with B. pseudomallei K96243 were delivered at day 70 via the intra-

195

peritoneal route (i.p.; the MLD at day 35 for B. pseudomallei K96243 via this route was

196

calculated to be 744 CFU (unpublished)). For organ bacterial enumeration, animals were

197

culled and organs removed. These were then mashed through 40μm sieves into PBS, serially

198

diluted and plated onto L-agar.

199

200

Analysis of antibody responses

201

Approximately 0.1ml of blood was collected from the tail veins of mice two weeks after the

202

final boost was administered. After clotting at 4°C, the serum was removed and stored at -

203

20°C until required for use. Responses directed against the OPS, CPS or LolC antigens were

204

assessed by ELISA essentially as previously described (40). A reading of twice background

205

or above was considered positive and the titer was determined to be the reciprocal of the final

206

positive dilution.

207 208

Statistical analysis

209

All graphs were produced using the program GraphPad PRISM V5.0. Survival data was

210

analysed using a log rank (Mantel-Cox) test. Bacterial burden data was transformed to the

211

logarithm of 10 and compared using a Mann-Whitney U-test. Antibody data was transformed

212

to the logarithm of 10 and compared using a Mann-Whitney U-test.

213 214

Results

215

Glycoconjugate synthesis

216

Polysaccharide-based glycoconjugates represent a rational approach for immunizing against

217

melioidosis. However, until recently isolating OPS and CPS antigens to develop these

218

vaccine candidates has been hampered by the fact that B. pseudomallei is a Select Agent

219

requiring specialized handling and containment practices. To address this issue, B.

220

pseudomallei RR2808 and RR2683 (derivatives of the Select Agent-excluded strain Bp82, a

221

ΔpurM derivative of 1026b) were created to enable us to produce these polysaccharides in a

222

safer and more cost-effective manner without the requirement for BSL-3 containment (19,

223

35).

224

To facilitate the construction of the glycoconjugates described in this study, the OPS

225

and CPS antigens purified from RR2808 and RR2683 were chemically activated with sodium

226

periodate and covalently linked to cBSA via reductive amination to produce OPS2B1 and

227

CPS2B1 (Figure 1A). Following conjugation, the samples were examined by SDS-PAGE.

228

Results of these analyses demonstrated that in both instances the polysaccharides had

229

covalently linked to the protein carriers as indicated by the shifts in molecular weights of the

230

glycoconjugates relative to the unconjugated cBSA controls (Figure 1B). Additionally,

231

Western immunoblotting confirmed that the structural integrity/antigenicity of the OPS and

232

CPS moieties remained intact following chemical activation and linkage to the protein

233

carriers based upon their reactivity with the Pp-PS-W or 3C5 mAbs (data not shown). Further

234

analysis of the constructs revealed that OPS2B1 and CPS2B1 contained 60% OPS or 53%

235

CPS (w/w) respectively.

236 237

Immunogenicity of the glycoconjugates

238

To compare the immunogenicity of the glycoconjugates, groups of BALB/c mice were

239

immunized with OPS2B1 or CPS2B1. For control purposes, groups of mice were also

240

immunized with a matching amount of antigen composed of mixed but unconjugated

241

polysaccharide and cBSA. Two weeks after the final boost, serum was obtained from the

242

mice and the LPS and CPS specific IgG titers were determined by ELISA (Figure 2). In both

243

cases, the glycoconjugates induced significantly higher antigen-specific IgG titers than the

244

unconjugated controls (p

Burkholderia pseudomallei capsular polysaccharide conjugates provide protection against acute melioidosis.

Burkholderia pseudomallei, the etiologic agent of melioidosis, is a CDC tier 1 select agent that causes severe disease in both humans and animals. Dia...
668KB Sizes 0 Downloads 0 Views